1
|
Huang X, Tan J, Chen M, Zheng W, Zou S, Ye X, Li Y, Wu M. Prognostic, Immunological, and Mutational Analysis of MTA2 in Pan-Cancer and Drug Screening for Hepatocellular Carcinoma. Biomolecules 2023; 13:883. [PMID: 37371463 DOI: 10.3390/biom13060883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Metastasis-associated protein 2 (MTA2) is a member of the metastasis-associated transcriptional regulator family and is a core component of the nucleosome remodeling and histone deacetylation complex. Despite growing evidence that MTA2 plays a crucial role in the tumorigenesis of certain cancers, no systematic pan-cancer analysis of MTA2 is available to date. Therefore, the aim of our study is to explore the prognostic value of MTA2 in 33 cancer types and to investigate its potential immune function. METHODS by comprehensive use of databases from TCGA, GTEx, GEO, UCSC xena, cBioPortal, comPPI, GeneMANIA, TCIA, MSigDB, and PDB, we applied various bioinformatics approaches to investigate the potential role of MTA2, including analyzing the association of MTA2 with MSI, prognosis, gene mutation, and immune cell infiltration in different tumors. We constructed a nomogram in TCGA-LIHC, performed single-cell sequencing (scRNA-seq) analysis of MTA2 in hepatocellular carcinoma (HCC), and screened drugs for the treatment of HCC. Finally, immunohistochemical experiments were performed to verify the expression and prognostic value of MTA2 in HCC. In vitro experiments were employed to observe the growth inhibition effects of MK-886 on the HCC cell line HepG2. RESULTS The results suggested that MTA2 was highly expressed in most cancers, and MTA2 expression was associated with the prognosis of different cancers. In addition, MTA2 expression was associated with Tumor Mutation Burden (TMB) in 12 cancer types and MSI in 8 cancer types. Immunoassays indicated that MTA2 positively correlated with activated memory CD4 T cells and M0 macrophage infiltration levels in HCC. ScRNA-seq analysis based on the GEO dataset discovered that MTA2 was significantly expressed in T cells in HCC. Finally, the eXtreme Sum (Xsum) algorithm was used to screen the antitumor drug MK-886, and the molecular docking technique was utilized to reveal the binding capacity between MK-886 and the MTA2 protein. The results demonstrated excellent binding sites between them, which bind to each other through Π-alkyl and alkyl interaction forces. An immunohistochemistry experiment showed that MTA2 protein was highly expressed in HCC, and high MTA2 expression was associated with poor survival in HCC patients. MK-886 significantly inhibited the proliferation and induced cell death of HepG2 cells in a dose-dependent manner. CONCLUSIONS Our study demonstrated that MTA2 plays crucial roles in tumor progression and tumor immunity, and it could be used as a prognostic marker for various malignancies. MK-886 might be a powerful drug for HCC.
Collapse
Affiliation(s)
- Xueshan Huang
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang 524000, China
| | - Jingyi Tan
- School of Pharmacy, Guangdong Medical University, Zhanjiang 524000, China
- School of Basic Medicine, Guangdong Medical University, Zhanjiang 524000, China
| | - Mei Chen
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang 524000, China
| | - Weirang Zheng
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang 524000, China
| | - Shanyang Zou
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang 524000, China
| | - Xiaoxia Ye
- School of Basic Medicine, Guangdong Medical University, Zhanjiang 524000, China
| | - Yutong Li
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang 524000, China
| | - Minhua Wu
- School of Basic Medicine, Guangdong Medical University, Zhanjiang 524000, China
| |
Collapse
|
2
|
Bibliometric Analysis of Hotspots and Frontiers of Immunotherapy in Pancreatic Cancer. Healthcare (Basel) 2023; 11:healthcare11030304. [PMID: 36766879 PMCID: PMC9914338 DOI: 10.3390/healthcare11030304] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most common malignant neoplasms with an increasing incidence, low rate of early diagnosis, and high degree of malignancy. In recent years, immunotherapy has made remarkable achievements in various cancer types including pancreatic cancer, due to the long-lasting antitumor responses elicited in the human body. Immunotherapy mainly relies on mobilizing the host's natural defense mechanisms to regulate the body state and exert anti-tumor effects. However, no bibliometric research about pancreatic cancer immunotherapy has been reported to date. This study aimed to assess research trends and offer possible new research directions in pancreatic cancer immunotherapy. METHODS The articles and reviews related to pancreatic cancer immunotherapy were collected from the Web of Science Core Collection. CiteSpace, VOSviewer, and an online platform, and were used to analyze co-authorship, citation, co-citation, and co-occurrence of terms retrieved from the literature highlighting the scientific advances in pancreatic cancer immunotherapy. RESULTS We collected 2475 publications and the number of articles was growing year by year. The United States had a strong presence worldwide with the most articles. The most contributing institution was Johns Hopkins University (103 papers). EM Jaffee was the most productive researcher with 43 papers, and L Zheng and RH Vonderheide ranked second and third, with 34 and 29 papers, respectively. All the keywords were grouped into four clusters: "immunotherapy", "clinical treatment study", "tumor immune cell expression", "tumor microenvironment". In the light of promising hotspots, keywords with recent citation bursts can be summarized into four aspects: immune microenvironment, adaptive immunotherapy, immunotherapy combinations, and molecular and gene therapy. CONCLUSIONS In recent decades, immunotherapy showed great promise for many cancer types, so various immunotherapy approaches have been introduced to treat pancreatic cancer. Understanding the mechanisms of immunosuppressive microenvironment, eliminating immune suppression and blocking immune checkpoints, and combining traditional treatments will be hotspots for future research.
Collapse
|
3
|
Positron emission tomography molecular imaging to monitor anti-tumor systemic response for immune checkpoint inhibitor therapy. Eur J Nucl Med Mol Imaging 2023; 50:1671-1688. [PMID: 36622406 PMCID: PMC10119238 DOI: 10.1007/s00259-022-06084-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/08/2022] [Indexed: 01/10/2023]
Abstract
Immune checkpoint inhibitors (ICIs) achieve a milestone in cancer treatment. Despite the great success of ICI, ICI therapy still faces a big challenge due to heterogeneity of tumor, and therapeutic response is complicated by possible immune-related adverse events (irAEs). Therefore, it is critical to assess the systemic immune response elicited by ICI therapy to guide subsequent treatment regimens. Positron emission tomography (PET) molecular imaging is an optimal approach in cancer diagnosis, treatment effect evaluation, follow-up, and prognosis prediction. PET imaging can monitor metabolic changes of immunocytes and specifically identify immuno-biomarkers to reflect systemic immune responses. Here, we briefly review the application of PET molecular imaging to date of systemic immune responses following ICI therapy and the associated rationale.
Collapse
|
4
|
Songjang W, Nensat C, Pongcharoen S, Jiraviriyakul A. The role of immunogenic cell death in gastrointestinal cancer immunotherapy (Review). Biomed Rep 2021; 15:86. [PMID: 34512974 PMCID: PMC8411483 DOI: 10.3892/br.2021.1462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
Modern cancer immunotherapy techniques are aimed at enhancing the responses of the patients' immune systems to fight against the cancer. The main promising strategies include active vaccination of tumor antigens, passive vaccination with antibodies specific to cancer antigens, adoptive transfer of cancer-specific T cells and manipulation of the patient's immune response by inhibiting immune checkpoints. The application of immunogenic cell death (ICD) inducers has been proven to enhance the immunity of patients undergoing various types of immunotherapy. The dying, stressed or injured cells release or present molecules on the cell surface, which function as either adjuvants or danger signals for detection by the innate immune system. These molecules are now termed 'damage-associated molecular patterns'. The term 'ICD' indicates a type of cell death that triggers an immune response against dead-cell antigens, particularly those derived from cancer cells, and it was initially proposed with regards to the effects of anticancer chemotherapy with conventional cytotoxic drugs. The aim of the present study was to review and discuss the role and mechanisms of ICD as a promising combined immunotherapy for gastrointestinal tumors.
Collapse
Affiliation(s)
- Worawat Songjang
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Chatchai Nensat
- Biomedical Sciences, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Sutatip Pongcharoen
- Division of Immunology, Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand
| | - Arunya Jiraviriyakul
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
5
|
Dyhl-Polk A, Mikkelsen MK, Ladekarl M, Nielsen DL. Clinical Trials of Immune Checkpoint Inhibitors in Hepatocellular Carcinoma. J Clin Med 2021; 10:2662. [PMID: 34208788 PMCID: PMC8234948 DOI: 10.3390/jcm10122662] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Introduction: Several immune checkpoint inhibitors (CPIs) are under clinical development in hepatocellular carcinoma (HCC) and the field is advancing rapidly. In this comprehensive review, we discuss published results and report on ongoing clinical trials. Methods: A literature search was carried out using PubMed and EMBASE; data reported at international meetings and clinicaltrials.gov were included as well. The search was updated 5 March 2021. We evaluated studies with monotherapy CPI's, combinations of CPI's and combinations of CPI's with other treatment modalities separately. Only studies with at least 10 included patients were considered. Results: We identified 2649 records published in the English language literature. After review, 29 studies remained, including 12 studies with preliminary data only. The obtained overall response rate of PD-1/PDL-1 monotherapy in phase II studies in the second-line setting was 15-20% with disease control in approximately 60% of patients. The responses were of long duration in a subset of patients. Furthermore, the safety profiles were manageable. However, a phase III study comparing nivolumab with sorafenib in the first-line setting and a phase III study evaluating pembrolizumab versus best supportive care in the second-line setting did not meet their prespecified endpoints. More recently, a phase I/II study of nivolumab and ipilimumab has resulted in a response rate of approximately 30% with a median OS of 22 months in the second-line setting. Multiple trials have been initiated to evaluate CPIs in combination with molecularly targeted drugs, especially anti-angiogenic drugs or local therapy. A phase III study investigating atezolizumab plus bevacizumab versus sorafenib in the first-line setting showed significantly increased survival in the combination arm. Conclusions: The combination of atezolizumab and bevacizumab represents a new standard of care in the first-line setting for fit patients with preserved liver function. CPIs can produce durable tumor remission and induce long-standing anti-tumor immunity in a subgroup of patients with advanced HCC. Although phase III trials of CPI monotherapy have been negative, the combination of PD-1/PD-L1 inhibitors with other anti-angiogenic drugs, CTLA-4 inhibitors or other modalities may result in new treatment options for patients with HCC. Research on predictive biomarkers is crucial for further development of CPIs in HCC.
Collapse
Affiliation(s)
- Anne Dyhl-Polk
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; (M.K.M.); (D.L.N.)
| | - Marta Kramer Mikkelsen
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; (M.K.M.); (D.L.N.)
| | - Morten Ladekarl
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Hobrovej 19-22, 9000 Aalborg, Denmark;
| | - Dorte Lisbet Nielsen
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; (M.K.M.); (D.L.N.)
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
6
|
Merino M, Lozano T, Casares N, Lana H, Troconiz IF, Ten Hagen TLM, Kochan G, Berraondo P, Zalba S, Garrido MJ. Dual activity of PD-L1 targeted Doxorubicin immunoliposomes promoted an enhanced efficacy of the antitumor immune response in melanoma murine model. J Nanobiotechnology 2021; 19:102. [PMID: 33849551 PMCID: PMC8042980 DOI: 10.1186/s12951-021-00846-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/27/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The immunomodulation of the antitumor response driven by immunocheckpoint inhibitors (ICIs) such as PD-L1 (Programmed Death Ligand-1) monoclonal antibody (α-PD-L1) have shown relevant clinical outcomes in a subset of patients. This fact has led to the search for rational combinations with other therapeutic agents such as Doxorubicin (Dox), which cytotoxicity involves an immune activation that may enhance ICI response. Therefore, this study aims to evaluate the combination of chemotherapy and ICI by developing Dox Immunoliposomes functionalized with monovalent-variable fragments (Fab') of α-PD-L1. RESULTS Immunoliposomes were assayed in vitro and in vivo in a B16 OVA melanoma murine cell line over-expressing PD-L1. Here, immune system activation in tumor, spleen and lymph nodes, together with the antitumor efficacy were evaluated. Results showed that immunoliposomes bound specifically to PD-L1+ cells, yielding higher cell interaction and Dox internalization, and decreasing up to 30-fold the IC50, compared to conventional liposomes. This mechanism supported a higher in vivo response. Indeed, immunoliposomes promoted full tumor regression in 20% of mice and increased in 1 month the survival rate. This formulation was the only treatment able to induce significant (p < 0.01) increase of activated tumor specific cytotoxic T lymphocytes at the tumor site. CONCLUSION PD-L1 targeted liposomes encapsulating Dox have proved to be a rational combination able to enhance the modulation of the immune system by blocking PD-L1 and selectively internalizing Dox, thus successfully providing a dual activity offered by both, chemo and immune therapeutic strategies.
Collapse
Affiliation(s)
- María Merino
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy, University of Navarra, 31008, Pamplona, Navarra, Spain
| | - Teresa Lozano
- Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdisNA), Pamplona, Spain
| | - Noelia Casares
- Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdisNA), Pamplona, Spain
| | - Hugo Lana
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy, University of Navarra, 31008, Pamplona, Navarra, Spain
| | - Iñaki F Troconiz
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy, University of Navarra, 31008, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IdisNA), Pamplona, Spain
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Grazyna Kochan
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain.,Department of Oncology, Navarrabiomed-Biomedical Research Centre, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdisNA), Pamplona, Spain
| | - Sara Zalba
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy, University of Navarra, 31008, Pamplona, Navarra, Spain. .,Navarra Institute for Health Research (IdisNA), Pamplona, Spain.
| | - María J Garrido
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy, University of Navarra, 31008, Pamplona, Navarra, Spain. .,Navarra Institute for Health Research (IdisNA), Pamplona, Spain.
| |
Collapse
|
7
|
Jiang J, Liu B, Liu R, Yang W. Overexpression of Taspase 1 Predicts Poor Prognosis in Patients with Hepatocellular Carcinoma. Cancer Manag Res 2021; 13:2517-2537. [PMID: 33758547 PMCID: PMC7981154 DOI: 10.2147/cmar.s296069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Background Taspase 1 (TASP1) is a highly conserved protease involved in site-specific proteolysis. Existing researches have revealed a link between TASP1 expression and carcinogenesis. However, limited data are available regarding the prognosis and functions of TASP1 in hepatocellular carcinoma (HCC). Methods Western Blotting and qRT-PCR were employed to evaluate the level of TASP1 in HCC cell lines and clinical specimens. TASP1 expression was further calculated in clinical specimens by immunohistochemistry and the mRNA level of TASP1 in HCC was analyzed using Oncomine and UALCAN databases. The TASP1 promoter methylation modification was shown via MEXPRESS and UALCAN. The association between TASP1 expression and postoperative prognosis was evaluated using Kaplan–Meier and Cox regression analysis in clinical patients. The effect of TASP1 on HCC prognosis was analyzed via Kaplan-Meier plotter, GEPIA and UALCAN. Additionally, the regulators, kinases, miRNA and transcription factor targets of TASP1 were identified using LinkedOmics. Moreover, cBioPortal was used to detect the genetic alteration of TASP1. Finally, TIMER was utilized to assess the relation between TASP1 and the immune cell infiltration, whereas the correlation of TASP1 with three immune factors was detected through TISIDB. Results TASP1 expression was increased in HCC cell lines and HCC tissues. CNV and DNA methylation of TASP1 were changed. Survival analysis revealed that high TASP1 expression was correlated with overall survival (OS). Functional network analysis about TASP1 in HCC showed that the double-strand break repair, peptidyl-threonine modification, spindle organization, peptidyl-lysine modification and microtubule-based movement were modulated. Furthermore, TASP1 expression revealed puissant relation to the infiltration of immune cells and three immune factors in HCC. Conclusion These data indicate that TASP1 may act as a potential prognostic marker in HCC and regulate HCC via multiple mechanisms.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Gastroenterology and Hepatology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Bin Liu
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Ruilin Liu
- Department of Pulmonary, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Wenzhuo Yang
- Department of Gastroenterology and Hepatology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
8
|
Basile MS, Mazzon E, Fagone P, Longo A, Russo A, Fallico M, Bonfiglio V, Nicoletti F, Avitabile T, Reibaldi M. Immunobiology of Uveal Melanoma: State of the Art and Therapeutic Targets. Front Oncol 2019; 9:1145. [PMID: 31750244 PMCID: PMC6848265 DOI: 10.3389/fonc.2019.01145] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Uveal Melanoma (UM) represents the most common primary intraocular malignant tumor in adults. Although it originates from melanocytes as cutaneous melanoma, it shows significant clinical and biological differences with the latter, including high resistance to immune therapy. Indeed, UM can evade immune surveillance via multiple mechanisms, such as the expression of inhibitory checkpoints (e.g., PD-L1, CD47, CD200) and the production of IDO-1 and soluble FasL, among others. More in-depth understanding of these mechanisms will suggest potential targets for the design of novel and more effective management strategies for UM patients.
Collapse
Affiliation(s)
- Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, Messina, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonio Longo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Andrea Russo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Matteo Fallico
- Department of Ophthalmology, University of Catania, Catania, Italy
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Michele Reibaldi
- Department of Ophthalmology, University of Catania, Catania, Italy
| |
Collapse
|
9
|
Henriksen A, Dyhl-Polk A, Chen I, Nielsen D. Checkpoint inhibitors in pancreatic cancer. Cancer Treat Rev 2019; 78:17-30. [DOI: 10.1016/j.ctrv.2019.06.005] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/18/2022]
|
10
|
Hu W, Zi Z, Jin Y, Li G, Shao K, Cai Q, Ma X, Wei F. CRISPR/Cas9-mediated PD-1 disruption enhances human mesothelin-targeted CAR T cell effector functions. Cancer Immunol Immunother 2019; 68:365-377. [PMID: 30523370 PMCID: PMC11028344 DOI: 10.1007/s00262-018-2281-2] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 12/01/2018] [Indexed: 12/30/2022]
Abstract
The interaction between programmed cell death protein 1 (PD-1) on activated T cells and its ligands on a target tumour may limit the capacity of chimeric antigen receptor (CAR) T cells to eradicate solid tumours. PD-1 blockade could potentially enhance CAR T cell function. Here, we show that mesothelin is overexpressed in human triple-negative breast cancer cells and can be targeted by CAR T cells. To overcome the suppressive effect of PD-1 on CAR T cells, we utilized CRISPR/Cas9 ribonucleoprotein-mediated editing to disrupt the programmed cell death-1 (PD-1) gene locus in human primary T cells, resulting in a significantly reduced PD-1hi population. This reduction had little effect on CAR T cell proliferation but strongly augmented CAR T cell cytokine production and cytotoxicity towards PD-L1-expressing cancer cells in vitro. CAR T cells with PD-1 disruption show enhanced tumour control and relapse prevention in vivo when compared with CAR T cells with or without αPD-1 antibody blockade. Our study demonstrates a potential advantage of integrated immune checkpoint blockade with CAR T cells in controlling solid tumours and provides an alternative CAR T cell strategy for adoptive transfer therapy.
Collapse
Affiliation(s)
- Wanghong Hu
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China
| | - Zhenguo Zi
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China
| | - Yanling Jin
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China
| | - Gaoxin Li
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China
| | - Kang Shao
- Affiliated Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qiliang Cai
- MOE and MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaojing Ma
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China
| | - Fang Wei
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China.
| |
Collapse
|
11
|
Merino M, Contreras A, Casares N, Troconiz IF, Ten Hagen TL, Berraondo P, Zalba S, Garrido MJ. A new immune-nanoplatform for promoting adaptive antitumor immune response. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:13-25. [PMID: 30654186 DOI: 10.1016/j.nano.2018.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 01/02/2023]
Abstract
Immunoliposomes (ILs), obtained with monoclonal antibodies (mAbs) decorating the liposome surface, are used for cancer treatment. These mAbs provide the recognition of molecules upregulated in cancer cells, like Programmed Death-Ligand 1 (PD-L1), an immune-checkpoint involved in tumor resistance, forming a complex that blocks this molecule and thereby, induces antitumor immune response. This mechanism introduces a new concept for ILs. ILs coupled to anti-PD-L1 or its Fab' fragment have been developed and in vitro/in vivo characterized. Factors such as coupling methods, PEG density and ligand size were optimized. In vitro data showed that Fab'-ILs displayed the highest PD-L1 cell interaction, correlating with a higher in vivo tumor accumulation and an increase of effector cytotoxic CD8+ T cells, providing tumor shrinkage and total regression in 20% of mice. Therefore, a novel immune-nanoplatform able to modulate the immune system has been developed, allowing the encapsulation of several agents for combinatorial therapies.
Collapse
Affiliation(s)
- María Merino
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdisNA)
| | - Ana Contreras
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdisNA)
| | - Noelia Casares
- Program of Immunology and Immunotherapy, CIMA, Pamplona, Spain; Navarra Institute for Health Research (IdisNA)
| | - Iñaki F Troconiz
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdisNA)
| | - Timo Lm Ten Hagen
- Laboratory of Experimental Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, CIMA, Pamplona, Spain; Navarra Institute for Health Research (IdisNA)
| | - Sara Zalba
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdisNA)
| | - María J Garrido
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdisNA).
| |
Collapse
|
12
|
Immune checkpoint blockade and its combination therapy with small-molecule inhibitors for cancer treatment. Biochim Biophys Acta Rev Cancer 2018; 1871:199-224. [PMID: 30605718 DOI: 10.1016/j.bbcan.2018.12.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 02/05/2023]
Abstract
Initially understood for its physiological maintenance of self-tolerance, the immune checkpoint molecule has recently been recognized as a promising anti-cancer target. There has been considerable interest in the biology and the action mechanism of the immune checkpoint therapy, and their incorporation with other therapeutic regimens. Recently the small-molecule inhibitor (SMI) has been identified as an attractive combination partner for immune checkpoint inhibitors (ICIs) and is becoming a novel direction for the field of combination drug design. In this review, we provide a systematic discussion of the biology and function of major immune checkpoint molecules, and their interactions with corresponding targeting agents. With both preclinical studies and clinical trials, we especially highlight the ICI + SMI combination, with its recent advances as well as its application challenges.
Collapse
|
13
|
Ahmed A, Adiga V, Nayak S, Uday Kumar JAJ, Dhar C, Sahoo PN, Sundararaj BK, Souza GD, Vyakarnam A. Circulating HLA-DR+CD4+ effector memory T cells resistant to CCR5 and PD-L1 mediated suppression compromise regulatory T cell function in tuberculosis. PLoS Pathog 2018; 14:e1007289. [PMID: 30231065 PMCID: PMC6166982 DOI: 10.1371/journal.ppat.1007289] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 10/01/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Chronic T cell activation is a hallmark of pulmonary tuberculosis (PTB). The mechanisms underpinning this important phenomenon are however, poorly elucidated, though known to rely on control of T effector cells (Teff) by regulatory T cells (Treg). Our studies show that circulating natural Treg cells in adults with PTB preserve their suppressive potential but Teff cells from such subjects are resistant to Treg-mediated suppression. We found this to be due to expansion of an activated Teff subset identified by Human Leukocyte Antigen (HLA)-DR expression. Sensitivity to suppression was restored to control levels by depletion of this subset. Comparative transcriptome analysis of Teff cells that contain HLA-DR+ cells versus the fraction depleted of this population identified putative resistance mechanisms linked to IFNG, IL17A, IL22, PD-L1 and β-chemokines CCL3L3, CCL4 expression. Antibody blocking experiments confirmed HLA-DR+ Teff cells, but not the fraction depleted of HLA-DR+ effectors, to be resistant to Treg suppression mediated via CCR5 and PD-L1 associated pathways. In the presence of HLA-DR+ Teff cells, activation of NFκB downstream of CCR5 and PD-L1 was perturbed. In addition, HLA-DR+ Teff cells expressed significantly higher levels of Th1/Th17 cytokines that may regulate Treg function through a reciprocal counter-balancing relationship. Taken together, our study provides novel insight on how activated HLA-DR+CD4+ T cells may contribute to disease associated inflammation by compromising Treg-mediated suppression in PTB. An important marker of progression to PTB following Mycobacterium tuberculosis (Mtb) infection in humans is elevated frequencies of HLA-DR+CD4+ T cells, reflecting chronic T cell activation. However, the mechanisms by which activated HLA-DR+CD4+ T cells contribute to disease process is not known. We show that CD25- HLA-DR+CD4+ memory Teff from PTB patients are resistant to suppression mediated by Treg cells. An unbiased transcriptome analysis identified several key pathways that contribute to this resistance. Specifically, presence of HLA-DR+CD4+ T cells renders the effector population resistant to CCR5 and PD-L1 mediated suppression by Treg cells. In addition, the HLA-DR+CD4+ memory Teff cells express elevated levels of Th1/Th17 cytokines known to counter-regulate and dampen Treg suppression. These findings provide fresh insight to disease process in TB and identify HLA-DR+ Teff resistant to Treg suppression as a potential functional marker of disease.
Collapse
Affiliation(s)
- Asma Ahmed
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Soumya Nayak
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | | | - Chirag Dhar
- Division of Infectious Diseases, St John’s Research Institute, Bangalore, India
| | - Pravat Nalini Sahoo
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Bharath K. Sundararaj
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - George D. Souza
- Dept. of Pulmonary Medicine & Division of Infectious Diseases, St John’s Research Institute, Bangalore, India
| | - Annapurna Vyakarnam
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Department of Infectious Diseases, King’s College London, London, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, Guy's Campus, London, United Kingdom
- * E-mail: ,
| |
Collapse
|
14
|
Checkpoint inhibitors in breast cancer – Current status. Cancer Treat Rev 2018; 63:122-134. [DOI: 10.1016/j.ctrv.2017.12.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
|
15
|
Lauss M, Donia M, Harbst K, Andersen R, Mitra S, Rosengren F, Salim M, Vallon-Christersson J, Törngren T, Kvist A, Ringnér M, Svane IM, Jönsson G. Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nat Commun 2017; 8:1738. [PMID: 29170503 PMCID: PMC5701046 DOI: 10.1038/s41467-017-01460-0] [Citation(s) in RCA: 331] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 09/19/2017] [Indexed: 01/01/2023] Open
Abstract
Adoptive T-cell therapy (ACT) is a highly intensive immunotherapy regime that has yielded remarkable response rates and many durable responses in clinical trials in melanoma; however, 50-60% of the patients have no clinical benefit. Here, we searched for predictive biomarkers to ACT in melanoma. Whole exome- and transcriptome sequencing and neoantigen prediction were applied to pre-treatment samples from 27 patients recruited to a clinical phase I/II trial of ACT in stage IV melanoma. All patients had previously progressed on other immunotherapies. We report that clinical benefit is associated with significantly higher predicted neoantigen load. High mutation and predicted neoantigen load are significantly associated with improved progression-free and overall survival. Further, clinical benefit is associated with the expression of immune activation signatures including a high MHC-I antigen processing and presentation score. These results improve our understanding of mechanisms behind clinical benefit of ACT in melanoma.
Collapse
Affiliation(s)
- Martin Lauss
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Scheelegatan 2, Medicon Village, 22185, Lund, Sweden
| | - Marco Donia
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Ringvej 75, 2730, Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Katja Harbst
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Scheelegatan 2, Medicon Village, 22185, Lund, Sweden
| | - Rikke Andersen
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Ringvej 75, 2730, Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Shamik Mitra
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Scheelegatan 2, Medicon Village, 22185, Lund, Sweden
| | - Frida Rosengren
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Scheelegatan 2, Medicon Village, 22185, Lund, Sweden
| | - Maryem Salim
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Scheelegatan 2, Medicon Village, 22185, Lund, Sweden
| | - Johan Vallon-Christersson
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Scheelegatan 2, Medicon Village, 22185, Lund, Sweden
| | - Therese Törngren
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Scheelegatan 2, Medicon Village, 22185, Lund, Sweden
| | - Anders Kvist
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Scheelegatan 2, Medicon Village, 22185, Lund, Sweden
| | - Markus Ringnér
- Department of Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Lund University, Sölvegatan 35, 22362, Lund, Sweden
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Ringvej 75, 2730, Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Göran Jönsson
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Scheelegatan 2, Medicon Village, 22185, Lund, Sweden
| |
Collapse
|
16
|
Kolloli A, Subbian S. Host-Directed Therapeutic Strategies for Tuberculosis. Front Med (Lausanne) 2017; 4:171. [PMID: 29094039 PMCID: PMC5651239 DOI: 10.3389/fmed.2017.00171] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/26/2017] [Indexed: 12/18/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a leading cause of morbidity and mortality in humans worldwide. Currently, the standard treatment for TB involves multiple antibiotics administered for at least 6 months. Although multiple antibiotics therapy is necessary to prevent the development of drug resistance, the prolonged duration of treatment, combined with toxicity of drugs, contributes to patient non-compliance that can leads to the development of drug-resistant Mtb (MDR and XDR) strains. The existence of comorbid conditions, including HIV infection, not only complicates TB treatment but also elevates the mortality rate of patients. These facts underscore the need for the development of new and/or improved TB treatment strategies. Host-directed therapy (HDT) is a new and emerging concept in the treatment of TB, where host response is modulated by treatment with small molecules, with or without adjunct antibiotics, to achieve better control of TB. Unlike antibiotics, HDT drugs act by directly modulating host cell functions; therefore, development of drug resistance by infecting Mtb is avoided. Thus, HDT is a promising treatment strategy for the management of MDR- and XDR-TB cases as well as for patients with existing chronic, comorbid conditions such as HIV infection or diabetes. Functionally, HDT drugs fine-tune the antimicrobial activities of host immune cells and limit inflammation and tissue damage associated with TB. However, current knowledge and clinical evidence is insufficient to implement HDT molecules as a stand-alone, without adjunct antibiotics, therapeutic modality to treat any form of TB in humans. In this review, we discuss the recent findings on small molecule HDT agents that target autophagy, vitamin D pathway, and anti-inflammatory response as adjunctive agents along with standard antibiotics for TB therapy. Data from recent publications show that this approach has the potential to improve clinical outcome and can help to reduce treatment duration. Thus, HDT can contribute to global TB control programs by potentially increasing the efficiency of anti-TB treatment.
Collapse
Affiliation(s)
- Afsal Kolloli
- Public Health Research Institute (PHRI) at New Jersey Medical School, Rutgers Biomedical and Health Sciences (RBHS), Rutgers University, The State University of New Jersey, Newark, NJ, United States
| | - Selvakumar Subbian
- Public Health Research Institute (PHRI) at New Jersey Medical School, Rutgers Biomedical and Health Sciences (RBHS), Rutgers University, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
17
|
Rao M, Valentini D, Dodoo E, Zumla A, Maeurer M. Anti-PD-1/PD-L1 therapy for infectious diseases: learning from the cancer paradigm. Int J Infect Dis 2017; 56:221-228. [PMID: 28163164 DOI: 10.1016/j.ijid.2017.01.028] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/19/2017] [Accepted: 01/22/2017] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Immune checkpoint pathways regulate optimal host immune responses against transformed cells, induce immunological memory, and limit tissue pathology. Conversely, aberrant immune checkpoint activity signifies a poor prognosis in cancer and infectious diseases. Host-directed therapy (HDT) via immune checkpoint blockade has revolutionized cancer treatment with therapeutic implications for chronic infections, thus laying the foundation for this review. METHODS Online literature searches were performed via PubMed, PubMed Central, and Google using the keywords "immune checkpoint inhibition"; "host-directed therapy"; "T cell exhaustion"; "cancer immunotherapy"; "anti-PD-1 therapy"; "anti-PD-L1 therapy"; "chronic infections"; "antigen-specific cells"; "tuberculosis"; "malaria"; "viral infections"; "human immunodeficiency virus"; "hepatitis B virus"; "hepatitis C virus"; "cytomegalovirus" and "Epstein-Barr virus". Search results were filtered based on relevance to the topics covered in this review. RESULTS The use of monoclonal antibodies directed against the antigen-experienced T-cell marker programmed cell death 1 (PD-1) and its ligand PD-L1 in the context of chronic infectious diseases is reviewed. The potential pitfalls and precautions, based on clinical experience from treating patients with cancer with PD-1/PD-L1 pathway inhibitors, are also described. CONCLUSIONS Anti-PD-1/PD-L1 therapy holds promise as adjunctive therapy for chronic infectious diseases such as tuberculosis and HIV, and must therefore be tested in randomized clinical trials.
Collapse
Affiliation(s)
- Martin Rao
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | - Davide Valentini
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden; Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ernest Dodoo
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden; Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London, and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK
| | - Markus Maeurer
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden; Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
18
|
Dutoit V, Migliorini D, Dietrich PY, Walker PR. Immunotherapy of Malignant Tumors in the Brain: How Different from Other Sites? Front Oncol 2016; 6:256. [PMID: 28003994 PMCID: PMC5141244 DOI: 10.3389/fonc.2016.00256] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/24/2016] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy is now advancing at remarkable pace for tumors located in various tissues, including the brain. Strategies launched decades ago, such as tumor antigen-specific therapeutic vaccines and adoptive transfer of tumor-infiltrating lymphocytes are being complemented by molecular engineering approaches allowing the development of tumor-specific TCR transgenic and chimeric antigen receptor T cells. In addition, the spectacular results obtained in the last years with immune checkpoint inhibitors are transfiguring immunotherapy, these agents being used both as single molecules, but also in combination with other immunotherapeutic modalities. Implementation of these various strategies is ongoing for more and more malignancies, including tumors located in the brain, raising the question of the immunological particularities of this site. This may necessitate cautious selection of tumor antigens, minimizing the immunosuppressive environment and promoting efficient T cell trafficking to the tumor. Once these aspects are taken into account, we might efficiently design immunotherapy for patients suffering from tumors located in the brain, with beneficial clinical outcome.
Collapse
Affiliation(s)
- Valérie Dutoit
- Laboratory of Tumor Immunology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| | - Denis Migliorini
- Oncology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| | - Pierre-Yves Dietrich
- Oncology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| | - Paul R Walker
- Laboratory of Tumor Immunology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| |
Collapse
|
19
|
Donia M, Pedersen M, Svane IM. Cancer immunotherapy in patients with preexisting autoimmune disorders. Semin Immunopathol 2016; 39:333-337. [DOI: 10.1007/s00281-016-0595-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/19/2016] [Indexed: 12/19/2022]
|
20
|
Millet A, Martin AR, Ronco C, Rocchi S, Benhida R. Metastatic Melanoma: Insights Into the Evolution of the Treatments and Future Challenges. Med Res Rev 2016; 37:98-148. [PMID: 27569556 DOI: 10.1002/med.21404] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/28/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023]
Abstract
Melanoma is the deadliest form of skin cancer. While associated survival prognosis is good when diagnosed early, it dramatically drops when melanoma progresses into its metastatic form. Prior to 2011, the favored therapies include interleukin-2 and chemotherapies, regardless of their low efficiency and their toxicity. Following key biological findings, two new types of therapy have been approved. First, there are the targeted therapies, which rely on small molecule B-Raf and MEK inhibitors and allow the treatment of patients with B-Raf mutated melanoma. Second, there are the immunotherapies, with anti-CTLA-4 and anti-PD-1 antibodies that are used for patients harboring a B-Raf wild-type status. Both approaches have significantly improved patient survival, compared with alkylating agents, in the treatment of unresectable melanoma. Herein, we review the evolution of the treatment of melanoma starting from early discoveries to current therapies. A focus will be provided on drug discovery, synthesis, and mode of action of relevant drugs and the future directions of the domain to overcome the emergence of the resistance events.
Collapse
Affiliation(s)
- Antoine Millet
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Nice, France
| | | | - Cyril Ronco
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Nice, France
| | - Stéphane Rocchi
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Equipe Biologie et Pathologie des cellules mélanocytaires: de la pigmentation cutanée au mélanome, Nice, France.,Université de Nice Sophia Antipolis, UFR de Médecine, Nice, France.,Service de Dermatologie, Hôpital Archet II, CHU Nice, France
| | - Rachid Benhida
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Nice, France
| |
Collapse
|
21
|
|
22
|
Munir Ahmad S, Martinenaite E, Hansen M, Junker N, Borch TH, Met Ö, Donia M, Svane IM, Andersen MH. PD-L1 peptide co-stimulation increases immunogenicity of a dendritic cell-based cancer vaccine. Oncoimmunology 2016; 5:e1202391. [PMID: 27622072 DOI: 10.1080/2162402x.2016.1202391] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/12/2016] [Accepted: 06/12/2016] [Indexed: 12/31/2022] Open
Abstract
We recently described naturally occurring PD-L1-specific T cells that recognize PD-L1-expressing immune cells as well as malignant cells. In the present study, we investigated whether the immunogenicity of a dendritic cell (DC)-based vaccine could be influenced by co-stimulation with a known PD-L1-derived epitope. We incubated a PD-L1-derived peptide epitope (19 amino acids long) or a control peptide (an irrelevant HIV epitope) with peripheral blood mononuclear cells from patients with malignant melanoma who had received a DC-based vaccine. We observed a significantly higher number of T cells that reacted to the vaccine in cultures that had been co-stimulated with the PD-L1 peptide epitope compared to cultures incubated with control peptide. Next, we characterized a novel PD-L1-derived epitope (23 amino acids long) and found that co-stimulation with both PD-L1 epitopes boosted the immune response elicited by the DC vaccine even further. Consequently, we observed a significant increase in the number of vaccine-reacting T cells in vitro. In conclusion, activation of PD-L1-specific T cells may directly modulate immunogenicity of DC vaccines. Addition of PD-L1 epitopes may thus be an easily applicable and attractive option to augment the effectiveness of cancer vaccines and other immunotherapeutic agents.
Collapse
Affiliation(s)
- Shamaila Munir Ahmad
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital , Herlev, Denmark
| | - Evelina Martinenaite
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital , Herlev, Denmark
| | - Morten Hansen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital , Herlev, Denmark
| | - Niels Junker
- Department of Oncology, Copenhagen University Hospital , Herlev, Denmark
| | - Troels Holz Borch
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Özcan Met
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Marco Donia
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Zumla A, Rao M, Dodoo E, Maeurer M. Potential of immunomodulatory agents as adjunct host-directed therapies for multidrug-resistant tuberculosis. BMC Med 2016; 14:89. [PMID: 27301245 PMCID: PMC4908783 DOI: 10.1186/s12916-016-0635-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/02/2016] [Indexed: 01/15/2023] Open
Abstract
Treatment of multidrug-resistant tuberculosis (MDR-TB) is extremely challenging due to the virulence of the etiologic strains of Mycobacterium tuberculosis (M. tb), the aberrant host immune responses and the diminishing treatment options with TB drugs. New treatment regimens incorporating therapeutics targeting both M. tb and host factors are urgently needed to improve the clinical management outcomes of MDR-TB. Host-directed therapies (HDT) could avert destructive tuberculous lung pathology, facilitate eradication of M. tb, improve survival and prevent long-term functional disability. In this review we (1) discuss the use of HDT for cancer and other infections, drawing parallels and the precedent they set for MDR-TB treatment, (2) highlight preclinical studies of pharmacological agents commonly used in clinical practice which have HDT potential, and (3) outline developments in cellular therapy to promote clinically beneficial immunomodulation to improve treatment outcomes in patients with pulmonary MDR-TB. The use of HDTs as adjuncts to MDR-TB therapy requires urgent evaluation.
Collapse
Affiliation(s)
- Alimuddin Zumla
- Division of Infection and Immunity, University College London, and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK
| | - Martin Rao
- F79, Therapeutic Immunology (TIM) division, Department of Laboratory Medicine (LABMED), Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Ernest Dodoo
- F79, Therapeutic Immunology (TIM) division, Department of Laboratory Medicine (LABMED), Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Markus Maeurer
- F79, Therapeutic Immunology (TIM) division, Department of Laboratory Medicine (LABMED), Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden. .,Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
24
|
Zhu X, Lang J. The significance and therapeutic potential of PD-1 and its ligands in ovarian cancer: A systematic review. Gynecol Oncol 2016; 142:184-189. [PMID: 27063803 DOI: 10.1016/j.ygyno.2016.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/28/2016] [Accepted: 04/01/2016] [Indexed: 02/07/2023]
Abstract
Surgery, radiotherapy and chemotherapy are the mainstay of malignant cancer treatments. However, with the development of immunology, the emerging immunotherapy represents a rational and alternative approach for the treatment of human cancer, including ovarian cancer (OC). Based on a body of evidence and the clinical success of immunotherapy in many malignancies, it is confirmed that blocking the programmed death 1 (PD-1) and its ligands in OC is feasible and valid both in animal models and patients. Immunotherapy may play a significant role in the future clinical management and improve the prognosis of OC. This review will focus on the biological functions, treatment response, toxicity and viable target of PD-1 and its ligands in OC. Recognition of the multiple functions of PD-1 and its ligands in ovarian cancer will serve to deepen our understanding of the nature of OC, develop novel immunotherapy approaches and discover possible diagnostic and prognostic biomarkers in future clinical decisions.
Collapse
Affiliation(s)
- Xinxin Zhu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
25
|
Sun Z, Zhu Y, Xia J, Sawakami T, Kokudo N, Zhang N. Status of and prospects for cancer vaccines against hepatocellular carcinoma in clinical trials. Biosci Trends 2015; 10:85-91. [PMID: 26522694 DOI: 10.5582/bst.2015.01128] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Current therapies to treat advanced hepatocellular carcinoma (HCC) are not satisfactory because of the high rate of recurrence after treatment and because of severe complications after surgery. Cancer vaccines have been studied for decades to achieve effective, micro-invasive, long-lasting anti-tumor action. Cancer vaccines are designed to promote tumor-specific immune responses and increase specific cytotoxic CD8-positive T cells. This review summarizes 16 phase I clinical trials of cancer vaccines against HCC that have been conducted over the past 10 years. According to those trials, the Alpha fetoprotein (AFP), Glypican-3 (GPC3), and Multidrug resistance-associated protein 3 (MRP3) vaccines were well tolerated and safe. Some early clinical trials have shown that vaccination resulted in a large number of T cells activated by a specific tumor-associated antigen in the circulation, but clinical outcomes were not satisfactory. This may be because targets for immunosuppressive agents have yet to be clearly determined in HCC. Therapeutic regimens that combine activative agents and suppressive agents may profoundly improve clinical outcomes for patients with HCC in the future.
Collapse
Affiliation(s)
- Zhipeng Sun
- Beijing Key Lab of Therapeutic Cancer Vaccines, Peking University Ninth School of Clinical Medicine (Cancer Center, BeijingShijitan Hospital, Capital Medical University)
| | | | | | | | | | | |
Collapse
|