1
|
Rosenthal I, Padler-Karavani V. Current Landscape of Therapeutic Cancer Vaccines. Methods Mol Biol 2025; 2926:1-14. [PMID: 40266513 DOI: 10.1007/978-1-0716-4542-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Cancer vaccines aim to harness the immune system for prevention or treatment of the disease. This is achieved by immunizing against target antigens that are specific/associated with cancer. Myriad of particulate materials or cell-based strategies have been developed to induce immune responses against cancer antigens, using protein/peptide antigens, DNA or RNA, viral antigens, viral or bacterial vectors, embryonic material, whole tumor lysate, dendritic cells, red blood cells, exosomes, liposomes, engineered bioinspired and biomimetic vaccines and glycosylation-based vaccines. Several key factors affect the safety and efficacy of therapeutic cancer vaccines, including identification of potent neoantigens, dosage, adjuvant and regimen of immunization, route of administration and delivery methods, as well as overcoming intrinsic and extrinsic resistance mechanisms. More recently, novel approaches for improving cancer vaccines have been explored, such as intratumoral vaccinations, oncolytic viruses, and combination immunotherapies. This chapter aims to provide a glimpse into some of the most common approaches for therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Inbal Rosenthal
- Department of Cell Research and Immunology, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Vered Padler-Karavani
- Department of Cell Research and Immunology, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
2
|
Shi R, Ran L, Tian Y, Guo W, Zhao L, Jin S, Cheng J, Zhang Z, Ma Y. Prospects and challenges of neoantigen applications in oncology. Int Immunopharmacol 2024; 143:113329. [PMID: 39405926 DOI: 10.1016/j.intimp.2024.113329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/11/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Neoantigen, unique peptides resulting from tumor-specific mutations, represent a promising frontier in oncology for personalized cancer immunotherapy. Their unique features allow for the development of highly specific and effective cancer treatments, which can potentially overcome the limitations of conventional therapies. This paper explores the current prospects and challenges associated with the application of neoantigens in oncology. We examine the latest advances in neoantigen identification, vaccine development, and adoptive T cell therapy. Additionally, we discuss the obstacles related to neoantigen heterogeneity, immunogenicity prediction, and the tumor microenvironment. Through a comprehensive analysis of current research and clinical trials, this paper aims to provide a detailed overview of how neoantigens could revolutionize cancer treatment and the hurdles that must be overcome to realize their full potential.
Collapse
Affiliation(s)
- Ranran Shi
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Ling Ran
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Yuan Tian
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Wei Guo
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China
| | - Lifang Zhao
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Shaoju Jin
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Jiang Cheng
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan 750000, China
| | - Zhe Zhang
- School of Sciences, Henan University of Technology, Zhengzhou 450001, China.
| | - Yongchao Ma
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China.
| |
Collapse
|
3
|
Wang Y, Hu M, Finn OJ, Wang XS. Tumor-Associated Antigen Burden Correlates with Immune Checkpoint Blockade Benefit in Tumors with Low Levels of T-cell Exhaustion. Cancer Immunol Res 2024; 12:1589-1602. [PMID: 39137006 PMCID: PMC11534523 DOI: 10.1158/2326-6066.cir-23-0932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 03/20/2024] [Accepted: 08/09/2024] [Indexed: 09/14/2024]
Abstract
Tumor-associated antigens (TAA) are important targets for cancer vaccines. However, TAA-based vaccines have not yet achieved their full potential in clinical trials. In contrast, immune checkpoint blockade (ICB) has emerged as an effective therapy, leading to durable responses in selected patients with cancer. To date, few generalizable associations between TAAs and ICB benefit have been reported, with most studies focusing on melanoma, which has the highest mutation rate in cancer. In this study, we developed a TAA burden (TAB) algorithm based on known and putative TAAs and investigated the association of TAB with ICB benefit. Analysis of the IMvigor210 patient cohort of urothelial carcinoma treated with anti-PDL1 revealed that high tumor mutation burden weakened the association of TAB with ICB benefit. Furthermore, TAB correlated with ICB efficacy in tumors characterized by negative PDL1 staining on immune cells; however, high levels of PDL1 staining on immune cells were linked to T-cell exhaustion. Validation across independent clinical datasets-including urothelial carcinoma cohorts treated with anti-PD1/PDL1 agents and neoadjuvant anti-PD1 trials for head and neck cancers-corroborated the finding that TAB correlates with ICB benefit in tumors with low T-cell exhaustion. Pan-cancer analyses revealed that in most cancer entities, tumors with higher T-cell exhaustion exhibited lower TAB levels, implying possible immunoediting of TAAs in tumors with established antitumor immunity. Our study challenges the prevailing notion of a lack of association between TAAs and ICB response. It also underscores the need for future investigations into the immunogenicity of TAAs and TAA-based vaccine strategies in tumors with low levels of T-cell exhaustion.
Collapse
Affiliation(s)
- Yue Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
| | - Mengying Hu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
| | - Olivera J Finn
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, U.S.A
| | - Xiao-Song Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
| |
Collapse
|
4
|
Makani VKK, Joglekar AV. Living in Syn: T Cell Antigen Identification Based on Synapse Sequencing. Cancer Immunol Res 2024; 12:515. [PMID: 38557780 DOI: 10.1158/2326-6066.cir-24-0239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
The pivotal role of T cell responses has been well studied in both protective and destructive scenarios. T cells recognize peptide epitopes presented on Human Leukocyte Antigens (HLA) through their surface T cell receptors (TCR). Advances in single-cell RNA sequencing have identified millions of TCRs, but only a minuscule fraction of them have known epitopes. Recently, cell-based T cell antigen discovery platforms have emerged onto the landscape. Here, Jin and colleagues, report a novel antigen discovery platform called Tsyn-seq that relies on sequencing TCR-peptide-HLA-induced synapses for genome-wide epitope screening. See related article by Jin et al., p. 530 (3).
Collapse
Affiliation(s)
- Venkata Krishna Kanth Makani
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Alok V Joglekar
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
5
|
Li Y, Rezvani K, Rafei H. Next-generation chimeric antigen receptors for T- and natural killer-cell therapies against cancer. Immunol Rev 2023; 320:217-235. [PMID: 37548050 PMCID: PMC10841677 DOI: 10.1111/imr.13255] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
Adoptive cellular therapy using chimeric antigen receptor (CAR) T cells has led to a paradigm shift in the treatment of various hematologic malignancies. However, the broad application of this approach for myeloid malignancies and solid cancers has been limited by the paucity and heterogeneity of target antigen expression, and lack of bona fide tumor-specific antigens that can be targeted without cross-reactivity against normal tissues. This may lead to unwanted on-target off-tumor toxicities that could undermine the desired antitumor effect. Recent advances in synthetic biology and genetic engineering have enabled reprogramming of immune effector cells to enhance their selectivity toward tumors, thus mitigating on-target off-tumor adverse effects. In this review, we outline the current strategies being explored to improve CAR selectivity toward tumor cells with a focus on natural killer (NK) cells, and the progress made in translating these strategies to the clinic.
Collapse
Affiliation(s)
- Ye Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hind Rafei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
6
|
Les I, Martínez M, Pérez-Francisco I, Cabero M, Teijeira L, Arrazubi V, Torrego N, Campillo-Calatayud A, Elejalde I, Kochan G, Escors D. Predictive Biomarkers for Checkpoint Inhibitor Immune-Related Adverse Events. Cancers (Basel) 2023; 15:1629. [PMID: 36900420 PMCID: PMC10000735 DOI: 10.3390/cancers15051629] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Immune-checkpoint inhibitors (ICIs) are antagonists of inhibitory receptors in the immune system, such as the cytotoxic T-lymphocyte-associated antigen-4, the programmed cell death protein-1 and its ligand PD-L1, and they are increasingly used in cancer treatment. By blocking certain suppressive pathways, ICIs promote T-cell activation and antitumor activity but may induce so-called immune-related adverse events (irAEs), which mimic traditional autoimmune disorders. With the approval of more ICIs, irAE prediction has become a key factor in improving patient survival and quality of life. Several biomarkers have been described as potential irAE predictors, some of them are already available for clinical use and others are under development; examples include circulating blood cell counts and ratios, T-cell expansion and diversification, cytokines, autoantibodies and autoantigens, serum and other biological fluid proteins, human leucocyte antigen genotypes, genetic variations and gene profiles, microRNAs, and the gastrointestinal microbiome. Nevertheless, it is difficult to generalize the application of irAE biomarkers based on the current evidence because most studies have been retrospective, time-limited and restricted to a specific type of cancer, irAE or ICI. Long-term prospective cohorts and real-life studies are needed to assess the predictive capacity of different potential irAE biomarkers, regardless of the ICI type, organ involved or cancer site.
Collapse
Affiliation(s)
- Iñigo Les
- Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Autoimmune Diseases Unit, Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Inflammatory and Immune-Mediated Diseases Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - Mireia Martínez
- Osakidetza Basque Health Service, Department of Medical Oncology, Araba University Hospital, 01009 Vitoria-Gasteiz, Spain
- Lung Cancer Research Group, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Inés Pérez-Francisco
- Breast Cancer Research Group, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - María Cabero
- Clinical Trials Platform, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Lucía Teijeira
- Medical Oncology Department, Navarre University Hospital, 31008 Pamplona, Spain
| | - Virginia Arrazubi
- Medical Oncology Department, Navarre University Hospital, 31008 Pamplona, Spain
| | - Nuria Torrego
- Osakidetza Basque Health Service, Department of Medical Oncology, Araba University Hospital, 01009 Vitoria-Gasteiz, Spain
- Lung Cancer Research Group, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Ana Campillo-Calatayud
- Inflammatory and Immune-Mediated Diseases Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - Iñaki Elejalde
- Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Autoimmune Diseases Unit, Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Inflammatory and Immune-Mediated Diseases Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - Grazyna Kochan
- Oncoimmunology Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - David Escors
- Oncoimmunology Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| |
Collapse
|
7
|
Zheng P, Yang Y, Fu Y, He J, Hu Y, Zheng X, Duan B, Wang M, Liu Q, Li W, Li D, Yang Y, Yang Z, Yang X, Huang W, Ma Y. Engineered Norovirus-Derived Nanoparticles as a Plug-and-Play Cancer Vaccine Platform. ACS NANO 2023; 17:3412-3429. [PMID: 36779845 DOI: 10.1021/acsnano.2c08840] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
In recent years, virus-derived self-assembled protein nanoparticles (NPs) have emerged as attractive antigen delivery platforms for developing both preventive and therapeutic vaccines. In this study, we exploited the genetically engineered Norovirus S domain (Nov-S) with SpyCatcher003 fused to the C-terminus to develop a robust, modular, and versatile NP-based carrier platform (Nov-S-Catcher003). The NPs can be conveniently armed in a plug-and-play pattern with SpyTag003-linked antigens. Nov-S-Catcher003 was efficiently expressed in Escherichia coli and self-assembled into highly uniform NPs with a purified protein yield of 97.8 mg/L. The NPs presented high stability at different maintained temperatures and after undergoing differing numbers of freeze-thaw cycles. Tumor vaccine candidates were easily obtained by modifying Nov-S-Catcher003 NPs with SpyTag003-linked tumor antigens. Nov-S-Catcher003-antigen NPs significantly promoted the maturation of bone marrow-derived dendritic cells in vitro and were capable of efficiently migrating to lymph nodes in vivo. In TC-1 and B16F10 tumor-bearing mice, the subcutaneous immunization of NPs elicited robust tumor-specific T-cell immunity, reshaped the tumor microenvironment, and inhibited tumor growth. In the TC-1 model, the NPs even completely abolished established tumors. In conclusion, the Nov-S-Catcher003 system is a promising delivery platform for facilitating the development of NP-based cancer vaccines.
Collapse
Affiliation(s)
- Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Ying Yang
- Cell Biology & Molecular Biology Laboratory of Experimental Teaching Center, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- Kunming Medical University, Kunming 650500, China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- Kunming Medical University, Kunming 650500, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Centers for Disease Control and Prevention, Kunming 650034, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| |
Collapse
|
8
|
Duhen T, Gough MJ, Leidner RS, Stanton SE. Development and therapeutic manipulation of the head and neck cancer tumor environment to improve clinical outcomes. FRONTIERS IN ORAL HEALTH 2022; 3:902160. [PMID: 35937775 PMCID: PMC9354490 DOI: 10.3389/froh.2022.902160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical response to cancer therapies involves the complex interplay between the systemic, tumoral, and stromal immune response as well as the direct impact of treatments on cancer cells. Each individual's immunological and cancer histories are different, and their carcinogen exposures may differ. This means that even though two patients with oral tumors may carry an identical mutation in TP53, they are likely to have different pre-existing immune responses to their tumors. These differences may arise due to their distinct accessory mutations, genetic backgrounds, and may relate to clinical factors including previous chemotherapy exposure and concurrent medical comorbidities. In isolation, their cancer cells may respond similarly to cancer therapy, but due to their baseline variability in pre-existing immune responses, patients can have different responses to identical therapies. In this review we discuss how the immune environment of tumors develops, the critical immune cell populations in advanced cancers, and how immune interventions can manipulate the immune environment of patients with pre-malignancies or advanced cancers to improve therapeutic outcomes.
Collapse
Affiliation(s)
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| | | | | |
Collapse
|
9
|
Xu R, Du S, Zhu J, Meng F, Liu B. Neoantigen-targeted TCR-T cell therapy for solid tumors: How far from clinical application. Cancer Lett 2022; 546:215840. [DOI: 10.1016/j.canlet.2022.215840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/09/2022] [Accepted: 07/22/2022] [Indexed: 11/25/2022]
|
10
|
Zhang L, Xu L, Wang Y, Liu J, Tan G, Huang F, He N, Lu Z. A novel therapeutic vaccine based on graphene oxide nanocomposite for tumor immunotherapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.01.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
11
|
Faraj S, Kemp EH, Gawkrodger DJ. Patho-immunological mechanisms of vitiligo: the role of the innate and adaptive immunities and environmental stress factors. Clin Exp Immunol 2022; 207:27-43. [PMID: 35020865 PMCID: PMC8802175 DOI: 10.1093/cei/uxab002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022] Open
Abstract
Epidermal melanocyte loss in vitiligo, triggered by stresses ranging from trauma to emotional stress, chemical exposure or metabolite imbalance, to the unknown, can stimulate oxidative stress in pigment cells, which secrete damage-associated molecular patterns that then initiate innate immune responses. Antigen presentation to melanocytes leads to stimulation of autoreactive T-cell responses, with further targeting of pigment cells. Studies show a pathogenic basis for cellular stress, innate immune responses and adaptive immunity in vitiligo. Improved understanding of the aetiological mechanisms in vitiligo has already resulted in successful use of the Jak inhibitors in vitiligo. In this review, we outline the current understanding of the pathological mechanisms in vitiligo and locate loci to which therapeutic attack might be directed.
Collapse
Affiliation(s)
- Safa Faraj
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | | | - David John Gawkrodger
- Department of Infection, Immunology and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| |
Collapse
|
12
|
Mercatelli D, Formaggio F, Caprini M, Holding A, Giorgi F. Detection of subtype-specific breast cancer surface protein biomarkers via a novel transcriptomics approach. Biosci Rep 2021; 41:BSR20212218. [PMID: 34750607 PMCID: PMC8655506 DOI: 10.1042/bsr20212218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Cell-surface proteins have been widely used as diagnostic and prognostic markers in cancer research and as targets for the development of anticancer agents. So far, very few attempts have been made to characterize the surfaceome of patients with breast cancer, particularly in relation with the current molecular breast cancer (BRCA) classification. In this view, we developed a new computational method to infer cell-surface protein activities from transcriptomics data, termed 'SURFACER'. METHODS Gene expression data from GTEx were used to build a normal breast network model as input to infer differential cell-surface proteins activity in BRCA tissue samples retrieved from TCGA versus normal samples. Data were stratified according to the PAM50 transcriptional subtypes (Luminal A, Luminal B, HER2 and Basal), while unsupervised clustering techniques were applied to define BRCA subtypes according to cell-surface proteins activity. RESULTS Our approach led to the identification of 213 PAM50 subtypes-specific deregulated surface genes and the definition of five BRCA subtypes, whose prognostic value was assessed by survival analysis, identifying a cell-surface activity configuration at increased risk. The value of the SURFACER method in BRCA genotyping was tested by evaluating the performance of 11 different machine learning classification algorithms. CONCLUSIONS BRCA patients can be stratified into five surface activity-specific groups having the potential to identify subtype-specific actionable targets to design tailored targeted therapies or for diagnostic purposes. SURFACER-defined subtypes show also a prognostic value, identifying surface-activity profiles at higher risk.
Collapse
Affiliation(s)
- Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Francesco Formaggio
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Marco Caprini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Andrew Holding
- York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, U.K
| | - Federico M. Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
13
|
Izosimova AV, Yuzhakova DV, Skatova VD, Volchkova LN, Zagainova EV, Chudakov DM, Sharonov GV. Deciphering Repertoire of B16 Melanoma Reactive TCRs by Immunization, In Vitro Restimulation and Sequencing of IFNγ-Secreting T Cells. Int J Mol Sci 2021; 22:ijms22189859. [PMID: 34576023 PMCID: PMC8469664 DOI: 10.3390/ijms22189859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
Recent advances in cancer immunotherapy have great promise for the treatment of solid tumors. One of the key limiting factors that hamper the decoding of physiological responses to these therapies is the inability to distinguish between specific and nonspecific responses. The identification of tumor-specific lymphocytes is also the most challenging step in cancer cell therapies such as adoptive cell transfer and T cell receptor (TCR) cloning. Here, we have elaborated a protocol for the identification of tumor-specific T lymphocytes and the deciphering of their repertoires. B16 melanoma engraftment following anti-PD1 checkpoint therapy provides better antitumor immunity compared to repetitive immunization with heat-shocked tumor cells. We have also revealed that the most error-prone part of dendritic cell (DC) generation, i.e., their maturation step, can be omitted if DCs are cultured at a sufficiently high density. Using this optimized protocol, we have achieved a robust IFNγ response to B16F0 antigens, but only within CD4+ T helper cells. A comparison of the repertoires of IFNγ-positive and -negative cells shows a prominent enrichment of certain clones with putative tumor specificity among the IFNγ+ fraction. In summary, our optimized protocol and the data provided here will aid in the acquisition of broad statistical data and the creation of a meaningful database of B16-specific TCRs.
Collapse
Affiliation(s)
- Anna V. Izosimova
- Institute of Experimental Oncology and Biomedicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia; (A.V.I.); (D.V.Y.); (L.N.V.); (E.V.Z.); (D.M.C.)
- Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Diana V. Yuzhakova
- Institute of Experimental Oncology and Biomedicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia; (A.V.I.); (D.V.Y.); (L.N.V.); (E.V.Z.); (D.M.C.)
| | - Valeria D. Skatova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 16/10 Miklukho-Maklaya, 117997 Moscow, Russia;
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, 1 Ostrovityanova, 117997 Moscow, Russia
| | - Lilia N. Volchkova
- Institute of Experimental Oncology and Biomedicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia; (A.V.I.); (D.V.Y.); (L.N.V.); (E.V.Z.); (D.M.C.)
| | - Elena V. Zagainova
- Institute of Experimental Oncology and Biomedicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia; (A.V.I.); (D.V.Y.); (L.N.V.); (E.V.Z.); (D.M.C.)
- Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Dmitry M. Chudakov
- Institute of Experimental Oncology and Biomedicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia; (A.V.I.); (D.V.Y.); (L.N.V.); (E.V.Z.); (D.M.C.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 16/10 Miklukho-Maklaya, 117997 Moscow, Russia;
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, 1 Ostrovityanova, 117997 Moscow, Russia
| | - George V. Sharonov
- Institute of Experimental Oncology and Biomedicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia; (A.V.I.); (D.V.Y.); (L.N.V.); (E.V.Z.); (D.M.C.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 16/10 Miklukho-Maklaya, 117997 Moscow, Russia;
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, 1 Ostrovityanova, 117997 Moscow, Russia
- Correspondence:
| |
Collapse
|
14
|
Koşaloğlu-Yalçın Z, Blazeska N, Carter H, Nielsen M, Cohen E, Kufe D, Conejo-Garcia J, Robbins P, Schoenberger SP, Peters B, Sette A. The Cancer Epitope Database and Analysis Resource: A Blueprint for the Establishment of a New Bioinformatics Resource for Use by the Cancer Immunology Community. Front Immunol 2021; 12:735609. [PMID: 34504503 PMCID: PMC8421848 DOI: 10.3389/fimmu.2021.735609] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022] Open
Abstract
Recent years have witnessed a dramatic rise in interest towards cancer epitopes in general and particularly neoepitopes, antigens that are encoded by somatic mutations that arise as a consequence of tumorigenesis. There is also an interest in the specific T cell and B cell receptors recognizing these epitopes, as they have therapeutic applications. They can also aid in basic studies to infer the specificity of T cells or B cells characterized in bulk and single-cell sequencing data. The resurgence of interest in T cell and B cell epitopes emphasizes the need to catalog all cancer epitope-related data linked to the biological, immunological, and clinical contexts, and most importantly, making this information freely available to the scientific community in a user-friendly format. In parallel, there is also a need to develop resources for epitope prediction and analysis tools that provide researchers access to predictive strategies and provide objective evaluations of their performance. For example, such tools should enable researchers to identify epitopes that can be effectively used for immunotherapy or in defining biomarkers to predict the outcome of checkpoint blockade therapies. We present here a detailed vision, blueprint, and work plan for the development of a new resource, the Cancer Epitope Database and Analysis Resource (CEDAR). CEDAR will provide a freely accessible, comprehensive collection of cancer epitope and receptor data curated from the literature and provide easily accessible epitope and T cell/B cell target prediction and analysis tools. The curated cancer epitope data will provide a transparent benchmark dataset that can be used to assess how well prediction tools perform and to develop new prediction tools relevant to the cancer research community.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Computational Biology
- Databases, Genetic
- Epitopes, B-Lymphocyte
- Epitopes, T-Lymphocyte
- Humans
- Immunotherapy
- Mutation
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Tumor Microenvironment
Collapse
Affiliation(s)
- Zeynep Koşaloğlu-Yalçın
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Nina Blazeska
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Hannah Carter
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
- Moore’s Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Morten Nielsen
- Department of Bio and Health Informatics, Technical University of Denmark, Lyngby, Denmark
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, San Martín, Argentina
| | - Ezra Cohen
- Moore’s Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Donald Kufe
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Jose Conejo-Garcia
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Paul Robbins
- National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Stephen P. Schoenberger
- Laboratory of Cellular Immunology, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
15
|
Arnaud M, Bobisse S, Chiffelle J, Harari A. The Promise of Personalized TCR-Based Cellular Immunotherapy for Cancer Patients. Front Immunol 2021; 12:701636. [PMID: 34394096 PMCID: PMC8363295 DOI: 10.3389/fimmu.2021.701636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Mutation-derived neoantigens are now established as attractive targets for cancer immunotherapy. The field of adoptive T cell transfer (ACT) therapy was significantly reshaped by tumor neoantigens and is now moving towards the genetic engineering of T cells with neoantigen-specific T cell receptors (TCRs). Yet, the identification of neoantigen-reactive TCRs remains challenging and the process needs to be adapted to clinical timelines. In addition, the state of recipient T cells for TCR transduction is critical and can affect TCR-ACT efficacy. Here we provide an overview of the main strategies for TCR-engineering, describe the selection and expansion of optimal carrier cells for TCR-ACT and discuss the next-generation methods for rapid identification of relevant TCR candidates for gene transfer therapy.
Collapse
Affiliation(s)
- Marion Arnaud
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Sara Bobisse
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Johanna Chiffelle
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alexandre Harari
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
16
|
Zhang LX, Hu J, Jia YB, Liu RT, Cai T, Xu ZP. Two-dimensional layered double hydroxide nanoadjuvant: recent progress and future direction. NANOSCALE 2021; 13:7533-7549. [PMID: 33876812 DOI: 10.1039/d1nr00881a] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Layered double hydroxide (LDH) is a 'sandwich'-like two-dimensional clay material that has been systematically investigated for biomedical application in the past two decades. LDH is an alum-similar adjuvant, which has a well-defined layered crystal structure and exhibits high adjuvanticity. The unique structure of LDH includes positively charged layers composed of divalent and trivalent cations and anion-exchangeable interlayer galleries. Among the many variants of LDH, MgAl-LDH (the cationic ions are Mg2+ and Al3+) has the highest affinity to antigens, bioadjuvants and drug molecules, and exhibits superior biosafety. Past research studies indicate that MgAl-LDH can simultaneously load antigens, bioadjuvants and molecular drugs to amplify the strength of immune responses, and induce broad-spectrum immune responses. Moreover, the size and dispersity of MgAl-LDH in biological environments can be well controlled to actively deliver antigens to the immune system, realizing the rapid induction and maintenance of durable immune responses. Furthermore, the functionalization of MgAl-LDH nanoadjuvants enables it to capture antigens in situ and induce personalized immune responses, thereby more effectively overcoming complex diseases. In this review, we comprehensively summarize the development and application of MgAl-LDH nanoparticles as a vaccine adjuvant, demonstrating that MgAl-LDH is the most potential adjuvant for clinical application.
Collapse
Affiliation(s)
- Ling-Xiao Zhang
- Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo 315010, China. and Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315010, China and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia. and Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang Univeristy, Hangzhou 310058, China
| | - Jing Hu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying-Bo Jia
- University of Chinese Academy of Sciences, Beijing 100049, China and State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Rui-Tian Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Ting Cai
- Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo 315010, China. and Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315010, China
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
17
|
Wang P, Chen Y, Wang C. Beyond Tumor Mutation Burden: Tumor Neoantigen Burden as a Biomarker for Immunotherapy and Other Types of Therapy. Front Oncol 2021; 11:672677. [PMID: 33996601 PMCID: PMC8117238 DOI: 10.3389/fonc.2021.672677] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/07/2021] [Indexed: 02/05/2023] Open
Abstract
Immunotherapy has significantly improved the clinical outcome of patients with cancer. However, the immune response rate varies greatly, possibly due to lack of effective biomarkers that can be used to distinguish responders from non-responders. Recently, clinical studies have associated high tumor neoantigen burden (TNB) with improved outcomes in patients treated with immunotherapy. Therefore, TNB has emerged as a biomarker for immunotherapy and other types of therapy. In the present review, the potential application of TNB as a biomarker was evaluated. The methods of neoantigen prediction were summarized and the mechanisms involved in TNB were investigated. The impact of high TNB and increased number of infiltrating immune cells on the efficacy of immunotherapy was also addressed. Finally, the future challenges of TNB were discussed.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Biotherapy, Cancer Center, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yueyun Chen
- Department of Biotherapy, Cancer Center, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Chun Wang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Liu C, Seeram NP, Ma H. Small molecule inhibitors against PD-1/PD-L1 immune checkpoints and current methodologies for their development: a review. Cancer Cell Int 2021; 21:239. [PMID: 33906641 PMCID: PMC8077906 DOI: 10.1186/s12935-021-01946-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Programmed death-1/programmed death ligand-1 (PD-1/PD-L1) based immunotherapy is a revolutionary cancer therapy with great clinical success. The majority of clinically used PD-1/PD-L1 inhibitors are monoclonal antibodies but their applications are limited due to their poor oral bioavailability and immune-related adverse effects (irAEs). In contrast, several small molecule inhibitors against PD-1/PD-L1 immune checkpoints show promising blockage effects on PD-1/PD-L1 interactions without irAEs. However, proper analytical methods and bioassays are required to effectively screen small molecule derived PD-1/PD-L1 inhibitors. Herein, we summarize the biophysical and biochemical assays currently employed for the measurements of binding capacities, molecular interactions, and blocking effects of small molecule inhibitors on PD-1/PD-L1. In addition, the discovery of natural products based PD-1/PD-L1 antagonists utilizing these screening assays are reviewed. Potential pitfalls for obtaining false leading compounds as PD-1/PD-L1 inhibitors by using certain binding bioassays are also discussed in this review.
Collapse
Affiliation(s)
- Chang Liu
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA.
| | - Navindra P Seeram
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA
| | - Hang Ma
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA.
| |
Collapse
|
19
|
Du XM, Wei YP, Gao Y, Li Z, Zhang JM, Chang H, Li Y. Clinicopathological characteristics of primary peritoneal epithelioid mesothelioma of clear cell type: A case report. Medicine (Baltimore) 2021; 100:e25264. [PMID: 33761726 PMCID: PMC9281963 DOI: 10.1097/md.0000000000025264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/04/2021] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Primary peritoneal epithelioid mesothelioma of clear cell type is an extremely rare entity composed of clear cytoplasm. It is challenging to diagnose because of the morphological resemblance to clear cell tumor. PATIENTS CONCERNS A 69-year-old male patient had swollen lymph nodes in the right inguinal region for 7 months and was constipated for 1 month. DIAGNOSIS The patient was diagnosed as peritoneal epithelioid mesothelioma of clear cell type based on computed tomography scan, pathology, immunohistochemistry, special staining and whole-exome sequencing. This patient harbored VHL gene alteration in exon 1 and homologous recombination defect (with a score of 45). This finding indicated that this patient might be sensitive to platinum-based therapy and Poly ADP-ribose Polymerase (PARP) inhibitor. This patient carried no microsatellite instability, a low level of tumor mutation burden, and a high extent of intratumoral heterogeneity. Eighteen neoantigens were detected. INTERVENTIONS The patient received surgery-based multidisciplinary treatment by integrating cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC). HIPEC was administered with docetaxel 120 mg plus cisplatin 120 mg, at 43°C, for 60 minutes. After operation, the patient received intravenous (IV) chemotherapy with docetaxel 60 mg, pemetrexed 750 mg and cisplatin 100 mg, and then intraperitoneal (IP) chemotherapy with docetaxel 40 mg. The patient received interventional therapy of hepatic artery embolization for 5 times. OUTCOMES Regular follow-up was performed until Oct 14, 2020. The patient died 31.6 months later owing to incomplete intestinal obstruction. LESSONS Primary peritoneal epithelioid mesothelioma of clear cell type needs to be differentiated from a variety of clear cell tumors. This disease is characterized by specific genetic alteration. Whole-exome sequencing contributes to guide individualized therapy. CRS-HIPEC helps achieve long-term overall survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yan Li
- Department of Pathology
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
20
|
Li W, Jing Z, Wang S, Li Q, Xing Y, Shi H, Li S, Hong Z. P22 virus-like particles as an effective antigen delivery nanoplatform for cancer immunotherapy. Biomaterials 2021; 271:120726. [PMID: 33636548 DOI: 10.1016/j.biomaterials.2021.120726] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022]
Abstract
As a new strategy for cancer immunotherapy, therapeutic cancer vaccines have been greatly improved in recent years. However, addressing the needs to quickly and efficiently elicit a high-intensity immune response against neoantigen peptides, especially to induce an effective cytotoxic lymphocyte (CTL) reaction, remain challenges in this field. In this study, virus-like particles (VLPs) derived from the phage P22 were adopted to load peptide antigens on the surface, to test whether VLP technology can be used as a platform for efficient peptide antigen delivery by therapeutic cancer vaccines. The B and T epitopes (OVAB peptide and OVAT peptide) of ovalbumin (OVA) were used here as model antigens and fused individually at the C terminus of the coat protein (CP), which allowed display on the surface of P22 particles to form two types of vaccine particles (VLP-OVAB and VLP-OVAT). Subsequent experiments showed that VLP-OVAB induced an antibody titer against the peptide antigen as high as 5.0 × 105 and that VLP-OVAT induced highly effective cross-presentation and then strongly activated a T epitope-specific CTL response. Mouse tumor model experiments showed that VLP-OVAT could significantly inhibit tumor growth by increasing the proportions of CD4+ T cells, CD8+ T cells and effector memory T cells (TEM cells) and lowering the proportion of myeloid-derived suppressor cells (MDSCs) among tumor-infiltrating lymphocytes and splenocytes. Compared with other chemically synthesized nanomaterials, VLPs have obvious advantages as vaccine carriers due to their clear chemical composition, fixed spatial structure, excellent biocompatibility, and relatively high potential for clinical translation. Therefore, this platform may lay a solid foundation for the design and preparation of personalized therapeutic vaccines based on neoantigen peptides.
Collapse
Affiliation(s)
- Wenjing Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Zhe Jing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Shuqing Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Qiyu Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Yutong Xing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Haobo Shi
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Shuang Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
21
|
Abstract
Cancer is a major burden on the healthcare system, and new therapies are needed. Recently, the development of immunotherapies, which aim to boost or use the immune system, or its constituents, as a tool to fight malignant cells, has provided a major new tool in the arsenal of clinicians and has revolutionized the treatment of many cancers.Cellular immunotherapies are based on the administration of living cells to patients and have developed hugely, especially since 2010 when Sipuleucel-T (Provenge), a DC vaccine, was the first cellular immunotherapy to be approved by the FDA. The ensuing years have seen two further cellular immunotherapies gain FDA approval: tisagenlecleucel (Kymriah) and axicabtagene ciloleucel (Yescarta).This review will give an overview of the principles of immunotherapies before focusing on the major forms of cellular immunotherapies individually, T cell-based, natural killer (NK) cell-based and dendritic cell (DC)-based, as well as detailing some of the clinical trials relevant to each therapy.
Collapse
Affiliation(s)
- Conall Hayes
- School of Medicine, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
22
|
Xu J, Wang B, Li S. Gene and cell therapy. REMINGTON 2021:463-488. [DOI: 10.1016/b978-0-12-820007-0.00025-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
23
|
Galgani M, Bruzzaniti S, La Rocca C, Micillo T, de Candia P, Bifulco M, Matarese G. Immunometabolism of regulatory T cells in cancer. Mol Aspects Med 2020; 77:100936. [PMID: 33250195 DOI: 10.1016/j.mam.2020.100936] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023]
Abstract
Regulatory T (Treg) cells are known to orchestrate the regulatory mechanisms aimed at suppressing pathological auto-reactive immune responses and are thus key in ensuring the maintenance of immune homeostasis. On the other hand, the presence of Treg cells with enhanced suppressive capability in a plethora of human cancers represents a major obstacle to an effective anti-cancer immune response. A relevant research effort has thus been dedicated to comprehend Treg cell biology, leading to a continuously refining characterization of their phenotype and function and unveiling the central role of metabolism in ensuring Treg cell fitness in cancer. Here we focus on how the peculiar biochemical characteristics of the tumor microenvironment actually support Treg cell metabolic activation and favor their selective survival and proliferation. Moreover, we examine the key metabolic pathways that may become useful targets of novel treatments directed at hampering tumor resident Treg cell proficiency, thus representing the next research frontier in cancer immunotherapy.
Collapse
Affiliation(s)
- Mario Galgani
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy; Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131, Napoli, Italy
| | - Sara Bruzzaniti
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131, Napoli, Italy; Dipartimento di Biologia, Università Degli Studi di Napoli "Federico II", 80126, Napoli, Italy
| | - Claudia La Rocca
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131, Napoli, Italy
| | - Teresa Micillo
- Unità di Neuroimmunologia, Fondazione Santa Lucia IRCCS, 00179, Roma, Italy
| | | | - Maurizio Bifulco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy; Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131, Napoli, Italy.
| |
Collapse
|
24
|
Rubio AJ, Porter T, Zhong X. Duality of B Cell-CXCL13 Axis in Tumor Immunology. Front Immunol 2020; 11:521110. [PMID: 33193299 PMCID: PMC7609404 DOI: 10.3389/fimmu.2020.521110] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor immunity is a rapidly evolving area of research consisting of many possible permutations of immune cell tumor interactions that are dependent upon cell type, tumor type, and stage in tumor progression. At the same time, the majority of cancer immunotherapies have been focused on modulating the T cell-mediated antitumor immune response and have largely ignored the potential utility that B cells possess with respect to tumor immunity. Therefore, this motivated an exploration into the role that B cells and their accompanying chemokine, CXCL13, play in tumor immunity across multiple tumor types. Both B cells and CXCL13 possess dualistic impacts on tumor progression and tumor immunity which is furthered detail in this review. Specifically, various B cells subtypes are able to suppress or enhance several important immunological functions. Paradoxically, CXCL13 has been shown to drive several pro-growth and invasive signaling pathways across multiple tumor types, while also, correlating with improved survival and immune cell tumor localization in other tumor types. Potential tools for better elucidating the mechanisms by which B cells and CXCL13 impact the antitumor immune response are also discussed. In addition, multiples strategies are proposed for modulating the B cell-CXCL13 axis for cancer immunotherapies.
Collapse
Affiliation(s)
- Angel J. Rubio
- Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, United States
| | - Tyrone Porter
- Department of Biomedical Engineering, University of Texas Austin, Austin, TX, United States
| | - Xuemei Zhong
- Hematology and Medical Oncology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
25
|
Wells DK, van Buuren MM, Dang KK, Hubbard-Lucey VM, Sheehan KCF, Campbell KM, Lamb A, Ward JP, Sidney J, Blazquez AB, Rech AJ, Zaretsky JM, Comin-Anduix B, Ng AHC, Chour W, Yu TV, Rizvi H, Chen JM, Manning P, Steiner GM, Doan XC, Merghoub T, Guinney J, Kolom A, Selinsky C, Ribas A, Hellmann MD, Hacohen N, Sette A, Heath JR, Bhardwaj N, Ramsdell F, Schreiber RD, Schumacher TN, Kvistborg P, Defranoux NA. Key Parameters of Tumor Epitope Immunogenicity Revealed Through a Consortium Approach Improve Neoantigen Prediction. Cell 2020; 183:818-834.e13. [PMID: 33038342 DOI: 10.1016/j.cell.2020.09.015] [Citation(s) in RCA: 314] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/08/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022]
Abstract
Many approaches to identify therapeutically relevant neoantigens couple tumor sequencing with bioinformatic algorithms and inferred rules of tumor epitope immunogenicity. However, there are no reference data to compare these approaches, and the parameters governing tumor epitope immunogenicity remain unclear. Here, we assembled a global consortium wherein each participant predicted immunogenic epitopes from shared tumor sequencing data. 608 epitopes were subsequently assessed for T cell binding in patient-matched samples. By integrating peptide features associated with presentation and recognition, we developed a model of tumor epitope immunogenicity that filtered out 98% of non-immunogenic peptides with a precision above 0.70. Pipelines prioritizing model features had superior performance, and pipeline alterations leveraging them improved prediction performance. These findings were validated in an independent cohort of 310 epitopes prioritized from tumor sequencing data and assessed for T cell binding. This data resource enables identification of parameters underlying effective anti-tumor immunity and is available to the research community.
Collapse
Affiliation(s)
- Daniel K Wells
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| | - Marit M van Buuren
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands; T Cell Immunology, Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech US, Cambridge, MA, USA
| | - Kristen K Dang
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA
| | | | - Kathleen C F Sheehan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Katie M Campbell
- Division of Hematology and Oncology, Department of Medicine, Johnson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Andrew Lamb
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA
| | - Jeffrey P Ward
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Ana B Blazquez
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew J Rech
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jesse M Zaretsky
- Division of Hematology and Oncology, Department of Medicine, Johnson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Begonya Comin-Anduix
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Department of Surgery, David Geffen School of Medicine, Johnson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - William Chour
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Thomas V Yu
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA
| | - Hira Rizvi
- Druckenmiller Center for Lung Cancer Research, MSKCC, New York, NY, USA
| | - Jia M Chen
- Division of Hematology and Oncology, Department of Medicine, Johnson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Patrice Manning
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Xengie C Doan
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA
| | - Taha Merghoub
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Department of Medicine, MSKCC, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Justin Guinney
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA; Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| | - Adam Kolom
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Anna-Maria Kellen Clinical Accelerator, Cancer Research Institute, New York, NY, USA
| | - Cheryl Selinsky
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Antoni Ribas
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Division of Hematology and Oncology, Department of Medicine, Johnson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Matthew D Hellmann
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Druckenmiller Center for Lung Cancer Research, MSKCC, New York, NY, USA; Department of Medicine, MSKCC, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Alessandro Sette
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - James R Heath
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Institute for Systems Biology, Seattle, WA, USA
| | - Nina Bhardwaj
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fred Ramsdell
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Robert D Schreiber
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Ton N Schumacher
- Division of Molecular Oncology and Immunology, Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | |
Collapse
|
26
|
Gaissmaier L, Elshiaty M, Christopoulos P. Breaking Bottlenecks for the TCR Therapy of Cancer. Cells 2020; 9:E2095. [PMID: 32937956 PMCID: PMC7564186 DOI: 10.3390/cells9092095] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/03/2020] [Accepted: 09/11/2020] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors have redefined the treatment of cancer, but their efficacy depends critically on the presence of sufficient tumor-specific lymphocytes, and cellular immunotherapies develop rapidly to fill this gap. The paucity of suitable extracellular and tumor-associated antigens in solid cancers necessitates the use of neoantigen-directed T-cell-receptor (TCR)-engineered cells, while prevention of tumor evasion requires combined targeting of multiple neoepitopes. These can be currently identified within 2 weeks by combining cutting-edge next-generation sequencing with bioinformatic pipelines and used to select tumor-reactive TCRs in a high-throughput manner for expeditious scalable non-viral gene editing of autologous or allogeneic lymphocytes. "Young" cells with a naive, memory stem or central memory phenotype can be additionally armored with "next-generation" features against exhaustion and the immunosuppressive tumor microenvironment, where they wander after reinfusion to attack heavily pretreated and hitherto hopeless neoplasms. Facilitated by major technological breakthroughs in critical manufacturing steps, based on a solid preclinical rationale, and backed by rapidly accumulating evidence, TCR therapies break one bottleneck after the other and hold the promise to become the next immuno-oncological revolution.
Collapse
Affiliation(s)
- Lena Gaissmaier
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany; (L.G.); (M.E.)
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Mariam Elshiaty
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany; (L.G.); (M.E.)
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany; (L.G.); (M.E.)
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| |
Collapse
|
27
|
Liu W, Zhang L, Xiu Z, Guo J, Wang L, Zhou Y, Jiao Y, Sun M, Cai J. Combination of Immune Checkpoint Inhibitors with Chemotherapy in Lung Cancer. Onco Targets Ther 2020; 13:7229-7241. [PMID: 32801752 PMCID: PMC7394580 DOI: 10.2147/ott.s255491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
Tremendous progress has been achieved in the field of immune checkpoint inhibitors (ICIs) therapy in lung cancer in recent years. To generate robust, long-lasting anti-tumor immune responses in lung cancer patients, combinational ICI therapies have been explored deeply. Conventionally, chemotherapy was considered as immunosuppressive. It is now recognized that chemotherapy could also reinstate cancer cell immune-surveillance and enable the perception of cancer cells as dangerous. That is to say that chemotherapeutic drugs are not only a source of direct cytotoxic effects but also an adjuvant for anti-tumor immunity. Recently, multiple clinical studies of ICIs combined with chemotherapeutic drugs have been explored and proved effective. However, there are still crucial questions that are not well addressed, such as the optimal dose and schedule for a given combination may differ across disease indications, and the appropriate strategy of selecting patient population that can benefit from ICIs remains unclear. To facilitate more rational lung cancer ICIs therapy development, this review summarizes the immune-regulatory effects and related mechanisms of chemotherapeutic drugs and the clinical progress of ICIs and their combination with chemotherapies in lung cancer treatment.
Collapse
Affiliation(s)
- Wei Liu
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China.,Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Lei Zhang
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China.,Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Zhiming Xiu
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Jian Guo
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Liye Wang
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX 77030, USA
| | - Yue Zhou
- Department of Statistics, North Dakota State University, Fargo, ND 58102, USA
| | - Yang Jiao
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Meiyan Sun
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Jianhui Cai
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China.,Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin 132013, People's Republic of China
| |
Collapse
|
28
|
Zhong XF, Sun X. Nanomedicines based on nanoscale metal-organic frameworks for cancer immunotherapy. Acta Pharmacol Sin 2020; 41:928-935. [PMID: 32355277 PMCID: PMC7468577 DOI: 10.1038/s41401-020-0414-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy, with an aim to enhance host immune responses, has been recognized as a promising therapeutic treatment for cancer. A diversity of immunomodulatory agents, including tumor-associated antigens, adjuvants, cytokines and immunomodulators, has been explored for their ability to induce a cascading adaptive immune response. Nanoscale metal-organic frameworks (nMOFs), a class of crystalline-shaped nanomaterials formed by the self-assembly of organic ligands and metal nodes, are attractive for cancer immunotherapy because they feature tunable pore size, high surface area and loading capacity, and intrinsic biodegradability. In this review we summarize recent progress in the development of nMOFs for cancer immunotherapy, including cancer vaccine delivery and combination of in situ vaccination with immunomodulators to reverse immune suppression. Current challenges and future perspectives for rational design of nMOF-based cancer immunotherapy are also discussed.
Collapse
Affiliation(s)
- Xiao-Fang Zhong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
29
|
Ishihara J, Ishihara A, Sasaki K, Lee SSY, Williford JM, Yasui M, Abe H, Potin L, Hosseinchi P, Fukunaga K, Raczy MM, Gray LT, Mansurov A, Katsumata K, Fukayama M, Kron SJ, Swartz MA, Hubbell JA. Targeted antibody and cytokine cancer immunotherapies through collagen affinity. Sci Transl Med 2020; 11:11/487/eaau3259. [PMID: 30971453 DOI: 10.1126/scitranslmed.aau3259] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 11/27/2018] [Accepted: 02/22/2019] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy with immune checkpoint inhibitors (CPIs) and interleukin-2 (IL-2) has demonstrated clinical efficacy but is frequently accompanied with severe adverse events caused by excessive and systemic immune system activation. Here, we addressed this need by targeting both the CPI antibodies anti-cytotoxic T lymphocyte antigen 4 antibody (αCTLA4) + anti-programmed death ligand 1 antibody (αPD-L1) and the cytokine IL-2 to tumors via conjugation (for the antibodies) or recombinant fusion (for the cytokine) to a collagen-binding domain (CBD) derived from the blood protein von Willebrand factor (VWF) A3 domain, harnessing the exposure of tumor stroma collagen to blood components due to the leakiness of the tumor vasculature. We show that intravenously administered CBD protein accumulated mainly in tumors. CBD conjugation or fusion decreases the systemic toxicity of both αCTLA4 + αPD-L1 combination therapy and IL-2, for example, eliminating hepatotoxicity with the CPI molecules and ameliorating pulmonary edema with IL-2. Both CBD-CPI and CBD-IL-2 suppressed tumor growth compared to their unmodified forms in multiple murine cancer models, and both CBD-CPI and CBD-IL-2 increased tumor-infiltrating CD8+ T cells. In an orthotopic breast cancer model, combination treatment with CPI and IL-2 eradicated tumors in 9 of 13 animals with the CBD-modified drugs, whereas it did so in only 1 of 13 animals with the unmodified drugs. Thus, the A3 domain of VWF can be used to improve safety and efficacy of systemically administered tumor drugs with high translational promise.
Collapse
Affiliation(s)
- Jun Ishihara
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ako Ishihara
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Koichi Sasaki
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Steve Seung-Young Lee
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | | | - Mariko Yasui
- Department of Pathology, University of Tokyo, 113-8655 Tokyo, Japan
| | - Hiroyuki Abe
- Department of Pathology, University of Tokyo, 113-8655 Tokyo, Japan
| | - Lambert Potin
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.,Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Peyman Hosseinchi
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Kazuto Fukunaga
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Michal M Raczy
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Laura T Gray
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Aslan Mansurov
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Kiyomitsu Katsumata
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Masashi Fukayama
- Department of Pathology, University of Tokyo, 113-8655 Tokyo, Japan
| | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Melody A Swartz
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.,Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Jeffrey A Hubbell
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
30
|
Abstract
Vitiligo is an autoimmune disease of the skin that targets pigment-producing melanocytes and results in patches of depigmentation that are visible as white spots. Recent research studies have yielded a strong mechanistic understanding of this disease. Autoreactive cytotoxic CD8+ T cells engage melanocytes and promote disease progression through the local production of IFN-γ, and IFN-γ-induced chemokines are then secreted from surrounding keratinocytes to further recruit T cells to the skin through a positive-feedback loop. Both topical and systemic treatments that block IFN-γ signaling can effectively reverse vitiligo in humans; however, disease relapse is common after stopping treatments. Autoreactive resident memory T cells are responsible for relapse, and new treatment strategies focus on eliminating these cells to promote long-lasting benefit. Here, we discuss basic, translational, and clinical research studies that provide insight into the pathogenesis of vitiligo, and how this insight has been utilized to create new targeted treatment strategies.
Collapse
Affiliation(s)
- Michael L. Frisoli
- University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA;, ,
| | - Kingsley Essien
- University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA;, ,
| | - John E. Harris
- University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA;, ,
| |
Collapse
|
31
|
Gridelli C, Ciuleanu T, Domine M, Szczesna A, Bover I, Cobo M, Kentepozidis N, Zarogoulidis K, Kalofonos C, Kazarnowisz A, Korozan M, de Las Penas R, Majem M, Chella A, Griesinger F, Bournakis E, Sadjadian P, Kotsakis A, Chinet T, Syrigos KN, Correale P, Gallou C, Jamet JM, Vetsika EK, Kosmatopoulos K, Georgoulias V. Clinical activity of a htert (vx-001) cancer vaccine as post-chemotherapy maintenance immunotherapy in patients with stage IV non-small cell lung cancer: final results of a randomised phase 2 clinical trial. Br J Cancer 2020; 122:1461-1466. [PMID: 32210365 PMCID: PMC7217860 DOI: 10.1038/s41416-020-0785-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 02/13/2020] [Accepted: 02/24/2020] [Indexed: 12/30/2022] Open
Abstract
Background The cancer vaccine Vx-001, which targets the universal tumour antigen TElomerase Reverse Transcriptase (TERT), can mount specific Vx-001/TERT572 CD8 + cytotoxic T cells; this immune response is associated with improved overall survival (OS) in patients with advanced/metastatic non-small cell lung cancer (NSCLC). Methods A randomised, double blind, phase 2b trial, in HLA-A*201-positive patients with metastatic, TERT-expressing NSCLC, who did not progress after first-line platinum-based chemotherapy were randomised to receive either Vx-001 or placebo. The primary endpoint of the trial was OS. Results Two hundred and twenty-one patients were randomised and 190 (101 and 89 patients in the placebo and the Vx-001 arm, respectively) were analysed for efficacy. There was not treatment-related toxicity >grade 2. The study did not meet its primary endpoint (median OS 11.3 and 14.3 months for the placebo and the Vx-001, respectively; p = 0.86) whereas the median Time to Treatment Failure (TTF) was 3.5 and 3.6 months, respectively. Disease control for >6months was observed in 30 (33.7%) and 26 (25.7%) patients treated with Vx-001 and placebo, respectively. There was no documented objective CR or PR. Long lasting TERT-specific immune response was observed in 29.2% of vaccinated patients who experienced a significantly longer OS compared to non-responders (21.3 and 13.4 months, respectively; p = 0.004). Conclusion Vx-001 could induce specific CD8+ immune response but failed to meet its primary endpoint. Subsequent studies have to be focused on the identification and treatment of subgroups of patients able to mount an effective immunological response to Vx-001. Clinical trial registration NCT01935154
Collapse
Affiliation(s)
| | | | | | | | | | - Manuel Cobo
- Hospital Regional Universitario Málaga, Instituto de Investigaciones Biomédicas (IBIMA), Málaga, Spain
| | | | | | | | | | | | | | | | | | | | | | | | - Athanasios Kotsakis
- Dpt of Medical Oncology, University General Hospital of Larissa, Larissa, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Yu H, Ye C, Li J, Pan C, Lin W, Chen H, Zhou Z, Ye Y. An altered HLA-A0201-restricted MUC1 epitope that could induce more efficient anti-tumor effects against gastric cancer. Exp Cell Res 2020; 390:111953. [PMID: 32156601 DOI: 10.1016/j.yexcr.2020.111953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/30/2022]
Abstract
MUC1 is a tumor-associated antigen (TAA) overexpressed in many tumor types, which makes it an attractive target for cancer immunotherapy. However, this marker is a non-mutated antigen without high immunogenicity. In this study, we designed several new altered peptides by replacing amino acids in their sequences, which were derived from a low-affinity MUC1 peptide, thus bypassing immune tolerance. Compared to the wild-type (WT) peptide, the altered MUC1 peptides (MUC11081-1089L2, MUC11156-1164L2, MUC11068-1076Y1) showed higher affinity to the HLA-A0201 molecule and stronger immunogenicity. Furthermore, these altered peptides resulted in the generation of more cytotoxic T lymphocytes (CTLs) that could cross-recognize gastric cancer cells expressing WT MUC1 peptides, in an HLA-A0201-restricted manner. In addition, M1.1 (MUC1950-958), a promising antitumor peptide that has been tested in multiple tumors, was not able to induce stronger antitumor responses. Collectively, our results demonstrated that altered peptides from MUC1, as potential HLA-A0201-restricted CTL epitopes, could serve as peptide vaccines or constitute components of peptide-loaded dendritic cell vaccines for gastric cancer treatment.
Collapse
Affiliation(s)
- Huahui Yu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Chunmei Ye
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Jieyu Li
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, Fujian Province, China
| | - Chunli Pan
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Wansong Lin
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, Fujian Province, China
| | - Huijing Chen
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, Fujian Province, China
| | - Zhifeng Zhou
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, Fujian Province, China
| | - Yunbin Ye
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China; Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, Fujian Province, China.
| |
Collapse
|
33
|
Blanc E, Holtgrewe M, Dhamodaran A, Messerschmidt C, Willimsky G, Blankenstein T, Beule D. Identification and ranking of recurrent neo-epitopes in cancer. BMC Med Genomics 2019; 12:171. [PMID: 31775766 PMCID: PMC6882202 DOI: 10.1186/s12920-019-0611-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/25/2019] [Indexed: 12/25/2022] Open
Abstract
Background Immune escape is one of the hallmarks of cancer and several new treatment approaches attempt to modulate and restore the immune system’s capability to target cancer cells. At the heart of the immune recognition process lies antigen presentation from somatic mutations. These neo-epitopes are emerging as attractive targets for cancer immunotherapy and new strategies for rapid identification of relevant candidates have become a priority. Methods We carefully screen TCGA data sets for recurrent somatic amino acid exchanges and apply MHC class I binding predictions. Results We propose a method for in silico selection and prioritization of candidates which have a high potential for neo-antigen generation and are likely to appear in multiple patients. While the percentage of patients carrying a specific neo-epitope and HLA-type combination is relatively small, the sheer number of new patients leads to surprisingly high reoccurence numbers. We identify 769 epitopes which are expected to occur in 77629 patients per year. Conclusion While our candidate list will definitely contain false positives, the results provide an objective order for wet-lab testing of reusable neo-epitopes. Thus recurrent neo-epitopes may be suitable to supplement existing personalized T cell treatment approaches with precision treatment options.
Collapse
Affiliation(s)
- Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany
| | - Manuel Holtgrewe
- Core Unit Bioinformatics, Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany
| | - Arunraj Dhamodaran
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, Berlin, 13092, Germany
| | - Clemens Messerschmidt
- Core Unit Bioinformatics, Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Lindenberger Weg 80, Berlin, 13125, Germany.,Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany.,German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, 69120, Germany
| | - Thomas Blankenstein
- Institute of Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Lindenberger Weg 80, Berlin, 13125, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, Berlin, 13092, Germany.,Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany. .,Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, Berlin, 13092, Germany.
| |
Collapse
|
34
|
Scheetz L, Park KS, Li Q, Lowenstein PR, Castro MG, Schwendeman A, Moon JJ. Engineering patient-specific cancer immunotherapies. Nat Biomed Eng 2019; 3:768-782. [PMID: 31406259 PMCID: PMC6783331 DOI: 10.1038/s41551-019-0436-x] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
Research into the immunological processes implicated in cancer has yielded a basis for the range of immunotherapies that are now considered the fourth pillar of cancer treatment (alongside surgery, radiotherapy and chemotherapy). For some aggressive cancers, such as advanced non-small-cell lung carcinoma, combination immunotherapies have resulted in unprecedented treatment efficacy for responding patients, and have become frontline therapies. Individualized immunotherapy, enabled by the identification of patient-specific mutations, neoantigens and biomarkers, and facilitated by advances in genomics and proteomics, promises to broaden the responder patient population. In this Perspective, we give an overview of immunotherapies leveraging engineering approaches, including the design of biomaterials, delivery strategies and nanotechnology solutions, for the realization of individualized cancer treatments such as nanoparticle vaccines customized with neoantigens, cell therapies based on patient-derived dendritic cells and T cells, and combinations of theranostic strategies. Developments in precision cancer immunotherapy will increasingly rely on the adoption of engineering principles.
Collapse
Affiliation(s)
- Lindsay Scheetz
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Kyung Soo Park
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Qiao Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
35
|
Gerber HP, Sibener LV, Lee LJ, Gee M. Intracellular targets as source for cleaner targets for the treatment of solid tumors. Biochem Pharmacol 2019; 168:275-284. [DOI: 10.1016/j.bcp.2019.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/12/2019] [Indexed: 01/02/2023]
|
36
|
Frankiw L, Baltimore D, Li G. Alternative mRNA splicing in cancer immunotherapy. Nat Rev Immunol 2019; 19:675-687. [PMID: 31363190 DOI: 10.1038/s41577-019-0195-7] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
Immunotherapies are yielding effective treatments for several previously untreatable cancers. Still, the identification of suitable antigens specific to the tumour that can be targets for cancer vaccines and T cell therapies is a challenge. Alternative processing of mRNA, a phenomenon that has been shown to alter the proteomic diversity of many cancers, may offer the potential of a broadened target space. Here, we discuss the promise of analysing mRNA processing events in cancer cells, with an emphasis on mRNA splicing, for the identification of potential new targets for cancer immunotherapy. Further, we highlight the challenges that must be overcome for this new avenue to have clinical applicability.
Collapse
Affiliation(s)
- Luke Frankiw
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - Guideng Li
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. .,Suzhou Institute of Systems Medicine, Suzhou, China.
| |
Collapse
|
37
|
Immunoepigenetics Combination Therapies: An Overview of the Role of HDACs in Cancer Immunotherapy. Int J Mol Sci 2019; 20:ijms20092241. [PMID: 31067680 PMCID: PMC6539010 DOI: 10.3390/ijms20092241] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/23/2019] [Accepted: 04/28/2019] [Indexed: 12/26/2022] Open
Abstract
Long-standing efforts to identify the multifaceted roles of histone deacetylase inhibitors (HDACis) have positioned these agents as promising drug candidates in combatting cancer, autoimmune, neurodegenerative, and infectious diseases. The same has also encouraged the evaluation of multiple HDACi candidates in preclinical studies in cancer and other diseases as well as the FDA-approval towards clinical use for specific agents. In this review, we have discussed how the efficacy of immunotherapy can be leveraged by combining it with HDACis. We have also included a brief overview of the classification of HDACis as well as their various roles in physiological and pathophysiological scenarios to target key cellular processes promoting the initiation, establishment, and progression of cancer. Given the critical role of the tumor microenvironment (TME) towards the outcome of anticancer therapies, we have also discussed the effect of HDACis on different components of the TME. We then have gradually progressed into examples of specific pan-HDACis, class I HDACi, and selective HDACis that either have been incorporated into clinical trials or show promising preclinical effects for future consideration. Finally, we have included examples of ongoing trials for each of the above categories of HDACis as standalone agents or in combination with immunotherapeutic approaches.
Collapse
|
38
|
The Cellular Immunotherapy Revolution: Arming the Immune System for Precision Therapy. Trends Immunol 2019; 40:292-309. [DOI: 10.1016/j.it.2019.02.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/30/2022]
|
39
|
Reuven EM, Leviatan Ben-Arye S, Yu H, Duchi R, Perota A, Conchon S, Bachar Abramovitch S, Soulillou JP, Galli C, Chen X, Padler-Karavani V. Biomimetic Glyconanoparticle Vaccine for Cancer Immunotherapy. ACS NANO 2019; 13:2936-2947. [PMID: 30840433 PMCID: PMC6756924 DOI: 10.1021/acsnano.8b07241] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Cancer immunotherapy aims to harness the immune system to combat malignant processes. Transformed cells harbor diverse modifications that lead to formation of neoantigens, including aberrantly expressed cell surface carbohydrates. Targeting tumor-associated carbohydrate antigens (TACA) hold great potential for cancer immunotherapy. N-glycolylneuraminic acid (Neu5Gc) is a dietary non-human immunogenic carbohydrate that accumulates on human cancer cells, thereby generating neoantigens. In mice, passive immunotherapy with anti-Neu5Gc antibodies inhibits growth of Neu5Gc-positive tumors. Here, we designed an active cancer vaccine immunotherapy strategy to target Neu5Gc-positive tumors. We generated biomimetic glyconanoparticles using engineered αGal knockout porcine red blood cells to form nanoghosts (NGs) that either express (NGpos) or lack expression (NGneg) of Neu5Gc-glycoconjugates in their natural context. We demonstrated that optimized immunization of "human-like" Neu5Gc-deficient Cmah-/- mice with NGpos glyconanoparticles induce a strong, diverse and persistent anti-Neu5Gc IgG immune response. The resulting anti-Neu5Gc IgG antibodies were also detected within Neu5Gc-positive tumors and inhibited tumor growth in vivo. Using detailed glycan microarray analysis, we further demonstrate that the kinetics and quality of the immune responses influence the efficacy of the vaccine. These findings reinforce the potential of TACA neoantigens and the dietary non-human sialic acid Neu5Gc, in particular, as immunotherapy targets.
Collapse
Affiliation(s)
- Eliran Moshe Reuven
- Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, 69978, Israel
| | | | - Hai Yu
- Department of Chemistry, University of California-Davis, Davis, CA 95616, USA
| | - Roberto Duchi
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
| | - Andrea Perota
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
| | - Sophie Conchon
- Institut de Transplantation–Urologie–Néphrologie, INSERM Unité Mixte de Recherche 1064, Centre Hospitalo Universitaire de Nantes, Nantes 44000, France
| | | | - Jean-Paul Soulillou
- Institut de Transplantation–Urologie–Néphrologie, INSERM Unité Mixte de Recherche 1064, Centre Hospitalo Universitaire de Nantes, Nantes 44000, France
| | - Cesare Galli
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
- FondazioneAvantea Cremona, Italy
| | - Xi Chen
- Department of Chemistry, University of California-Davis, Davis, CA 95616, USA
| | - Vered Padler-Karavani
- Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, 69978, Israel
- Corresponding Author: Department of Cell Research & Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978 Israel. Tel: +972-3-640-6737. Fax: +972-3-642-2046.
| |
Collapse
|
40
|
Cui M, Liao Q, Zhao Y. Enteral Immunonutrition Promotes Immune and Inflammatory Recovery after Surgery for Gastric Cancer. J INVEST SURG 2019; 33:960-961. [PMID: 30897981 DOI: 10.1080/08941939.2019.1583295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Ming Cui
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| |
Collapse
|
41
|
Andersson AMC, Buldun CM, Pattinson DJ, Draper SJ, Howarth M. SnoopLigase peptide-peptide conjugation enables modular vaccine assembly. Sci Rep 2019; 9:4625. [PMID: 30874593 PMCID: PMC6420506 DOI: 10.1038/s41598-019-40985-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 02/11/2019] [Indexed: 02/06/2023] Open
Abstract
For many infectious diseases there is still no vaccine, even though potential protective antigens have been identified. Suitable platforms and conjugation routes are urgently needed to convert the promise of such antigens into broadly protective and scalable vaccines. Here we apply a newly established peptide-peptide ligation approach, SnoopLigase, for specific and irreversible coupling of antigens onto an oligomerization platform. SnoopLigase was engineered from a Streptococcus pneumoniae adhesin and enables isopeptide bond formation between two peptide tags: DogTag and SnoopTagJr. We expressed in bacteria DogTag linked to the self-assembling coiled-coil nanoparticle IMX313. This platform was stable over months at 37 °C when lyophilized, remaining reactive even after boiling. IMX-DogTag was efficiently coupled to two blood-stage malarial proteins (from PfEMP1 or CyRPA), with SnoopTagJr fused at the N- or C-terminus. We also showed SnoopLigase-mediated coupling of a telomerase peptide relevant to cancer immunotherapy. SnoopLigase-mediated nanoassembly enhanced the antibody response to both malaria antigens in a prime-boost model. Including or depleting SnoopLigase from the conjugate had little effect on the antibody response to the malarial antigens. SnoopLigase decoration represents a promising and accessible strategy for modular plug-and-display vaccine assembly, as well as providing opportunities for robust nanoconstruction in synthetic biology.
Collapse
Affiliation(s)
| | - Can M Buldun
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | | | - Simon J Draper
- Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Mark Howarth
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
42
|
Snook JP, Kim C, Williams MA. TCR signal strength controls the differentiation of CD4 + effector and memory T cells. Sci Immunol 2019; 3:3/25/eaas9103. [PMID: 30030369 DOI: 10.1126/sciimmunol.aas9103] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/21/2018] [Indexed: 12/21/2022]
Abstract
CD4+ T cell responses are composed of heterogeneous T cell receptor (TCR) signals that influence the acquisition of effector and memory characteristics. We sought to define early TCR-dependent activation events that control T cell differentiation. A polyclonal panel of TCRs specific for the same viral antigen demonstrated substantial variability in TCR signal strength, expression of CD25, and activation of nuclear factor of activated T cells and nuclear factor κB. After viral infection, strong TCR signals corresponded to T helper cell (TH1) differentiation, whereas T follicular helper cell and memory T cell differentiation were most efficient when TCR signals were comparatively lower. We observed substantial heterogeneity in TCR-dependent CD25 expression in vivo, and the vast majority of CD4+ memory T cells were derived from CD25lo effector cells that displayed decreased TCR signaling in vivo. Nevertheless, memory T cells derived from either CD25lo or CD25hi effector cells responded vigorously to rechallenge, indicating that, although early clonal differences in CD25 expression predicted memory T cell numbers, they did not predict memory T cell function on a per cell basis. Gene transcription analysis demonstrated expression clustering based on CD25 expression and enrichment of transcripts associated with enhanced T follicular helper cell and memory development within CD25lo effector cells. Direct enhancement of TCR signaling via knockdown of Src homology region 2 domain-containing phosphatase 1, a tyrosine phosphatase that suppresses early TCR signaling events, favored the differentiation of TH1 effector and memory cells. We conclude that strong TCR signals during early T cell activation favor terminal TH1 differentiation over long-term TH1 and T follicular helper cell memory responses.
Collapse
Affiliation(s)
- Jeremy P Snook
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Chulwoo Kim
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew A Williams
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
43
|
Fink K. Can We Improve Vaccine Efficacy by Targeting T and B Cell Repertoire Convergence? Front Immunol 2019; 10:110. [PMID: 30814993 PMCID: PMC6381292 DOI: 10.3389/fimmu.2019.00110] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 01/15/2019] [Indexed: 01/31/2023] Open
Abstract
Traditional vaccine development builds on the assumption that healthy individuals have virtually unlimited antigen recognition repertoires of receptors in B cells and T cells [the B cell receptor (BCR) and TCR respectively]. However, there are indications that there are "holes" in the breadth of repertoire diversity, where no or few B or T cell are able to bind to a given antigen. Repertoire diversity may in these cases be a limiting factor for vaccine efficacy. Assuming that it is possible to predict which B and T cell receptors will respond to a given immunogen, vaccine strategies could be optimized and personalized. In addition, vaccine testing could be simplified if we could predict responses through sequencing BCR and TCRs. Bulk sequencing has shown putatively specific converging sequences after infection or vaccination. However, only single cell technologies have made it possible to capture the sequence of both heavy and light chains of a BCR or the alpha and beta chains the TCR. This has enabled the cloning of receptors and the functional validation of a predicted specificity. This review summarizes recent evidence of converging sequences in infectious diseases. Current and potential future applications of single cell technology in immune repertoire analysis are then discussed. Finally, possible short- and long- term implications for vaccine research are highlighted.
Collapse
Affiliation(s)
- Katja Fink
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
44
|
Li G, Bethune MT, Wong S, Joglekar AV, Leonard MT, Wang JK, Kim JT, Cheng D, Peng S, Zaretsky JM, Su Y, Luo Y, Heath JR, Ribas A, Witte ON, Baltimore D. T cell antigen discovery via trogocytosis. Nat Methods 2019; 16:183-190. [PMID: 30700903 DOI: 10.1038/s41592-018-0305-7] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/13/2018] [Indexed: 01/04/2023]
Abstract
T cell receptor (TCR) ligand discovery is essential for understanding and manipulating immune responses to tumors. We developed a cell-based selection platform for TCR ligand discovery that exploits a membrane transfer phenomenon called trogocytosis. We discovered that T cell membrane proteins are transferred specifically to target cells that present cognate peptide-major histocompatibility complex (MHC) molecules. Co-incubation of T cells expressing an orphan TCR with target cells collectively presenting a library of peptide-MHCs led to specific labeling of cognate target cells, enabling isolation of these target cells and sequencing of the cognate TCR ligand. We validated this method for two clinically employed TCRs and further used the platform to identify the cognate neoepitope for a subject-derived neoantigen-specific TCR. Thus, target cell trogocytosis is a robust tool for TCR ligand discovery that will be useful for studying basic tumor immunology and identifying new targets for immunotherapy.
Collapse
Affiliation(s)
- Guideng Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA. .,Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. .,Suzhou Institute of Systems Medicine, Suzhou, China.
| | - Michael T Bethune
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - Stephanie Wong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Alok V Joglekar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Michael T Leonard
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jessica K Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jocelyn T Kim
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Donghui Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Songming Peng
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jesse M Zaretsky
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yapeng Su
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.,Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Yicheng Luo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - James R Heath
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Parker Institute for Cancer Immunotherapy (PICI) Center, California Institute of Technology, Pasadena, CA, USA
| | - Antoni Ribas
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Division of Hematology & Oncology, Department of Medicine, and Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.,Parker Institute for Cancer Immunotherapy (PICI) Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Owen N Witte
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Parker Institute for Cancer Immunotherapy (PICI) Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA. .,Parker Institute for Cancer Immunotherapy (PICI) Center, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
45
|
Joglekar AV, Leonard MT, Jeppson JD, Swift M, Li G, Wong S, Peng S, Zaretsky JM, Heath JR, Ribas A, Bethune MT, Baltimore D. T cell antigen discovery via signaling and antigen-presenting bifunctional receptors. Nat Methods 2019; 16:191-198. [PMID: 30700902 PMCID: PMC6755906 DOI: 10.1038/s41592-018-0304-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 12/07/2018] [Indexed: 01/22/2023]
Abstract
CD8+ T cells recognize and eliminate tumors in an antigen-specific manner. Despite progress in characterizing the antitumor T cell repertoire and function, identifying their target antigens remains a challenge. Here, we describe the use of chimeric receptors called Signaling and Antigen-presenting Bifunctional Receptors (SABRs) in a novel cell-based platform for T Cell Receptor (TCR) antigen discovery. SABRs present an extracellular peptide-MHC complex and induce intracellular signaling via a TCR-like signal upon binding with a cognate TCR. We devised a strategy for antigen discovery using SABR libraries to screen thousands of antigenic epitopes. We validated this platform by identifying the targets recognized by public TCRs of known specificities. Moreover, we extended this approach for personalized neoantigen discovery. The antigen discovery platform reported here will provide a scalable and versatile way to develop novel targets for immunotherapy.
Collapse
Affiliation(s)
- Alok V Joglekar
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - Michael T Leonard
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - John D Jeppson
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Margaret Swift
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Guideng Li
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.,Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Suzhou Institute of Systems Medicine, Suzhou, China
| | - Stephanie Wong
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Songming Peng
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jesse M Zaretsky
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - James R Heath
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA.,Parker Institute for Cancer Immunotherapy (PICI), University of California, Los Angeles and California Institute of Technology, Pasadena, CA, USA
| | - Antoni Ribas
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Parker Institute for Cancer Immunotherapy (PICI), University of California, Los Angeles and California Institute of Technology, Pasadena, CA, USA.,Division of Hematology & Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael T Bethune
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - David Baltimore
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA. .,Parker Institute for Cancer Immunotherapy (PICI), University of California, Los Angeles and California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
46
|
Recent Development and Clinical Application of Cancer Vaccine: Targeting Neoantigens. J Immunol Res 2018; 2018:4325874. [PMID: 30662919 PMCID: PMC6313977 DOI: 10.1155/2018/4325874] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/18/2018] [Indexed: 12/30/2022] Open
Abstract
Recently, increasing data show that immunotherapy could be a powerful weapon against cancers. Comparing to the traditional surgery, chemotherapy or radiotherapy, immunotherapy more specifically targets cancer cells, giving rise to the opportunities to the patients to have higher response rates and better quality of life and even to cure the disease. Cancer vaccines could be designed to target tumor-associated antigens (TAAs), cancer germline antigens, virus-associated antigens, or tumor-specific antigens (TSAs), which are also called neoantigens. The cancer vaccines could be cell-based (e.g., dendritic cell vaccine provenge (sipuleucel-T) targeting prostatic acid phosphatase for metastatic prostate cancer), peptide/protein-based, or gene- (DNA/RNA) based, with the different kinds of adjuvants. Neoantigens are tumor-specific and could be presented by MHC molecules and recognized by T lymphocytes, serving the ideal immune targets to increase the therapeutic specificity and decrease the risk of nonspecific autoimmunity. By targeting the shared antigens and private epitopes, the cancer vaccine has potential to treat the disease. Accordingly, personalized neoantigen-based immunotherapies are emerging. In this article, we review the literature and evidence of the advantage and application of cancer vaccine. We summarize the recent clinical trials of neoantigen cancer vaccines which were designed according to the patients' personal mutanome. With the rapid development of personalized immunotherapy, it is believed that tumors could be efficiently controlled and become curable in the new era of precision medicine.
Collapse
|
47
|
Valdes-Mora F, Handler K, Law AMK, Salomon R, Oakes SR, Ormandy CJ, Gallego-Ortega D. Single-Cell Transcriptomics in Cancer Immunobiology: The Future of Precision Oncology. Front Immunol 2018; 9:2582. [PMID: 30483257 PMCID: PMC6240655 DOI: 10.3389/fimmu.2018.02582] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer is a heterogeneous and complex disease. Tumors are formed by cancer cells and a myriad of non-cancerous cell types that together with the extracellular matrix form the tumor microenvironment. These cancer-associated cells and components contribute to shape the progression of cancer and are deeply involved in patient outcome. The immune system is an essential part of the tumor microenvironment, and induction of cancer immunotolerance is a necessary step involved in tumor formation and growth. Immune mechanisms are intimately associated with cancer progression, invasion, and metastasis; as well as to tumor dormancy and modulation of sensitivity to drug therapy. Transcriptome analyses have been extensively used to understand the heterogeneity of tumors, classifying tumors into molecular subtypes and establishing signatures that predict response to therapy and patient outcomes. However, the classification of the tumor cell diversity and specially the identification of rare populations has been limited in these transcriptomic analyses of bulk tumor cell populations. Massively-parallel single-cell RNAseq analysis has emerged as a powerful method to unravel heterogeneity and to study rare cell populations in cancer, through unsupervised sampling and modeling of transcriptional states in single cells. In this context, the study of the role of the immune system in cancer would benefit from single cell approaches, as it will enable the characterization and/or discovery of the cell types and pathways involved in cancer immunotolerance otherwise missed in bulk transcriptomic information. Thus, the analysis of gene expression patterns at single cell resolution holds the potential to provide key information to develop precise and personalized cancer treatment including immunotherapy. This review is focused on the latest single-cell RNAseq methodologies able to agnostically study thousands of tumor cells as well as targeted single-cell RNAseq to study rare populations within tumors. In particular, we will discuss methods to study the immune system in cancer. We will also discuss the current challenges to the study of cancer at the single cell level and the potential solutions to the current approaches.
Collapse
Affiliation(s)
- Fatima Valdes-Mora
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Kristina Handler
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Andrew M K Law
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Robert Salomon
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Samantha R Oakes
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Christopher J Ormandy
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - David Gallego-Ortega
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
48
|
Bethune MT, Li XH, Yu J, McLaughlin J, Cheng D, Mathis C, Moreno BH, Woods K, Knights AJ, Garcia-Diaz A, Wong S, Hu-Lieskovan S, Puig-Saus C, Cebon J, Ribas A, Yang L, Witte ON, Baltimore D. Isolation and characterization of NY-ESO-1-specific T cell receptors restricted on various MHC molecules. Proc Natl Acad Sci U S A 2018; 115:E10702-E10711. [PMID: 30348802 PMCID: PMC6233129 DOI: 10.1073/pnas.1810653115] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tumor-specific T cell receptor (TCR) gene transfer enables specific and potent immune targeting of tumor antigens. Due to the prevalence of the HLA-A2 MHC class I supertype in most human populations, the majority of TCR gene therapy trials targeting public antigens have employed HLA-A2-restricted TCRs, limiting this approach to those patients expressing this allele. For these patients, TCR gene therapy trials have resulted in both tantalizing successes and lethal adverse events, underscoring the need for careful selection of antigenic targets. Broad and safe application of public antigen-targeted TCR gene therapies will require (i) selecting public antigens that are highly tumor-specific and (ii) targeting multiple epitopes derived from these antigens by obtaining an assortment of TCRs restricted by multiple common MHC alleles. The canonical cancer-testis antigen, NY-ESO-1, is not expressed in normal tissues but is aberrantly expressed across a broad array of cancer types. It has also been targeted with A2-restricted TCR gene therapy without adverse events or notable side effects. To enable the targeting of NY-ESO-1 in a broader array of HLA haplotypes, we isolated TCRs specific for NY-ESO-1 epitopes presented by four MHC molecules: HLA-A2, -B07, -B18, and -C03. Using these TCRs, we pilot an approach to extend TCR gene therapies targeting NY-ESO-1 to patient populations beyond those expressing HLA-A2.
Collapse
Affiliation(s)
- Michael T Bethune
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Xiao-Hua Li
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Jiaji Yu
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
- Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Jami McLaughlin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Donghui Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Colleen Mathis
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Blanca Homet Moreno
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
| | - Katherine Woods
- Cancer Immunobiology Laboratory, Olivia Newton-John Cancer Research Institute, Austin Hospital, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Cancer Immunobiology Laboratory, Ludwig Institute for Cancer Research, Heidelberg, VIC 3084, Australia
| | - Ashley J Knights
- Cancer Immunobiology Laboratory, Ludwig Institute for Cancer Research, Heidelberg, VIC 3084, Australia
| | - Angel Garcia-Diaz
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
| | - Stephanie Wong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Siwen Hu-Lieskovan
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
| | - Cristina Puig-Saus
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
| | - Jonathan Cebon
- Cancer Immunobiology Laboratory, Olivia Newton-John Cancer Research Institute, Austin Hospital, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Cancer Immunobiology Laboratory, Ludwig Institute for Cancer Research, Heidelberg, VIC 3084, Australia
| | - Antoni Ribas
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Medicine, University of California, Los Angeles, CA 90095
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, CA 90095
| | - Lili Yang
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095;
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Owen N Witte
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095;
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, CA 90095
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125;
| |
Collapse
|
49
|
Servín-Blanco R, Chávaro-Ortiz RM, Zamora-Alvarado R, Martínez-Cortes F, Gevorkian G, Manoutcharian K. Generation of cancer vaccine immunogens derived from major histocompatibility complex (MHC) class I molecules using variable epitope libraries. Immunol Lett 2018; 204:47-54. [PMID: 30339819 DOI: 10.1016/j.imlet.2018.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/04/2018] [Accepted: 10/15/2018] [Indexed: 12/22/2022]
Abstract
Although various immune checkpoint inhibitors (ICIs), used for the treatment of advanced cancer, showed remarkably durable tumor regression in a subset of patients, there are important limitations in a large group of non-responders, and the generation of novel immunogens capable of inducing protective cellular immune responses is a priority in cancer immunotherapy field. During the last decades, several types of vaccine immunogens have been used in numerous preclinical studies and clinical trials. However, although immunity to tumor Ags can be elicited by most vaccines tested, their clinical efficacy remains modest. Recently, we have developed an innovative vaccine concept, called Variable Epitope Libraries (VELs), with the purpose to exploit the high antigenic variability of many important pathogens and tumor cells as starting points for the construction of a new class of vaccine immunogens capable of inducing the largest possible repertoire of both B and T cells. In the present study, we decided to generate VEL immunogens derived from both classical and non-classical major histocompatibility complex (MHC) class I molecules. The MHC molecules, responsible for antigen presentation and subsequent activation of T lymphocytes, undergo multiple modifications that directly affect their proper function, resulting in immune escape of tumor cells. Two large VELs derived from multi-epitope region of H2-Kd and Qa-2 sequences (46 and 34 amino acids long, respectively), along with their wild type counterparts have been generated as synthetic peptides and tested in an aggressive 4T1 mouse model of breast cancer. Significant inhibition of tumor growth and the reduction of metastatic lesions in the lungs of immunized mice were observed. This study demonstrated for the first time the successful application of VELs carrying combinatorial libraries of epitope variants derived from MHC class I molecules as novel vaccine immunogens.
Collapse
Affiliation(s)
- Rodolfo Servín-Blanco
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria, CDMX, 04510, Mexico
| | - Rosa Mariana Chávaro-Ortiz
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria, CDMX, 04510, Mexico
| | - Rubén Zamora-Alvarado
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria, CDMX, 04510, Mexico
| | - Fernando Martínez-Cortes
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria, CDMX, 04510, Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria, CDMX, 04510, Mexico
| | - Karen Manoutcharian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria, CDMX, 04510, Mexico.
| |
Collapse
|
50
|
RNA editing derived epitopes function as cancer antigens to elicit immune responses. Nat Commun 2018; 9:3919. [PMID: 30254248 PMCID: PMC6156571 DOI: 10.1038/s41467-018-06405-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023] Open
Abstract
In addition to genomic mutations, RNA editing is another major mechanism creating sequence variations in proteins by introducing nucleotide changes in mRNA sequences. Deregulated RNA editing contributes to different types of human diseases, including cancers. Here we report that peptides generated as a consequence of RNA editing are indeed naturally presented by human leukocyte antigen (HLA) molecules. We provide evidence that effector CD8+ T cells specific for edited peptides derived from cyclin I are present in human tumours and attack tumour cells that are presenting these epitopes. We show that subpopulations of cancer patients have increased peptide levels and that levels of edited RNA correlate with peptide copy numbers. These findings demonstrate that RNA editing extends the classes of HLA presented self-antigens and that these antigens can be recognised by the immune system. RNA editing is a biological process that creates sequence variation. Here the authors show that peptides generated as a consequence of RNA editing are naturally presented by human leukocyte antigen (HLA) and serve as antigens to elicit anti-tumour immune responses.
Collapse
|