1
|
Wang X, Cheng M, Chen S, Zhang C, Ling R, Qiu S, Chen K, Zhou B, Li Q, Lei W, Chen D. Resistance to anti-LAG-3 plus anti-PD-1 therapy in head and neck cancer is mediated by Sox9+ tumor cells interaction with Fpr1+ neutrophils. Nat Commun 2025; 16:3975. [PMID: 40295483 PMCID: PMC12037843 DOI: 10.1038/s41467-025-59050-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Relatlimab and nivolumab combination therapy shows significant efficacy in treating various types of cancer. Current research on the molecular mechanisms of this treatment is abundant, but in-depth investigations into post-treatment resistance remain notably lacking. In this study, we identify significant enrichment of SRY (sex determining region Y)-box 9 (Sox9)+ tumor cells in resistant samples using single cell RNA sequencing (scRNAseq) in a head and neck squamous cell carcinoma (HNSCC) mouse model. In addition, Sox9 directly regulates the expression of annexin A1 (Anxa1), mediating apoptosis of formyl peptide receptor 1 (Fpr1)+ neutrophils through the Anxa1-Fpr1 axis, which promotes mitochondrial fission, inhibits mitophagy by downregulating BCL2/adenovirus E1B interacting protein 3 (Bnip3) expression and ultimately prevents the accumulation of neutrophils in tumor tissues. The reduction of Fpr1+ neutrophils impairs the infiltration and tumor cell-killing ability of cytotoxic Cd8 T and γδT cells within the tumor microenvironment, thereby leading to the development of resistance to the combination therapy. We further validate these findings using various transgenic mouse models. Overall, this study comprehensively explains the mechanisms underlying resistance to the anti-LAG-3 plus anti-PD-1 combination therapy and identifies potential therapeutic targets to overcome this resistance.
Collapse
Affiliation(s)
- Xiaochen Wang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Maosheng Cheng
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuang Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Caihua Zhang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Rongsong Ling
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuqing Qiu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ke Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Qiuli Li
- Department of Head and Neck Surgery, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Wenbin Lei
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Demeng Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
2
|
Xue J, Chen X, An X, Wang J, Zang M, Mao B, Guo S, Yang T, Kumari R, Li QX. An NGS-based assay for accurate detection and quantification of immune gene expression in mouse tumor models. PLoS One 2024; 19:e0303171. [PMID: 38768113 PMCID: PMC11104603 DOI: 10.1371/journal.pone.0303171] [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: 12/30/2023] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
Tumor microenvironment (TME) is a complex dynamic system with many tumor-interacting components including tumor-infiltrating leukocytes (TILs), cancer associated fibroblasts, blood vessels, and other stromal constituents. It intrinsically affects tumor development and pharmacology of oncology therapeutics, particularly immune-oncology (IO) treatments. Accurate measurement of TME is therefore of great importance for understanding the tumor immunity, identifying IO treatment mechanisms, developing predictive biomarkers, and ultimately, improving the treatment of cancer. Here, we introduce a mouse-IO NGS-based (NGSmIO) assay for accurately detecting and quantifying the mRNA expression of 1080 TME related genes in mouse tumor models. The NGSmIO panel was shown to be superior to the commonly used microarray approach by hosting 300 more relevant genes to better characterize various lineage of immune cells, exhibits improved mRNA and protein expression correlation to flow cytometry, shows stronger correlation with mRNA expression than RNAseq with 10x higher sequencing depth, and demonstrates higher sensitivity in measuring low-expressed genes. We describe two studies; firstly, detecting the pharmacodynamic change of interferon-γ expression levels upon anti-PD-1: anti-CD4 combination treatment in MC38 and Hepa 1-6 tumors; and secondly, benchmarking baseline TILs in 14 syngeneic tumors using transcript level expression of lineage specific genes, which demonstrate effective and robust applications of the NGSmIO panel.
Collapse
Affiliation(s)
- Jia Xue
- Crown Bioscience, Inc., San Diego, California, United States of America
| | - Xiaobo Chen
- Crown Bioscience, Inc., San Diego, California, United States of America
| | - Xiaoyu An
- Crown Bioscience, Inc., San Diego, California, United States of America
| | - Jingjing Wang
- Crown Bioscience, Inc., San Diego, California, United States of America
| | - Mingfa Zang
- Crown Bioscience, Inc., San Diego, California, United States of America
| | - Binchen Mao
- Crown Bioscience, Inc., San Diego, California, United States of America
| | - Sheng Guo
- Crown Bioscience, Inc., San Diego, California, United States of America
| | - Tao Yang
- Crown Bioscience, Inc., San Diego, California, United States of America
| | - Rajendra Kumari
- Crown Bioscience, Inc., San Diego, California, United States of America
| | - Qi-Xiang Li
- Crown Bioscience, Inc., San Diego, California, United States of America
| |
Collapse
|
3
|
Boccalatte F, Mina R, Aroldi A, Leone S, Suryadevara CM, Placantonakis DG, Bruno B. Advances and Hurdles in CAR T Cell Immune Therapy for Solid Tumors. Cancers (Basel) 2022; 14:5108. [PMID: 36291891 PMCID: PMC9600451 DOI: 10.3390/cancers14205108] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/28/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells in solid tumors have so far yielded limited results, in terms of therapeutic effects, as compared to the dramatic results observed for hematological malignancies. Many factors involve both the tumor cells and the microenvironment. The lack of specific target antigens and severe, potentially fatal, toxicities caused by on-target off-tumor toxicities constitute major hurdles. Furthermore, the tumor microenvironment is usually characterized by chronic inflammation, the presence of immunosuppressive molecules, and immune cells that can reduce CAR T cell efficacy and facilitate antigen escape. Nonetheless, solid tumors are under investigation as possible targets despite their complexity, which represents a significant challenge. In preclinical mouse models, CAR T cells are able to efficiently recognize and kill several tumor xenografts. Overall, in the next few years, there will be intensive research into optimizing novel cell therapies to improve their effector functions and keep untoward effects in check. In this review, we provide an update on the state-of-the-art CAR T cell therapies in solid tumors, focusing on the preclinical studies and preliminary clinical findings aimed at developing optimal strategies to reduce toxicity and improve efficacy.
Collapse
Affiliation(s)
- Francesco Boccalatte
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Roberto Mina
- Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, University of Torino, 10126 Torino, TO, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, TO, Italy
| | - Andrea Aroldi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, MB, Italy
| | - Sarah Leone
- Department of Population Health, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Carter M. Suryadevara
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Dimitris G. Placantonakis
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
- Brain and Spine Tumor Center/Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Benedetto Bruno
- Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, University of Torino, 10126 Torino, TO, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, TO, Italy
| |
Collapse
|
4
|
Shang P, Yu L, Cao S, Guo C, Zhang W. An improved cell line-derived xenograft humanized mouse model for evaluation of PD-1/PD-L1 blocker BMS202-induced immune responses in colorectal cancer. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1497-1506. [PMID: 36269133 PMCID: PMC9827804 DOI: 10.3724/abbs.2022145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The establishment of an in vivo mouse model mimicking human tumor-immune environments provides a promising platform for immunotherapy assessment, drug discovery and clinical decision guidance. To this end, we construct humanized NCG mice by transplanting human hCD34 + hematopoietic progenitors into non-obese diabetic (NOD) Cg- Prkdc scidIL2rg tm1Wjl /Sz (null; NCG) mice and monitoring the development of human hematopoietic and immune systems (Hu-NCG). The cell line-derived xenograft (CDX) Hu-NCG mouse models are set up to assess the outcome of immunotherapy mediated by the small molecule BMS202. As a PD-1/PD-L1 blocker, BMS202 shows satisfactory antitumour efficacy in the HCT116 and SW480 xenograft Hu-NCG mouse models. Mechanistically, BMS202 exerts antitumour efficacy by improving the tumor microenvironment and enhancing the infiltration of hCD8 + T cells and the release of hIFNγ in tumor tissue. Thus, tumor-bearing Hu-NCG mice are a suitable and important in vivo model for preclinical study, particularly in cancer immunotherapy.
Collapse
Affiliation(s)
- Pengzhao Shang
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing210009China
| | - Liting Yu
- Department of PharmacyBinzhou Medical UniversityYantai264003China
| | - Shucheng Cao
- School of EngineeringChina Pharmaceutical UniversityNanjing210009China
| | - Changying Guo
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing210009China,Correspondence address. Tel: +86-15737957481; (W.Z.) / Tel: +86-18252099426; (C.G.) @cpu.edu.cn
| | - Wanheng Zhang
- Department of Pharmacythe First Affiliated Hospitaland College of Clinical Medicine of Henan University of Science and TechnologyLuoyang471003China,Correspondence address. Tel: +86-15737957481; (W.Z.) / Tel: +86-18252099426; (C.G.) @cpu.edu.cn
| |
Collapse
|
5
|
Chiu WC, Ou DL, Tan CT. Mouse Models for Immune Checkpoint Blockade Therapeutic Research in Oral Cancer. Int J Mol Sci 2022; 23:ijms23169195. [PMID: 36012461 PMCID: PMC9409124 DOI: 10.3390/ijms23169195] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
The most prevalent oral cancer globally is oral squamous cell carcinoma (OSCC). The invasion of adjacent bones and the metastasis to regional lymph nodes often lead to poor prognoses and shortened survival times in patients with OSCC. Encouraging immunotherapeutic responses have been seen with immune checkpoint inhibitors (ICIs); however, these positive responses to monotherapy have been limited to a small subset of patients. Therefore, it is urgent that further investigations into optimizing immunotherapies are conducted. Areas of research include identifying novel immune checkpoints and targets and tailoring treatment programs to meet the needs of individual patients. Furthermore, the advancement of combination therapies against OSCC is also critical. Thus, additional studies are needed to ensure clinical trials are successful. Mice models are advantageous in immunotherapy research with several advantages, such as relatively low costs and high tumor growth success rate. This review paper divided methods for establishing OSCC mouse models into four categories: syngeneic tumor models, chemical carcinogen induction, genetically engineered mouse, and humanized mouse. Each method has advantages and disadvantages that influence its application in OSCC research. This review comprehensively surveys the literature and summarizes the current mouse models used in immunotherapy, their advantages and disadvantages, and details relating to the cell lines for oral cancer growth. This review aims to present evidence and considerations for choosing a suitable model establishment method to investigate the early diagnosis, clinical treatment, and related pathogenesis of OSCC.
Collapse
Affiliation(s)
- Wei-Chiao Chiu
- Department of Medical Research, Fu-Jen Catholic University Hospital, Fu-Jen Catholic University, New Taipei City 24352, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei City 100225, Taiwan
| | - Da-Liang Ou
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei City 10051, Taiwan
- YongLin Institute of Health, National Taiwan University, Taipei City 10672, Taiwan
| | - Ching-Ting Tan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei City 100225, Taiwan
- Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei City 10051, Taiwan
- Department of Otolaryngology, College of Medicine, National Taiwan University, Taipei City 100233, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302058, Taiwan
- Correspondence: ; Tel.: +886-2-23123456 (ext. 88649)
| |
Collapse
|
6
|
Letchuman V, Ampie L, Shah AH, Brown DA, Heiss JD, Chittiboina P. Syngeneic murine glioblastoma models: reactionary immune changes and immunotherapy intervention outcomes. Neurosurg Focus 2022; 52:E5. [PMID: 35104794 PMCID: PMC10851929 DOI: 10.3171/2021.11.focus21556] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
Abstract
Glioblastoma is the most common primary malignant brain neoplasm with dismal 10-year survival rates of < 1%. Despite promising preliminary results from several novel therapeutic agents, clinical responses have been modest due to several factors, including tumor heterogeneity, immunosuppressive tumor microenvironment, and treatment resistance. Novel immunotherapeutics have been developed to reverse tumor-induced immunosuppression in patients with glioblastomas. In order to recapitulate the tumor microenvironment, reliable in vivo syngeneic murine models are critical for the development of new targeted agents as these models demonstrate rapid tumor induction and reliable tumor growth over multiple generations. Despite the clear advantages of murine models, choosing an appropriate model from an immunological perspective can be difficult and have significant ramifications on the translatability of the results from murine to human trials. Herein, the authors reviewed the 4 most commonly used immunocompetent syngeneic murine glioma models (GL261 [C57BL/6], SB28 [C57BL/6], CT-2A [C57BL/6], and SMA-560 [VM/Dk]) and compared their strengths and weaknesses from an immunological standpoint.
Collapse
Affiliation(s)
- Vijay Letchuman
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Leonel Ampie
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Ashish H. Shah
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Desmond A. Brown
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - John D. Heiss
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Prashant Chittiboina
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
7
|
Investigating T Cell Immunity in Cancer: Achievements and Prospects. Int J Mol Sci 2021; 22:ijms22062907. [PMID: 33809369 PMCID: PMC7999898 DOI: 10.3390/ijms22062907] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 12/21/2022] Open
Abstract
T cells play a key role in tumour surveillance, both identifying and eliminating transformed cells. However, as tumours become established they form their own suppressive microenvironments capable of shutting down T cell function, and allowing tumours to persist and grow. To further understand the tumour microenvironment, including the interplay between different immune cells and their role in anti-tumour immune responses, a number of studies from mouse models to clinical trials have been performed. In this review, we examine mechanisms utilized by tumour cells to reduce their visibility to CD8+ Cytotoxic T lymphocytes (CTL), as well as therapeutic strategies trialled to overcome these tumour-evasion mechanisms. Next, we summarize recent advances in approaches to enhance CAR T cell activity and persistence over the past 10 years, including bispecific CAR T cell design and early evidence of efficacy. Lastly, we examine mechanisms of T cell infiltration and tumour regression, and discuss the strengths and weaknesses of different strategies to investigate T cell function in murine tumour models.
Collapse
|
8
|
Pilot Acute Safety Evaluation of Innocell™ Cancer Immunotherapy in Canine Subjects. J Immunol Res 2020; 2020:7142375. [PMID: 33102607 PMCID: PMC7568153 DOI: 10.1155/2020/7142375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/09/2020] [Accepted: 09/22/2020] [Indexed: 12/30/2022] Open
Abstract
Background We are developing cancer immunotherapy based on the use of autologous tumor tissue that has been rendered replication-incompetent but maintains phenotype and metabolic activity post-preparation. Aim The aim of this study was to evaluate safety and tolerance to injection of the inactivated tumor cell and adjuvant preparation (Innocell™) within 24 hours of administration in a pilot study in canine patients with solid organ tumors. Methodology. Three canine patients demonstrating accessible solid organ tumors of various types were assessed in this study. The local site injection was monitored post-treatment. Clinical signs of adverse reactions were monitored for 24 hours post-treatment. Blood samples were taken pre-treatment and at 8 and 24 hours post-treatment for all subjects. One subject provided samples at 7 days post-treatment. All blood samples were analyzed for cytokine content for both immune system-associated and tumor-associated cytokines. Results No signs of adverse reactions at the site of injection or systemically were observed in the study period. A slight fever and lethargy were reported in one subject by the owner post-vaccination. Immune system-associated cytokine levels in two of the three animals were elevated post-treatment. Tumor-associated cytokine levels in all three subjects declined post-treatment from baseline levels with the effect most prominent in the subject with a non-excised tumor. Conclusion Subcutaneous injection of the inactivated tumor cells and adjuvant was well tolerated in this pilot study. Cytokine responses observed were in line with the intended use of the treatment in stimulating immune response without adverse clinical observations. Additional evaluation is warranted.
Collapse
|
9
|
Chulpanova DS, Kitaeva KV, Rutland CS, Rizvanov AA, Solovyeva VV. Mouse Tumor Models for Advanced Cancer Immunotherapy. Int J Mol Sci 2020; 21:E4118. [PMID: 32526987 PMCID: PMC7312663 DOI: 10.3390/ijms21114118] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 12/21/2022] Open
Abstract
Recent advances in the development of new methods of cancer immunotherapy require the production of complex cancer animal models that reliably reflect the complexity of the tumor and its microenvironment. Mice are good animals to create tumor models because they are low cost, have a short reproductive cycle, exhibit high tumor growth rates, and can be easily genetically modified. However, the obvious problem of these models is the high failure rate observed in human clinical trials after promising results obtained in mouse models. In order to increase the reliability of the results obtained in mice, the tumor model should reflect the heterogeneity of the tumor, contain components of the tumor microenvironment, in particular immune cells, to which the action of immunotherapeutic drugs are directed. This review discusses the current immunocompetent and immunocompromised mouse models of human tumors that are used to evaluate the effectiveness of immunotherapeutic agents, in particular chimeric antigen receptor (CAR) T-cells and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Daria S. Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| | - Kristina V. Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| | - Catrin S. Rutland
- Faculty of Medicine and Health Sciences, University of Medicine, Nottingham NG7 2HA, UK;
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| |
Collapse
|
10
|
Whyte CE, Osman M, Kara EE, Abbott C, Foeng J, McKenzie DR, Fenix KA, Harata-Lee Y, Foyle KL, Boyle ST, Kochetkova M, Aguilera AR, Hou J, Li XY, Armstrong MA, Pederson SM, Comerford I, Smyth MJ, McColl SR. ACKR4 restrains antitumor immunity by regulating CCL21. J Exp Med 2020; 217:e20190634. [PMID: 32289156 PMCID: PMC7971131 DOI: 10.1084/jem.20190634] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 02/03/2020] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
Current immunotherapies involving CD8+ T cell responses show remarkable promise, but their efficacy in many solid tumors is limited, in part due to the low frequency of tumor-specific T cells in the tumor microenvironment (TME). Here, we identified a role for host atypical chemokine receptor 4 (ACKR4) in controlling intratumor T cell accumulation and activation. In the absence of ACKR4, an increase in intratumor CD8+ T cells inhibited tumor growth, and nonhematopoietic ACKR4 expression was critical. We show that ACKR4 inhibited CD103+ dendritic cell retention in tumors through regulation of the intratumor abundance of CCL21. In addition, preclinical studies indicate that ACKR4 and CCL21 are potential therapeutic targets to enhance responsiveness to immune checkpoint blockade or T cell costimulation.
Collapse
Affiliation(s)
- Carly E. Whyte
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Maleika Osman
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Ervin E. Kara
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Caitlin Abbott
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jade Foeng
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Duncan R. McKenzie
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kevin A. Fenix
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Yuka Harata-Lee
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kerrie L. Foyle
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Sarah T. Boyle
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Marina Kochetkova
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Amelia Roman Aguilera
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jiajie Hou
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Xian-Yang Li
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mark A. Armstrong
- Bioinformatics Hub, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Stephen M. Pederson
- Bioinformatics Hub, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Iain Comerford
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Shaun R. McColl
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
11
|
Bresnahan E, Lindblad KE, Ruiz de Galarreta M, Lujambio A. Mouse Models of Oncoimmunology in Hepatocellular Carcinoma. Clin Cancer Res 2020; 26:5276-5286. [PMID: 32327473 DOI: 10.1158/1078-0432.ccr-19-2923] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/10/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Liver cancer is the fourth leading cause of cancer-related mortality worldwide and incidence is on the rise. Hepatocellular carcinoma (HCC) is the most common form of liver cancer, with a complex etiology and limited treatment options. The standard-of-care treatment for patients with advanced HCC is sorafenib, a tyrosine kinase inhibitor that offers limited survival benefit. In the past years, therapeutic options for the treatment of advanced HCC have increased substantially, including additional multikinase inhibitors as well as immune checkpoint inhibitors. Nivolumab and pembrolizumab were approved in 2017 and 2018, respectively, as second-line treatment in advanced HCC. These drugs, both targeting the programmed death-1 pathway, demonstrate unprecedented results, with objective response rates of approximately 20%. However, the majority of patients do not respond, necessitating the identification of biomarkers of response and resistance to immunotherapy. With the recent success of immunotherapies in oncology, mouse models that better recapitulate the human disease and antitumor immune response are needed. This review lists ongoing clinical trials testing immunotherapy in HCC, briefly discusses the unique immunosuppressive environment of the liver, and then delves into the most applicable current murine model systems to study oncoimmunology within the context of HCC, including syngeneic, genetically engineered, and humanized models.
Collapse
Affiliation(s)
- Erin Bresnahan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Katherine E Lindblad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marina Ruiz de Galarreta
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York. .,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
12
|
Wu H, Wang Y, Wang G, Qiu Z, Hu X, Zhang H, Yan X, Ke F, Zou A, Wang M, Liao Y, Chen X. A bivalent antihypertensive vaccine targeting L-type calcium channels and angiotensin AT 1 receptors. Br J Pharmacol 2019; 177:402-419. [PMID: 31625597 DOI: 10.1111/bph.14875] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 09/09/2019] [Accepted: 09/15/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Hypertension has been the leading preventable cause of premature death worldwide. The aim of this study was to design a more efficient vaccine against novel targets for the treatment of hypertension. EXPERIMENTAL APPROACH The epitope CE12, derived from the human L-type calcium channel (CaV 1.2), was designed and conjugated with Qβ bacteriophage virus-like particles to test the efficacy in hypertensive animals. Further, the hepatitis B core antigen (HBcAg)-CE12-CQ10 vaccine, a bivalent vaccine based on HBcAg virus-like particles and targeting both human angiotensin AT1 receptors and CaV 1.2 channels, was developed and evaluated in hypertensive rodents. KEY RESULTS The Qβ-CE12 vaccine effectively decreased the BP in hypertensive rodents. A monoclonal antibody against CE12 specifically bound to L-type calcium channels and inhibited channel activity. Injection with monoclonal antibody against CE12 effectively reduced the BP in angiotensin II-induced hypertensive mice. The HBcAg-CE12-CQ10 vaccine showed antihypertensive effects in hypertensive mice and relatively superior antihypertensive effects in spontaneously hypertensive rats and ameliorated L-NAME-induced renal injury. In addition, no obvious immune-mediated damage or electrophysiological adverse effects were detected. CONCLUSION AND IMPLICATIONS Immunotherapy against both AT1 receptors and CaV 1.2 channels decreased the BP in hypertensive rodents effectively and provided protection against hypertensive target organ damage without obvious feedback activation of renin-angiotensin system or induction of dominant antibodies against the carrier protein. Thus, the HBcAg-CE12-CQ10 vaccine may provide a novel and promising therapeutic approach for hypertension.
Collapse
Affiliation(s)
- Hailang Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyi Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gongxin Wang
- Electrophysiological Laboratory, Qingdao Haiwei Biopharma Co. Ltd, Qingdao, China
| | - Zhihua Qiu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiajun Hu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongrong Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaole Yan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Ke
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anruo Zou
- Electrophysiological Laboratory, Qingdao Haiwei Biopharma Co. Ltd, Qingdao, China
| | - Min Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhua Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Mouse Models for Immunotherapy in Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11111800. [PMID: 31731753 PMCID: PMC6896030 DOI: 10.3390/cancers11111800] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022] Open
Abstract
Liver cancer is one of the dominant causes of cancer-related mortality, and the survival rate of liver cancer is among the lowest for all cancers. Immunotherapy for hepatocellular carcinoma (HCC) has yielded some encouraging results, but the percentage of patients responding to single-agent therapies remains low. Therefore, potential directions for improved immunotherapies include identifying new immune targets and checkpoints and customizing treatment procedures for individual patients. The development of combination therapies for HCC is also crucial and urgent and, thus, further studies are required. Mice have been utilized in immunotherapy research due to several advantages, for example, being low in cost, having high success rates for inducing tumor growth, and so on. Moreover, immune-competent mice are used in immunotherapy research to clarify the role that the immune system plays in cancer growth. In this review paper, the advantages and disadvantages of mouse models for immunotherapy, the equipment that are used for monitoring HCC, and the cell strains used for inducing HCC are reviewed.
Collapse
|
14
|
Li XY, Moesta AK, Xiao C, Nakamura K, Casey M, Zhang H, Madore J, Lepletier A, Aguilera AR, Sundarrajan A, Jacoberger-Foissac C, Wong C, Dela Cruz T, Welch M, Lerner AG, Spatola BN, Soros VB, Corbin J, Anderson AC, Effern M, Hölzel M, Robson SC, Johnston RL, Waddell N, Smith C, Bald T, Geetha N, Beers C, Teng MWL, Smyth MJ. Targeting CD39 in Cancer Reveals an Extracellular ATP- and Inflammasome-Driven Tumor Immunity. Cancer Discov 2019; 9:1754-1773. [PMID: 31699796 DOI: 10.1158/2159-8290.cd-19-0541] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 09/04/2019] [Accepted: 10/01/2019] [Indexed: 01/24/2023]
Abstract
We explored the mechanism of action of CD39 antibodies that inhibit ectoenzyme CD39 conversion of extracellular ATP (eATP) to AMP and thus potentially augment eATP-P2-mediated proinflammatory responses. Using syngeneic and humanized tumor models, we contrast the potency and mechanism of anti-CD39 mAbs with other agents targeting the adenosinergic pathway. We demonstrate the critical importance of an eATP-P2X7-ASC-NALP3-inflammasome-IL18 pathway in the antitumor activity mediated by CD39 enzyme blockade, rather than simply reducing adenosine as mechanism of action. Efficacy of anti-CD39 activity was underpinned by CD39 and P2X7 coexpression on intratumor myeloid subsets, an early signature of macrophage depletion, and active IL18 release that facilitated the significant expansion of intratumor effector T cells. More importantly, anti-CD39 facilitated infiltration into T cell-poor tumors and rescued anti-PD-1 resistance. Anti-human CD39 enhanced human T-cell proliferation and Th1 cytokine production and suppressed human B-cell lymphoma in the context of autologous Epstein-Barr virus-specific T-cell transfer. SIGNIFICANCE: Overall, these data describe a potent and novel mechanism of action of antibodies that block mouse or human CD39, triggering an eATP-P2X7-inflammasome-IL18 axis that reduces intratumor macrophage number, enhances intratumor T-cell effector function, overcomes anti-PD-1 resistance, and potentially enhances the efficacy of adoptive T-cell transfer.This article is highlighted in the In This Issue feature, p. 1631.
Collapse
Affiliation(s)
- Xian-Yang Li
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | - Christos Xiao
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Kyohei Nakamura
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Mika Casey
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Haiyan Zhang
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Jason Madore
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ailin Lepletier
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Amelia Roman Aguilera
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ashmitha Sundarrajan
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Celia Jacoberger-Foissac
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | | | - Megan Welch
- Tizona Therapeutics, San Francisco, California
| | | | | | | | - John Corbin
- Tizona Therapeutics, San Francisco, California
| | - Ana C Anderson
- Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Maike Effern
- Unit of RNA Biology, Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Experimental Oncology (IEO), University Hospital Bonn, University of Bonn, Bonn, Germany
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Michael Hölzel
- Unit of RNA Biology, Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Experimental Oncology (IEO), University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Simon C Robson
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Rebecca L Johnston
- Medical Genomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Nicola Waddell
- Medical Genomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Corey Smith
- Translational and Human Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Tobias Bald
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Nishamol Geetha
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | - Michele W L Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.
| |
Collapse
|
15
|
Faithful preclinical mouse models for better translation to bedside in the field of immuno-oncology. Int J Clin Oncol 2019; 25:831-841. [PMID: 31407168 DOI: 10.1007/s10147-019-01520-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/01/2019] [Indexed: 12/19/2022]
Abstract
The success of immunotherapy using immune checkpoint inhibitors has changed the practice of cancer treatment tremendously. However, there are still many clinical challenges, such as drug resistance, predictive biomarker development, exploration of combination therapies, and prediction of immune-related adverse events in preclinical settings. To overcome these problems, it is essential to establish faithful preclinical mouse models that recapitulate the clinical features, molecular genetics, biological heterogeneity, and immune microenvironment of human cancers. Here we review the advantages and disadvantages of current preclinical mouse models, including syngeneic murine tumor cell lines, autochthonous tumor models, cancer cell line-derived xenografts, patient-derived-xenografts, and various kinds of immunologically humanized mice. We discuss how these models should be characterized and applied in preclinical settings, and how we should prepare preclinical studies for successful translation from bench to bedside.
Collapse
|
16
|
Meraz IM, Majidi M, Meng F, Shao R, Ha MJ, Neri S, Fang B, Lin SH, Tinkey PT, Shpall EJ, Morris J, Roth JA. An Improved Patient-Derived Xenograft Humanized Mouse Model for Evaluation of Lung Cancer Immune Responses. Cancer Immunol Res 2019; 7:1267-1279. [PMID: 31186248 PMCID: PMC7213862 DOI: 10.1158/2326-6066.cir-18-0874] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/08/2019] [Accepted: 05/31/2019] [Indexed: 12/17/2022]
Abstract
Human tumor xenograft models do not replicate the human immune system and tumor microenvironment. We developed an improved humanized mouse model, derived from fresh cord blood CD34+ stem cells (CD34+ HSC), and combined it with lung cancer cell line-derived human xenografts or patient-derived xenografts (Hu-PDX). Fresh CD34+ HSCs could reconstitute detectable mature human leukocytes (hCD45+) in mice at four weeks without the onset of graft-versus-host disease (GVHD). Repopulated human T cells, B cells, natural killer (NK) cells, dendritic cells (DC), and myeloid-derived suppressor cells (MDSC) increased in peripheral blood, spleen, and bone marrow over time. Although cultured CD34+ HSCs labeled with luciferase could be detected in mice, the cultured HSCs did not develop into mature human immune cells by four weeks, unlike fresh CD34+ HSCs. Ex vivo, reconstituted T cells, obtained from the tumor-bearing humanized mice, secreted IFNγ upon treatment with phorbol myristate acetate (PMA) or exposure to human A549 lung tumor cells and mediated antigen-specific CTL responses, indicating functional activity. Growth of engrafted PDXs and tumor xenografts was not dependent on the human leukocyte antigen status of the donor. Treatment with the anti-PD-1 checkpoint inhibitors pembrolizumab or nivolumab inhibited tumor growth in humanized mice significantly, and correlated with an increased number of CTLs and decreased MDSCs, regardless of the donor HLA type. In conclusion, fresh CD34+HSCs are more effective than their expanded counterparts in humanizing mice, and do so in a shorter time. The Hu-PDX model provides an improved platform for evaluation of immunotherapy.
Collapse
Affiliation(s)
- Ismail M Meraz
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Mourad Majidi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Feng Meng
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - RuPing Shao
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Min Jin Ha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shinya Neri
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peggy T Tinkey
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey Morris
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Thoracic Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
17
|
Mouse-Derived Isograft (MDI) In Vivo Tumor Models I. Spontaneous sMDI Models: Characterization and Cancer Therapeutic Approaches. Cancers (Basel) 2019; 11:cancers11020244. [PMID: 30791466 PMCID: PMC6406567 DOI: 10.3390/cancers11020244] [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: 12/01/2018] [Revised: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 01/05/2023] Open
Abstract
Syngeneic in vivo tumor models are valuable for the development and investigation of immune-modulating anti-cancer drugs. In the present study, we established a novel syngeneic in vivo model type named mouse-derived isografts (MDIs). Spontaneous MDIs (sMDIs) were obtained during a long-term observation period (more than one to two years) of naïve and untreated animals of various mouse strains (C3H/HeJ, CBA/J, DBA/2N, BALB/c, and C57BL/6N). Primary tumors or suspicious tissues were assessed macroscopically and re-transplanted in a PDX-like manner as small tumor pieces into sex-matched syngeneic animals. Nine outgrowing primary tumors were histologically characterized either as adenocarcinomas, histiocytic carcinomas, or lymphomas. Growth of the tumor pieces after re-transplantation displayed model heterogeneity. The adenocarcinoma sMDI model JA-0009 was further characterized by flow cytometry, RNA-sequencing, and efficacy studies. M2 macrophages were found to be the main tumor infiltrating leukocyte population, whereas only a few T cells were observed. JA-0009 showed limited sensitivity when treated with antibodies against inhibitory checkpoint molecules (anti-mPD-1 and anti-mCTLA-4), but high sensitivity to gemcitabine treatment. The generated sMDI are spontaneously occurring tumors of low passage number, propagated as tissue pieces in mice without any tissue culturing, and thus conserving the original tumor characteristics and intratumoral immune cell populations.
Collapse
|
18
|
Affiliation(s)
- Jenny Lou
- Department of Medical BiophysicsUniversity of Toronto Toronto M5G 1L7 Canada
- Princess Margaret Cancer CenterUniversity Health Network Toronto M5G 2C1 Canada
- Centre for Pharmaceutical OncologyUniversity of Toronto Toronto M5S 3M2 Canada
| | - Li Zhang
- Toronto General Hospital Research InstituteUniversity Health Network Toronto M5G 2C4 Canada
- Department of ImmunologyUniversity of Toronto Toronto M5S 1A8 Canada
- Department of Laboratory Medicine and PathobiologyUniversity of Toronto Toronto M5S 1A8 Canada
| | - Gang Zheng
- Department of Medical BiophysicsUniversity of Toronto Toronto M5G 1L7 Canada
- Princess Margaret Cancer CenterUniversity Health Network Toronto M5G 2C1 Canada
- Centre for Pharmaceutical OncologyUniversity of Toronto Toronto M5S 3M2 Canada
| |
Collapse
|
19
|
Shao Q, O'Flanagan S, Lam T, Roy P, Pelaez F, Burbach BJ, Azarin SM, Shimizu Y, Bischof JC. Engineering T cell response to cancer antigens by choice of focal therapeutic conditions. Int J Hyperthermia 2019; 36:130-138. [DOI: 10.1080/02656736.2018.1539253] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Qi Shao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Stephen O'Flanagan
- Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Tiffany Lam
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Priyatanu Roy
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Francisco Pelaez
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Brandon J Burbach
- Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Yoji Shimizu
- Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
20
|
Zhao Y, Shuen TWH, Toh TB, Chan XY, Liu M, Tan SY, Fan Y, Yang H, Lyer SG, Bonney GK, Loh E, Chang KTE, Tan TC, Zhai W, Chan JKY, Chow EKH, Chee CE, Lee GH, Dan YY, Chow PKH, Toh HC, Lim SG, Chen Q. Development of a new patient-derived xenograft humanised mouse model to study human-specific tumour microenvironment and immunotherapy. Gut 2018; 67:1845-1854. [PMID: 29602780 PMCID: PMC6145285 DOI: 10.1136/gutjnl-2017-315201] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 03/07/2018] [Accepted: 03/17/2018] [Indexed: 12/27/2022]
Abstract
OBJECTIVE As the current therapeutic strategies for human hepatocellular carcinoma (HCC) have been proven to have limited effectiveness, immunotherapy becomes a compelling way to tackle the disease. We aim to provide humanised mouse (humice) models for the understanding of the interaction between human cancer and immune system, particularly for human-specific drug testing. DESIGN Patient-derived xenograft tumours are established with type I human leucocyte antigen matched human immune system in NOD-scid Il2rg-/- (NSG) mice. The longitudinal changes of the tumour and immune responses as well as the efficacy of immune checkpoint inhibitors are investigated. RESULTS Similar to the clinical outcomes, the human immune system in our model is educated by the tumour and exhibits exhaustion phenotypes such as a significant declination of leucocyte numbers, upregulation of exhaustion markers and decreased the production of human proinflammatory cytokines. Notably, cytotoxic immune cells decreased more rapidly compared with other cell types. Tumour infiltrated T cells have much higher expression of exhaustion markers and lower cytokine production compared with peripheral T cells. In addition, tumour-associated macrophages and myeloid-derived suppressor cells are found to be highly enriched in the tumour microenvironment. Interestingly, the tumour also changes gene expression profiles in response to immune responses by upregulating immune checkpoint ligands. Most importantly, in contrast to the NSG model, our model demonstrates both therapeutic and side effects of immune checkpoint inhibitors pembrolizumab and ipilimumab. CONCLUSIONS Our work provides a model for immune-oncology study and a useful parallel-to-human platform for anti-HCC drug testing, especially immunotherapy.
Collapse
Affiliation(s)
- Yue Zhao
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | | | - Tan Boon Toh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xue Ying Chan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Min Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sue Yee Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hechuan Yang
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shridhar Ganpathi Lyer
- Division of Hepatobiliary and Liver Transplantation Surgery, National University Health System, Singapore
| | - Glenn Kunnath Bonney
- Division of Hepatobiliary and Liver Transplantation Surgery, National University Health System, Singapore
| | - Eva Loh
- Department of Pathology and Laboratory Medicine, KK Women’s and Children’s Hospital, Singapore
| | - Kenneth Tou En Chang
- Department of Pathology and Laboratory Medicine, KK Women’s and Children’s Hospital, Singapore
| | - Thiam Chye Tan
- Department of Obstetrics and Gynaecology, KK Women’s and Children’s Hospital, Singapore
| | - Weiwei Zhai
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, Singapore
- Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Cheng Ean Chee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Guan Huei Lee
- Division of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
| | - Yock Young Dan
- Division of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
| | - Pierce Kah-Hoe Chow
- Division of Surgical Oncology, National Cancer Center Singapore, Singapore
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, Singapore General Hospital, Singapore
- Duke-NUS Graduate Medical School, Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Seng Gee Lim
- Division of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
21
|
Nanomaterials for modulating innate immune cells in cancer immunotherapy. Asian J Pharm Sci 2018; 14:16-29. [PMID: 32104435 PMCID: PMC7032173 DOI: 10.1016/j.ajps.2018.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/24/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy has been intensively investigated in both preclinical and clinical studies. Whereas chemotherapies use cytotoxic drugs to kill tumor cells, cancer immunotherapy is based on the ability of the immune system to fight cancer. Tumors are intimately associated with the immune system: they can suppress the immune response and/or control immune cells to support tumor growth. Immunotherapy has yielded promising results in clinical practice, but some patients show limited responses. This may reflect the complexities of the relationship between a tumor and the immune system. In an effort to improve the current immunotherapies, researchers have exploited nanomaterials in creating new strategies to cure tumors via modulation of the immune system in tumor tissues. Although extensive studies have examined the use of immune checkpoint-based immunotherapy, rather less work has focused on manipulating the innate immune cells. This review examines the recent approaches and challenges in the use of nanomaterials to modulate innate immune cells.
Collapse
|
22
|
Buqué A, Galluzzi L. Modeling Tumor Immunology and Immunotherapy in Mice. Trends Cancer 2018; 4:599-601. [PMID: 30149876 DOI: 10.1016/j.trecan.2018.07.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 07/09/2018] [Indexed: 10/28/2022]
Abstract
Figure 1. Main Applications of Mouse Models for Tumor Immunology and Immunotherapy. Immunodeficient mice xenografted with human cancer cell lines have been at the foundation of in vivo cancer research for several decades, providing ground for the regulatory approval of multiple chemotherapeutics and targeted anticancer agents, but are intrinsically unsuitable for studying tumor immunology and immunotherapy. Similarly, patient-derived xenografts (PDXs) established in immunodeficient mice are not subjected to immunosurveillance by the host, although (depending on the protocol employed for PDX generation) some components of the patient's immune system may also be transferred to the mouse and be active, at least for some time. Considerable efforts are being devoted to the generation of humanized mice to circumvent these limitations. The establishment of PDXs in immunodeficient mice that are also engrafted with matched patient-derived peripheral blood mononuclear cells (PBMCs) is also being investigated as a means to screen for the efficacy of (immuno)therapeutic agents in support of clinical decision making. Mouse cancer cell lines grafted subcutaneously or orthotopically in immunocompetent syngeneic hosts have been instrumental for the development of a variety of immunotherapeutics, as well as for the discovery that conventional anticancer regimens, including some forms of chemotherapy and radiation therapy, can trigger tumor-targeting immune responses. Carcinogen-driven tumors established in immunocompetent versus immunodeficient animals were critical in the early days of modern tumor immunology, as they enabled the discovery of natural anticancer immunosurveillance. Moreover, they allow for investigating the immunological versus non-immunological efficacy of anticancer (immuno)therapeutics in the context of natural immunoediting, clinically relevant immunobiological heterogeneity, and high mutation load. Transgene-driven tumors have generated in-depth insights into the crosstalk between oncogenic drivers and the tumor microenvironment, in both its immunological and non-immunological components. Each of these models is associated with specific advantages and disadvantages (see Key Facts). Figure 2. Key Features of Mouse Models for Tumor Immunology and Immunotherapy. Key parameters that should be taken into careful consideration when choosing the most appropriate mouse model for the study of tumor immunology and immunotherapy include not only the immunological competence of the host (which is influenced by strain, sex, and age) and its immunological compatibility with malignant cells (which is dictated by strain), but also the mutational load of the latter, their immunological history (previous immunoediting), proliferative potential, propensity for neovascularization and metastatic dissemination, as well as their ability to generate an immunostimulatory versus immunosuppressive microenvironment. Inoculation site is also an important parameter to keep under consideration (not shown). The precise objective of each study dictates which specific combination of such features should be preferred.
Collapse
Affiliation(s)
- Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, 10065 NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, 10065 NY, USA; Sandra and Edward Meyer Cancer Center, New York, 10065 NY, USA; Université Paris Descartes/Paris V, 75006 Paris, France.
| |
Collapse
|
23
|
A novel tetrahydroisoquinoline (THIQ) analogue induces mitochondria-dependent apoptosis. Eur J Med Chem 2018; 150:719-728. [PMID: 29573707 DOI: 10.1016/j.ejmech.2018.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 12/31/2022]
Abstract
Lung cancer continues to be a leading cause of cancer-related death worldwide, with non-small cell lung cancer (NSCLC) accounting for more than 80% of lung cancer cases. Current therapies for NSCLC have only limited effect and treatment resistance develops rapidly. In a previous study, we have shown that C1-phenylethynyl tetrahydroisoquinoline (THIQ) analogue 4 has anti-proliferative activity against PC3 human prostate cancer cells. However, this anticancer effect was achieved with relatively high IC50 in A549 lung cancer cells. To improve the potency of the drug, in the present study, a series of novel THIQ analogues (analogues 5a-d) were prepared by using an oxidative C-H functionalization strategy, and their potential anticancer activities on A549 lung cancer cells were investigated. Among these analogues, analogue 5c can markedly inhibit A549 cell proliferation in a dose-dependent manner with a reasonable IC50 of 14.61 ± 1.03 μM. This effect was mediated by analogue 5c-induced G0/G1 phase arrest and cell apoptosis. Treatment with analogue 5c was shown to induce reactive oxygen species (ROS) accumulation, disruption of mitochondrial membrane potential, reduction of glutathione, elevation of intracellular calcium ion (Ca2+), and activation of Caspase-3. Furthermore, analogue 5c can lead to DNA double-strand break and the activation of p53 pathway in A549 cells. In conclusion, the oxidative C-H functionalization strategy to generate analogue 5c could improve the drug anticancer efficacy by inducing mitochondria-dependent apoptosis in A549 cells.
Collapse
|
24
|
Obermajer N, Urban J, Wieckowski E, Muthuswamy R, Ravindranathan R, Bartlett DL, Kalinski P. Promoting the accumulation of tumor-specific T cells in tumor tissues by dendritic cell vaccines and chemokine-modulating agents. Nat Protoc 2018; 13:335-357. [PMID: 29345636 DOI: 10.1038/nprot.2017.130] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This protocol describes how to induce large numbers of tumor-specific cytotoxic T cells (CTLs) in the spleens and lymph nodes of mice receiving dendritic cell (DC) vaccines and how to modulate tumor microenvironments (TMEs) to ensure effective homing of the vaccination-induced CTLs to tumor tissues. We also describe how to evaluate the numbers of tumor-specific CTLs within tumors. The protocol contains detailed information describing how to generate a specialized DC vaccine with augmented ability to induce tumor-specific CTLs. We also describe methods to modulate the production of chemokines in the TME and show how to quantify tumor-specific CTLs in the lymphoid organs and tumor tissues of mice receiving different treatments. The combined experimental procedure, including tumor implantation, DC vaccine generation, chemokine-modulating (CKM) approaches, and the analyses of tumor-specific systemic and intratumoral immunity is performed over 30-40 d. The presented ELISpot-based ex vivo CTL assay takes 6 h to set up and 5 h to develop. In contrast to other methods of evaluating tumor-specific immunity in tumor tissues, our approach allows detection of intratumoral T-cell responses to nonmanipulated weakly immunogenic cancers. This detection method can be performed using basic laboratory skills, and facilitates the development and preclinical evaluation of new immunotherapies.
Collapse
Affiliation(s)
- Nataša Obermajer
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julie Urban
- Immunotransplantation Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Eva Wieckowski
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Immunotransplantation Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - David L Bartlett
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Pawel Kalinski
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Immunotransplantation Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
25
|
Leveraging Epigenetics to Enhance the Cellular Response to Chemotherapies and Improve Tumor Immunogenicity. Adv Cancer Res 2018; 138:1-39. [PMID: 29551125 DOI: 10.1016/bs.acr.2018.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer chemotherapeutic drugs have greatly advanced our ability to successfully treat a variety of human malignancies. The different forms of stress produced by these agents in cancer cells result in both cell autonomous and cell nonautonomous effects. Desirable cell autonomous effects include reduced proliferative potential, cellular senescence, and cell death. More recently recognized cell nonautonomous effects, usually in the form of stimulating an antitumor immune response, have significant roles in therapeutic efficiency for a select number of chemotherapies. Unfortunately, the success of these therapeutics is not universal as not all tumors respond to treatment, and those that do respond will frequently relapse into therapy-resistant disease. Numerous strategies have been developed to sensitize tumors toward chemotherapies as a means to either improve initial responses, or serve as a secondary treatment strategy for therapy-resistant disease. Recently, targeting epigenetic regulators has emerged as a viable method of sensitizing tumors to the effects of chemotherapies, many of which are cytotoxic. In this review, we summarize these strategies and propose a path for future progress.
Collapse
|
26
|
Carpenter DJ, Granot T, Matsuoka N, Senda T, Kumar BV, Thome JJC, Gordon CL, Miron M, Weiner J, Connors T, Lerner H, Friedman A, Kato T, Griesemer AD, Farber DL. Human immunology studies using organ donors: Impact of clinical variations on immune parameters in tissues and circulation. Am J Transplant 2018; 18:74-88. [PMID: 28719147 PMCID: PMC5740015 DOI: 10.1111/ajt.14434] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/23/2017] [Accepted: 07/11/2017] [Indexed: 01/25/2023]
Abstract
Organ donors are sources of physiologically healthy organs and tissues for life-saving transplantation, and have been recently used for human immunology studies which are typically confined to the sampling of peripheral blood. Donors comprise a diverse population with different causes of death and clinical outcomes during hospitalization, and the effects of such variations on immune parameters in blood and tissues are not known. We present here a coordinate analysis of innate and adaptive immune components in blood, lymphoid (bone marrow, spleen, lymph nodes), and mucosal (lungs, intestines) sites from a population of brain-dead organ donors (2 months-93 years; n = 291) across eight clinical parameters. Overall, the blood of donors exhibited similar monocyte and lymphocyte content and low serum levels of pro-inflammatory cytokines as healthy controls; however, donor blood had increased neutrophils and serum levels of IL-8, IL-6, and MCP-1 which varied with cause of death. In tissues, the frequency and composition of monocytes, neutrophils, B lymphocytes and T cell subsets in lymphoid or mucosal sites did not vary with clinical state, and was similar in donors independent of the extent of clinical complications. Our results reveal that organ donors maintain tissue homeostasis, and are a valuable resource for fundamental studies in human immunology.
Collapse
Affiliation(s)
- D J Carpenter
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - T Granot
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - N Matsuoka
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - T Senda
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - B V Kumar
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, USA
| | - J J C Thome
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, USA
| | - C L Gordon
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - M Miron
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, USA
| | - J Weiner
- Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - T Connors
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | | | | | - T Kato
- Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - A D Griesemer
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - D L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Medical Center, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
27
|
Hochnadel I, Kossatz-Boehlert U, Jedicke N, Lenzen H, Manns MP, Yevsa T. Cancer vaccines and immunotherapeutic approaches in hepatobiliary and pancreatic cancers. Hum Vaccin Immunother 2017; 13:2931-2952. [PMID: 29112462 PMCID: PMC5718787 DOI: 10.1080/21645515.2017.1359362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatobiliary and pancreatic cancers along with other gastrointestinal malignancies remain the leading cause of cancer-related deaths worldwide. Strategies developed in the recent years on immunotherapy and cancer vaccines in the setting of primary liver cancer as well as in pancreatic cancer are the scope of this review. Significance of orthotopic and autochthonous animal models which mimic and/or closely reflect human malignancies allowing for a prompt and trustworthy analysis of new therapeutics is underlined. Combinational approaches that on one hand, specifically target a defined cancer-driving pathway, and on the other hand, restore the functions of immune cells, which effector functions are often suppressed by a tumor milieu, are shown to have the strongest perspectives and future directions. Among combinational immunotherapeutic approaches a personalized- and individual cancer case-based therapy is of special importance.
Collapse
Affiliation(s)
- Inga Hochnadel
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Uta Kossatz-Boehlert
- b Institute for Neuroanatomy, Eberhard-Karls University Tuebingen , Tuebingen , Germany
| | - Nils Jedicke
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Henrike Lenzen
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Michael P Manns
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Tetyana Yevsa
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| |
Collapse
|
28
|
VAN Noord RA, Thomas T, Krook M, Chukkapalli S, Hoenerhoff MJ, Dillman JR, Lawlor ER, Opipari VP, Newman EA. Tissue-directed Implantation Using Ultrasound Visualization for Development of Biologically Relevant Metastatic Tumor Xenografts. ACTA ACUST UNITED AC 2017; 31:779-791. [PMID: 28882943 DOI: 10.21873/invivo.11131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Advances in cancer therapeutics depend on reliable in vivo model systems. To develop biologically relevant xenografts, ultrasound was utilized for tissue-directed implantation of neuroblastoma (NB) cell line and patient-derived tumors in the adrenal gland, and for renal subcapsular engraftment of Ewing's sarcoma (ES). MATERIALS AND METHODS NB xenografts were established by direct adrenal injection of luciferase-transfected NB cell lines (IMR32, SH-SY5Y, SK-N-BE2) or NB patient-derived tumor cells (UMNBL001, UMNBL002). ES xenografts were established by renal subcapsular injection of TC32, A673, CHLA-25, or A4573 cells. Progression was monitored by in vivo imaging. RESULTS Tumors progressed to local disease with metastasis evident by 5 weeks. Metastatic sites included cortical bone, lung, liver, and lymph nodes. Xenografted tumors retained immunochemical features of the original cancer. CONCLUSION Human NB adrenal xenografts, including two patient-derived orthotopic, and ES renal subcapsular xenografts were established by ultrasound without open surgery. Tissue-directed implantation is an effective technique for developing metastatic preclinical models.
Collapse
Affiliation(s)
- Raelene A VAN Noord
- Department of Surgery, C.S Mott Children's and Women's Hospital, Mott Solid Tumor Oncology Program, The University of Michigan Medical School, Ann Arbor, MI, U.S.A
| | - Tina Thomas
- Department of Surgery, C.S Mott Children's and Women's Hospital, Mott Solid Tumor Oncology Program, The University of Michigan Medical School, Ann Arbor, MI, U.S.A
| | - Melanie Krook
- Department of Pathology, C.S Mott Children's and Women's Hospital, Mott Solid Tumor Oncology Program, The University of Michigan Medical School, Ann Arbor, MI, U.S.A
| | - Sahiti Chukkapalli
- Department of Surgery, C.S Mott Children's and Women's Hospital, Mott Solid Tumor Oncology Program, The University of Michigan Medical School, Ann Arbor, MI, U.S.A
| | - Mark J Hoenerhoff
- Unit for Laboratory Animal Medicine, The University of Michigan Medical School, Ann Arbor, MI, U.S.A
| | - Jonathan R Dillman
- Department of Radiology, C.S Mott Children's and Women's Hospital, Mott Solid Tumor Oncology Program, The University of Michigan Medical School, Ann Arbor, MI, U.S.A
| | - Elizabeth R Lawlor
- Department of Pathology, C.S Mott Children's and Women's Hospital, Mott Solid Tumor Oncology Program, The University of Michigan Medical School, Ann Arbor, MI, U.S.A.,Department of Pediatrics, C.S Mott Children's and Women's Hospital, Mott Solid Tumor Oncology Program, The University of Michigan Medical School, Ann Arbor, MI, U.S.A
| | - Valerie P Opipari
- Department of Pediatrics, C.S Mott Children's and Women's Hospital, Mott Solid Tumor Oncology Program, The University of Michigan Medical School, Ann Arbor, MI, U.S.A
| | - Erika A Newman
- Department of Surgery, C.S Mott Children's and Women's Hospital, Mott Solid Tumor Oncology Program, The University of Michigan Medical School, Ann Arbor, MI, U.S.A.
| |
Collapse
|
29
|
Abstract
Immuno-oncology (I/O) research has intensified significantly in recent years due to the breakthrough development and the regulatory approval of several immune checkpoint inhibitors, leading to the rapid expansion of the new discovery of novel I/O therapies, new checkpoint inhibitors and beyond. However, many I/O questions remain unanswered, including why only certain subsets of patients respond to these treatments, who the responders would be, and how to expand patient response (the conversion of non-responders or maximizing response in partial responders). All of these require relevant I/O experimental systems, particularly relevant preclinical animal models. Compared to other oncology drug discovery, e.g. cytotoxic and targeted drugs, a lack of relevant animal models is a major obstacle in I/O drug discovery, and an urgent and unmet need. Despite the obvious importance, and the fact that much I/O research has been performed using many different animal models, there are few comprehensive and introductory reviews on this topic. This article attempts to review the efforts in development of a variety of such models, as well as their applications and limitations for readers new to the field, particularly those in the pharmaceutical industry.
Collapse
Affiliation(s)
- Qi-Xiang Li
- Crown Bioscience Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA 95054, USA; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| | - Gerold Feuer
- HuMurine Technologies, Inc., 2700 Stockton Blvd, Rm. 1403, Sacramento, CA 95817, USA
| | - Xuesong Ouyang
- Crown Bioscience Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA 95054, USA
| | - Xiaoyu An
- Crown Bioscience Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA 95054, USA; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| |
Collapse
|
30
|
Abstract
Fundamental cancer research and the development of efficacious antineoplastic treatments both rely on experimental systems in which the relationship between malignant cells and immune cells can be studied. Mouse models of transplantable, carcinogen-induced or genetically engineered malignancies - each with their specific advantages and difficulties - have laid the foundations of oncoimmunology. These models have guided the immunosurveillance theory that postulates that evasion from immune control is an essential feature of cancer, the concept that the long-term effects of conventional cancer treatments mostly rely on the reinstatement of anticancer immune responses and the preclinical development of immunotherapies, including currently approved immune checkpoint blockers. Specific aspects of pharmacological development, as well as attempts to personalize cancer treatments using patient-derived xenografts, require the development of mouse models in which murine genes and cells are replaced with their human equivalents. Such 'humanized' mouse models are being progressively refined to characterize the leukocyte subpopulations that belong to the innate and acquired arms of the immune system as they infiltrate human cancers that are subjected to experimental therapies. We surmise that the ever-advancing refinement of murine preclinical models will accelerate the pace of therapeutic optimization in patients.
Collapse
Affiliation(s)
- Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), INSERM U1015, 114 rue Edouard Vaillant, 94805 Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer, CICBT1428, GRCC, 94805 Villejuif, France
| | - Jonathan M Pitt
- Gustave Roussy Cancer Campus (GRCC), INSERM U1015, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | - Romain Daillère
- Gustave Roussy Cancer Campus (GRCC), INSERM U1015, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; University of Queensland, Herston, QLD, Australia
| | - Guido Kroemer
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France
- University of Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
- University of Pierre et Marie Curie, 75006 Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, 75015 Paris, France
- Metabolomics and Cell Biology Platforms, GRCC, 94805 Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| |
Collapse
|