51
|
Yang J, Zhang W, Jia L, Shi F, Cao M, Sun J, Xu C, Li Z, Cheng Z, Zhao S, Wang J, Yang C. A novel autophagy activator ginsenoside Rh2 enhances the efficacy of immunogenic chemotherapy. Clin Transl Med 2023; 13:e1109. [PMID: 36732082 PMCID: PMC9894730 DOI: 10.1002/ctm2.1109] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 02/04/2023] Open
Affiliation(s)
- Jing Yang
- Department of NephrologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Wei Zhang
- Department of NephrologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Lin Jia
- College of PharmacyShenzhen Technology UniversityShenzhenChina
| | - Fei Shi
- Emergency Department, Institute of Shenzhen Respiratory DiseasesShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Min Cao
- Department of NephrologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Jichao Sun
- Department of NephrologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Chengchao Xu
- Department of NephrologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Zhijie Li
- Department of NephrologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Zhiqiang Cheng
- Department of PathologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Shan‐Chao Zhao
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
- Department of UrologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Jigang Wang
- Department of NephrologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
- Artemisinin Research Center, Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Chuanbin Yang
- Department of NephrologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| |
Collapse
|
52
|
Mondal P, Patel NS, Bailey K, Aravind S, Cartwright SB, Hollingsworth MA, Lazenby AJ, Carlson MA. Induction of pancreatic neoplasia in the KRAS/TP53 Oncopig. Dis Model Mech 2023; 16:286617. [PMID: 36579622 PMCID: PMC9884120 DOI: 10.1242/dmm.049699] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/13/2022] [Indexed: 12/30/2022] Open
Abstract
The 5-year survival of pancreatic cancer (PC) remains low. Murine models may not adequately mimic human PC and can be too small for medical device development. A large-animal PC model could address these issues. We induced and characterized pancreatic tumors in Oncopigs (transgenic swine containing KRASG12D and TP53R167H). The oncopigs underwent injection of adenovirus expressing Cre recombinase (AdCre) into one of the main pancreatic ducts. Resultant tumors were characterized by histology, cytokine expression, exome sequencing and transcriptome analysis. Ten of 14 Oncopigs (71%) had gross tumor within 3 weeks. At necropsy, all of these subjects had gastric outlet obstruction secondary to pancreatic tumor and phlegmon. Oncopigs with injections without Cre recombinase and wild-type pigs with AdCre injection did not show notable effect. Exome and transcriptome analysis of the porcine pancreatic tumors revealed similarity to the molecular signatures and pathways of human PC. Although further optimization and validation of this porcine PC model would be beneficial, it is anticipated that this model will be useful for focused research and development of diagnostic and therapeutic technologies for PC. This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Pinaki Mondal
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Neesha S. Patel
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Katie Bailey
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Shruthishree Aravind
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Sara B. Cartwright
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Audrey J. Lazenby
- Department of Pathology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mark A. Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA,Author for correspondence ()
| |
Collapse
|
53
|
Yu R, Maswikiti EP, Yu Y, Gao L, Ma C, Ma H, Deng X, Wang N, Wang B, Chen H. Advances in the Application of Preclinical Models in Photodynamic Therapy for Tumor: A Narrative Review. Pharmaceutics 2023; 15:pharmaceutics15010197. [PMID: 36678826 PMCID: PMC9867105 DOI: 10.3390/pharmaceutics15010197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/09/2023] Open
Abstract
Photodynamic therapy (PDT) is a non-invasive laser light local treatment that has been utilized in the management of a wide variety of solid tumors. Moreover, the evaluation of efficacy, adverse reactions, the development of new photosensitizers and the latest therapeutic regimens are inseparable from the preliminary exploration in preclinical studies. Therefore, our aim was to better comprehend the characteristics and limitations of these models and to provide a reference for related research. METHODS We searched the databases, including PubMed, Web of Science and Scopus for the past 25 years of original research articles on the feasibility of PDT in tumor treatment based on preclinical experiments and animal models. We provided insights into inclusion and exclusion criteria and ultimately selected 40 articles for data synthesis. RESULTS After summarizing and comparing the methods and results of these studies, the experimental model selection map was drawn. There are 7 main preclinical models, which are used for different research objectives according to their characteristics. CONCLUSIONS Based on this narrative review, preclinical experimental models are crucial to the development and promotion of PDT for tumors. The traditional animal models have some limitations, and the emergence of organoids may be a promising new insight.
Collapse
Affiliation(s)
- Rong Yu
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | | | - Yang Yu
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Lei Gao
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Chenhui Ma
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Huanhuan Ma
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Xiaobo Deng
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Na Wang
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Bofang Wang
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Hao Chen
- Department of Surgical Oncology, Second Hospital of Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Digestive System Tumor of Gansu Province, Second Hospital of Lanzhou University, Lanzhou 730030, China
- Correspondence: ; Tel.: +86-0931-5190550
| |
Collapse
|
54
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 174:xv-xxv. [PMID: 37039770 DOI: 10.1016/s0091-679x(23)00036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
55
|
Sun LY, Zhang KJ, Xie YM, Liu JW, Xiao ZQ. Immunotherapies for advanced hepatocellular carcinoma. Front Pharmacol 2023; 14:1138493. [PMID: 37025485 PMCID: PMC10070708 DOI: 10.3389/fphar.2023.1138493] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Primary liver cancer is the second leading cause of tumor-related deaths in China, with hepatocellular carcinoma (HCC) accounting for 80%-90% of these. Since there is a lack of symptoms in the early stages of HCC, a large proportion of patients were identified with unresectable HCC when diagnosed. Due to the severe resistance to chemotherapy, patients with advanced HCC were traditionally treated with systematic therapy in the past decades, and the tyrosine kinase inhibitor (TKI) sorafenib has remained the only treatment option for advanced HCC since 2008. Immunotherapies, particularly immune checkpoint inhibitors (ICIs), have shown a strong anti-tumor effect and have been supported by several guidelines recently. ICIs, for example programmed cell death-1 (PD-1) inhibitors such as nivolumab and pembrolizumab, programmed cell death ligand 1 (PD-L1) inhibitors such as atezolizumab, and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors such as ipilimumab, the ICI-based combination with TKIs, and VEGF-neutralizing antibody or systematic or local anti-tumor therapies, are being further studied in clinical trials. However, immune-related adverse events (irAEs) including cutaneous toxicity, gastrointestinal toxicity, and hepatotoxicity may lead to the termination of ICI treatment or even threaten patients' lives. This review aims to summarize currently available immunotherapies and introduce the irAEs and their managements in order to provide references for clinical application and further research.
Collapse
Affiliation(s)
- Li-Yang Sun
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Cancer Center, General Surgery, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kang-Jun Zhang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Cancer Center, General Surgery, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ya-Ming Xie
- Cancer Center, General Surgery, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jun-Wei Liu
- Cancer Center, General Surgery, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- *Correspondence: Jun-Wei Liu, ; Zun-Qiang Xiao,
| | - Zun-Qiang Xiao
- Cancer Center, General Surgery, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- *Correspondence: Jun-Wei Liu, ; Zun-Qiang Xiao,
| |
Collapse
|
56
|
Mirjolet C, Baude J, Galluzzi L. Dual impact of radiation therapy on tumor-targeting immune responses. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:xiii-xxiv. [PMID: 37438022 DOI: 10.1016/s1937-6448(23)00114-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Affiliation(s)
- Céline Mirjolet
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, GF Leclerc Centre, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France.
| | - Jérémy Baude
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, GF Leclerc Centre, Unicancer, Dijon, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
57
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 180:xv-xxv. [PMID: 37890936 DOI: 10.1016/s0091-679x(23)00166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Affiliation(s)
- Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Jeffrey Kraynak
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Ariel E Marciscano
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
58
|
Xu L, Li B, Pi C, Zhu Z, Tao F, Xie K, Feng Y, Xu X, Yin Y, Gu H, Fang J. Targeting CD89 on tumor-associated macrophages overcomes resistance to immune checkpoint blockade. J Immunother Cancer 2022; 10:jitc-2022-005447. [PMID: 36460336 PMCID: PMC9723960 DOI: 10.1136/jitc-2022-005447] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Despite the survival benefits observed with immune checkpoint blockade (ICB) treatment-programmed cell death-1/programmed cell death ligand-1 (PD-1/PD-L1), many patients with cancer have not benefited from these agents because of impaired antigen presentation and other resistance mechanisms. To overcome resistance to checkpoint therapy, we designed bispecific antibodies (BsAbs) targeting CD89 and tumor antigens. We demonstrated their immunomodulatory efficacy as a separate treatment strategy or combined with immune checkpoint inhibitors. METHODS We have previously generated a heterodimeric one-arm IgG1 Fc-based bispecific antibody. For animal efficacy studies, murine tumors in a humanized transgenic mice model were used to determine the effects of CD89-bispecific antibodies on antigen presentation and immune cell recruitment. The efficacy of the CD89 bispecific antibody against tumors resistant to pembrolizumab was evaluated in double-transgenic mice. RESULTS BsAbs targeting CD89 on tumor-associated macrophages (TAMs) increased the ratio of M1:M2 and activated the antigen presentation, leading to increased cytotoxic T cell-mediated tumor regression. CD89-BsAbs also potentiated a combinational antitumor effect with PD-1/PD-L1 inhibitors and overcame the ICB resistance by augmenting cytotoxic T-cell infiltration and reshaping tumor immune microenvironment. In an hCD89/hPD-1 double transgenic mouse model engrafted with pembrolizumab-resistant B16-HER2 tumor cells, the HER2-CD89 bispecific antibody potently inhibited tumor growth. CONCLUSIONS CD89 BsAbs targeting tumor antigens and TAMs controlled tumor growth in animal models by improving antigen presentation and T-cell infiltration. Our results suggest a general strategy for overcoming resistance to ICB.
Collapse
Affiliation(s)
- Lijun Xu
- Laboratory of Molecular Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China,College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Bingyu Li
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Chenyu Pi
- Laboratory of Molecular Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhaohua Zhu
- Laboratory of Molecular Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Fei Tao
- Laboratory of Molecular Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Kun Xie
- Laboratory of Molecular Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yan Feng
- Laboratory of Molecular Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoqing Xu
- Laboratory of Molecular Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yanxin Yin
- Tongji University Suzhou Institute, Suzhou, Jiangsu, China
| | - Hua Gu
- Laboratory of Molecular Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jianmin Fang
- Laboratory of Molecular Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China,Tongji Hospital Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
59
|
Mechanisms of Resistance and Strategies to Combat Resistance in PD-(L)1 Blockade. IMMUNO 2022. [DOI: 10.3390/immuno2040041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Prolonged survival and durable responses in several late-stage cancers such as melanoma and lung cancer have been made possible with the use of immune checkpoint inhibitors targeting the programmed cell-death protein 1 (PD-1) or its ligand PD-L1. While it is prudent to focus on the unprecedented and durable clinical responses, there are subsets of cancer patients that do not respond to immunotherapies or respond early and then relapse later. Many pathways of resistance have been characterized, and more continue to be uncovered. To overcome the development of resistance, an in-depth investigation is necessary to identify alternative immune receptors and signals with the overarching goal of expanding treatment options for those with demonstrated resistance to PD1 checkpoint immunotherapy. In this mini-review, we will discuss the mechanisms by which tumors exhibit resistance to anti-PD-1/PD-L1 immunotherapy and explore strategies to overcome such resistances.
Collapse
|
60
|
Budhwani KI, Patel ZH, Guenter RE, Charania AA. A hitchhiker's guide to cancer models. Trends Biotechnol 2022; 40:1361-1373. [PMID: 35534320 PMCID: PMC9588514 DOI: 10.1016/j.tibtech.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/31/2022] [Accepted: 04/08/2022] [Indexed: 01/21/2023]
Abstract
Cancer is a complex and uniquely personal disease. More than 1.7 million people in the United States are diagnosed with cancer every year. As the burden of cancer grows, so does the need for new, more effective therapeutics and for predictive tools to identify optimal, personalized treatment options for every patient. Cancer models that recapitulate various aspects of the disease are fundamental to making advances along the continuum of cancer treatment from benchside discoveries to bedside delivery. In this review, we use a thought experiment as a vehicle to arrive at four broad categories of cancer models and explore the strengths, weaknesses, opportunities, and threats for each category in advancing our understanding of the disease and improving treatment strategies.
Collapse
Affiliation(s)
- Karim I Budhwani
- CerFlux, Inc., Birmingham, AL, USA; Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; Department of Physics, Coe College, Cedar Rapids, IA, USA.
| | | | | | | |
Collapse
|
61
|
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: 1.0] [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
|
62
|
Liu J, Yu Y, Liu C, Gao C, Zhuang J, Liu L, Wu Q, Ma W, Zhang Q, Sun C. Combinatorial regimens of chemotherapeutic agents: A new perspective on raising the heat of the tumor immune microenvironment. Front Pharmacol 2022; 13:1035954. [PMID: 36304169 PMCID: PMC9593050 DOI: 10.3389/fphar.2022.1035954] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Harnessing the broad immunostimulatory capabilities of chemotherapy in combination with immune checkpoint inhibitors has improved immunotherapy outcomes in patients with cancer. Certain chemotherapeutic agents can extensively modify the tumor microenvironment (TME), resulting in the reprogramming of local immune responses. Although chemotherapeutic agents with an enhanced generation of potent anti-tumor immune responses have been tested in preclinical animal models and clinical trials, this strategy has not yet shown substantial therapeutic efficacy in selected difficult-to-treat cancer types. In addition, the efficacy of chemotherapeutic agent-based monotherapy in eliciting a long-term anti-tumor immune response is restricted by the immunosuppressive TME. To enhance the immunomodulatory effect of chemotherapy, researchers have made many attempts, mainly focusing on improving the targeted distribution of chemotherapeutic agents and designing combination therapies. Here, we focused on the mechanisms of the anti-tumor immune response to chemotherapeutic agents and enumerated the attempts to advance the use of chemo-immunotherapy. Furthermore, we have listed the important considerations in designing combinations of these drugs to maximize efficacy and improve treatment response rates in patients with cancer.
Collapse
Affiliation(s)
- Jingyang Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Qiming Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Qiming Zhang, ; Changgang Sun,
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
- *Correspondence: Qiming Zhang, ; Changgang Sun,
| |
Collapse
|
63
|
Zanella ER, Grassi E, Trusolino L. Towards precision oncology with patient-derived xenografts. Nat Rev Clin Oncol 2022; 19:719-732. [PMID: 36151307 DOI: 10.1038/s41571-022-00682-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2022] [Indexed: 11/09/2022]
Abstract
Under the selective pressure of therapy, tumours dynamically evolve multiple adaptive mechanisms that make static interrogation of genomic alterations insufficient to guide treatment decisions. Clinical research does not enable the assessment of how various regulatory circuits in tumours are affected by therapeutic insults over time and space. Likewise, testing different precision oncology approaches informed by composite and ever-changing molecular information is hard to achieve in patients. Therefore, preclinical models that incorporate the biology and genetics of human cancers, facilitate analyses of complex variables and enable adequate population throughput are needed to pinpoint randomly distributed response predictors. Patient-derived xenograft (PDX) models are dynamic entities in which cancer evolution can be monitored through serial propagation in mice. PDX models can also recapitulate interpatient diversity, thus enabling the identification of response biomarkers and therapeutic targets for molecularly defined tumour subgroups. In this Review, we discuss examples from the past decade of the use of PDX models for precision oncology, from translational research to drug discovery. We elaborate on how and to what extent preclinical observations in PDX models have confirmed and/or anticipated findings in patients. Finally, we illustrate emerging methodological efforts that could broaden the application of PDX models by honing their predictive accuracy or improving their versatility.
Collapse
Affiliation(s)
| | - Elena Grassi
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Italy.,Department of Oncology, University of Torino, Candiolo, Italy
| | - Livio Trusolino
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Italy. .,Department of Oncology, University of Torino, Candiolo, Italy.
| |
Collapse
|
64
|
Sun Q, Wang Y, Ji H, Sun X, Xie S, Chen L, Li S, Zeng W, Chen R, Tang Q, Zuo J, Hou L, Hosaka K, Lu Y, Liu Y, Ye Y, Yang Y. Lenvatinib for effectively treating antiangiogenic drug-resistant nasopharyngeal carcinoma. Cell Death Dis 2022; 13:724. [PMID: 35985991 PMCID: PMC9391381 DOI: 10.1038/s41419-022-05171-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/30/2022] [Accepted: 08/08/2022] [Indexed: 01/21/2023]
Abstract
Nasopharyngeal carcinoma (NPC) clinical trials show that antiangiogenic drugs (AADs) fail to achieve the expected efficacy, and combining AAD with chemoradiotherapy does not show superiority over chemoradiotherapy alone. Accumulating evidence suggests the intrinsic AAD resistance in NPC patients with poorly understood molecular mechanisms. Here, we describe NPC-specific FGF-2 expression-triggered, VEGF-independent angiogenesis as a mechanism of AAD resistance. Angiogenic factors screening between AAD-sensitive cancer type and AAD-resistant NPC showed high FGF-2 expression in NPC in both xenograft models and clinical samples. Mechanistically, the FGF-2-FGFR1-MYC axis drove endothelial cell survival and proliferation as an alternative to VEGF-VEGFR2-MYC signaling. Genetic knockdown of FGF-2 in NPC tumor cells reduced tumor angiogenesis, enhanced AAD sensitivity, and reduced pulmonary metastasis. Moreover, lenvatinib, an FDA recently approved multi-kinase inhibitor targeting both VEGFR2 and FGFR1, effectively inhibits the tumor vasculature, and exhibited robust anti-tumor effects in NPC-bearing nude mice and humanized mice compared with an agent equivalent to bevacizumab. These findings provide mechanistic insights on FGF-2 signaling in the modulation of VEGF pathway activation in the NPC microenvironment and propose an effective NPC-targeted therapy by using a clinically available drug.
Collapse
Affiliation(s)
- Qi Sun
- grid.8547.e0000 0001 0125 2443Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Yujie Wang
- grid.452847.80000 0004 6068 028XDepartment of Otolaryngology, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, 518035 Shenzhen, Guangdong China
| | - Hong Ji
- grid.452509.f0000 0004 1764 4566Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu China
| | - Xiaoting Sun
- grid.8547.e0000 0001 0125 2443Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China ,grid.4714.60000 0004 1937 0626Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden ,grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vison and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P. R. China
| | - Sisi Xie
- grid.8547.e0000 0001 0125 2443Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China ,grid.256112.30000 0004 1797 9307Longyan First Hospital Affiliated to Fujian Medical University, 364000 Longyan, Fujian China
| | - Longtian Chen
- grid.256112.30000 0004 1797 9307Longyan First Hospital Affiliated to Fujian Medical University, 364000 Longyan, Fujian China
| | - Sen Li
- grid.8547.e0000 0001 0125 2443Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Weifan Zeng
- grid.8547.e0000 0001 0125 2443Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Ruibo Chen
- grid.8547.e0000 0001 0125 2443Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Qi Tang
- grid.8547.e0000 0001 0125 2443Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Ji Zuo
- grid.8547.e0000 0001 0125 2443Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| | - Likun Hou
- grid.412532.3Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P. R. China
| | - Kayoko Hosaka
- grid.4714.60000 0004 1937 0626Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yongtian Lu
- grid.452847.80000 0004 6068 028XDepartment of Otolaryngology, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, 518035 Shenzhen, Guangdong China
| | - Ying Liu
- grid.39436.3b0000 0001 2323 5732Institute of Translational Medicine, Shanghai University, 99 Shangda Road, 200444 Shanghai, China
| | - Ying Ye
- grid.24516.340000000123704535Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yunlong Yang
- grid.8547.e0000 0001 0125 2443Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032 Shanghai, China
| |
Collapse
|
65
|
Genta S, Coburn B, Cescon DW, Spreafico A. Patient-derived cancer models: Valuable platforms for anticancer drug testing. Front Oncol 2022; 12:976065. [PMID: 36033445 PMCID: PMC9413077 DOI: 10.3389/fonc.2022.976065] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Molecularly targeted treatments and immunotherapy are cornerstones in oncology, with demonstrated efficacy across different tumor types. Nevertheless, the overwhelming majority metastatic disease is incurable due to the onset of drug resistance. Preclinical models including genetically engineered mouse models, patient-derived xenografts and two- and three-dimensional cell cultures have emerged as a useful resource to study mechanisms of cancer progression and predict efficacy of anticancer drugs. However, variables including tumor heterogeneity and the complexities of the microenvironment can impair the faithfulness of these platforms. Here, we will discuss advantages and limitations of these preclinical models, their applicability for drug testing and in co-clinical trials and potential strategies to increase their reliability in predicting responsiveness to anticancer medications.
Collapse
Affiliation(s)
- Sofia Genta
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Bryan Coburn
- Division of Infectious Diseases, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - David W. Cescon
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
66
|
Li P, Rozich N, Wang J, Wang J, Xu Y, Herbst B, Yu R, Muth S, Niu N, Li K, Funes V, Gai J, Osipov A, Edil BH, Wolfgang CL, Lei M, Liang T, Zheng L. Anti-IL-8 antibody activates myeloid cells and potentiates the anti-tumor activity of anti-PD-1 antibody in the humanized pancreatic cancer murine model. Cancer Lett 2022; 539:215722. [PMID: 35533951 PMCID: PMC9485862 DOI: 10.1016/j.canlet.2022.215722] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 12/20/2022]
Abstract
Pancreatic ductal adenocarcinoma(PDAC) does not respond to single-agent immune checkpoint inhibitor therapy, including anti-PD-1 antibody(aPD-1) therapy. Higher plasma levels of IL-8 are associated with poorer outcomes in patients who receive aPD-1 therapies, providing a rationale for combination immunotherapy with an anti-IL-8 antibody(aIL-8) and aPD-1. We thus investigated whether human aIL-8 therapy can potentiate the antitumor activity of aPD-1 and further investigated how the combination affects the immune response by regulating myeloid cells in the tumor microenvironment in a humanized murine model of PDAC with a reconstituted immune system consisting of human T cells and a combination of CD14+ and CD16+ myeloid cells. The results show that the combination of aIL-8 and aPD-1 treatment significantly enhanced antitumor activity following the infusion of myeloid cells. Our results further showed that the target of IL-8 is mainly present in CD16+ myeloid cells and is likely to be granulocytes. FACS analysis showed that aIL-8 treatment increased granulocytic myeloid cells in tumors. Consistently, single-nuclear RNA-sequencing analysis of tumor tissue showed that the innate immune response and cytokine response pathways in the myeloid cell cluster were activated by aIL-8 treatment. This is the first preclinical study using a humanized mouse model for new combination immunotherapeutic development and supports the further clinical testing of aIL-8 in combination with aPD-1 for PDAC treatment. This study also suggests that peripherally derived myeloid cells can potentiate the antitumor response of T cells, likely through the innate immune response, and aIL-8 re-educates tumor-infiltrating myeloid cells by activating the innate immune response.
Collapse
Affiliation(s)
- Pan Li
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Noah Rozich
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA; University of Oklahoma, Oklahoma City, OK, USA
| | - Jianxin Wang
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junke Wang
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yao Xu
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian Herbst
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raymond Yu
- NovaRock, Biotherapeutics Ltd., Ewing, NJ, USA
| | - Stephen Muth
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nan Niu
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Keyu Li
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vanessa Funes
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica Gai
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arsen Osipov
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Cedar-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Christopher L Wolfgang
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA; New York University, New York, NY, USA
| | - Ming Lei
- NovaRock, Biotherapeutics Ltd., Ewing, NJ, USA.
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lei Zheng
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Department of Surgery, and the Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
67
|
Merlano MC, Denaro N, Galizia D, Ruatta F, Occelli M, Minei S, Abbona A, Paccagnella M, Ghidini M, Garrone O. How Chemotherapy Affects the Tumor Immune Microenvironment: A Narrative Review. Biomedicines 2022; 10:biomedicines10081822. [PMID: 36009369 PMCID: PMC9405073 DOI: 10.3390/biomedicines10081822] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Chemotherapy is much more effective in immunocompetent mice than in immunodeficient ones, and it is now acknowledged that an efficient immune system is necessary to optimize chemotherapy activity and efficacy. Furthermore, chemotherapy itself may reinvigorate immune response in different ways: by targeting cancer cells through the induction of cell stress, the release of damage signals and the induction of immunogenic cell death, by targeting immune cells, inhibiting immune suppressive cells and/or activating immune effector cells; and by targeting the host physiology through changes in the balance of gut microbiome. All these effects acting on immune and non-immune components interfere with the tumor microenvironment, leading to the different activity and efficacy of treatments. This article describes the correlation between chemotherapy and the immune changes induced in the tumor microenvironment. Our ultimate aim is to pave the way for the identification of the best drugs or combinations, the doses, the schedules and the right sequences to use when chemotherapy is combined with immunotherapy.
Collapse
Affiliation(s)
- Marco Carlo Merlano
- Scientific Direction, Candiolo Cancer Institute, FPO-IRCCS Candiolo, 10060 Torino, Italy
- Correspondence:
| | - Nerina Denaro
- Department of Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.D.); (F.R.); (M.G.); (O.G.)
| | - Danilo Galizia
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo, Italy;
| | - Fiorella Ruatta
- Department of Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.D.); (F.R.); (M.G.); (O.G.)
| | - Marcella Occelli
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy;
| | - Silvia Minei
- Post-Graduate School of Specialization Medical Oncology, University of Bari “A.Moro”, 70120 Bari, Italy;
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, 70120 Bari, Italy
| | - Andrea Abbona
- Translational Oncology ARCO Foundation, 12100 Cuneo, Italy; (A.A.); (M.P.)
| | - Matteo Paccagnella
- Translational Oncology ARCO Foundation, 12100 Cuneo, Italy; (A.A.); (M.P.)
| | - Michele Ghidini
- Department of Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.D.); (F.R.); (M.G.); (O.G.)
| | - Ornella Garrone
- Department of Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.D.); (F.R.); (M.G.); (O.G.)
| |
Collapse
|
68
|
The cell-line-derived subcutaneous tumor model in preclinical cancer research. Nat Protoc 2022; 17:2108-2128. [PMID: 35859135 DOI: 10.1038/s41596-022-00709-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 03/31/2022] [Indexed: 01/09/2023]
Abstract
Tumor-bearing experimental animals are essential for preclinical cancer drug development. A broad range of tumor models is available, with the simplest and most widely used involving a tumor of mouse or human origin growing beneath the skin of a mouse: the subcutaneous tumor model. Here, we outline the different types of in vivo tumor model, including some of their advantages and disadvantages and how they fit into the drug-development process. We then describe in more detail the subcutaneous tumor model and key steps needed to establish it in the laboratory, namely: choosing the mouse strain and tumor cells; cell culture, preparation and injection of tumor cells; determining tumor volume; mouse welfare; and an appropriate experimental end point. The protocol leads to subcutaneous tumor growth usually within 1-3 weeks of cell injection and is suitable for those with experience in tissue culture and mouse experimentation.
Collapse
|
69
|
Lê H, Seitlinger J, Lindner V, Olland A, Falcoz PE, Benkirane-Jessel N, Quéméneur E. Patient-Derived Lung Tumoroids—An Emerging Technology in Drug Development and Precision Medicine. Biomedicines 2022; 10:biomedicines10071677. [PMID: 35884982 PMCID: PMC9312903 DOI: 10.3390/biomedicines10071677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/16/2022] Open
Abstract
Synthetic 3D multicellular systems derived from patient tumors, or tumoroids, have been developed to complete the cancer research arsenal and overcome the limits of current preclinical models. They aim to represent the molecular and structural heterogeneity of the tumor micro-environment, and its complex network of interactions, with greater accuracy. They are more predictive of clinical outcomes, of adverse events, and of resistance mechanisms. Thus, they increase the success rate of drug development, and help clinicians in their decision-making process. Lung cancer remains amongst the deadliest of diseases, and still requires intensive research. In this review, we analyze the merits and drawbacks of the current preclinical models used in lung cancer research, and the position of tumoroids. The introduction of immune cells and healthy regulatory cells in autologous tumoroid models has enabled their application to most recent therapeutic concepts. The possibility of deriving tumoroids from primary tumors within reasonable time has opened a direct approach to patient-specific features, supporting their future role in precision medicine.
Collapse
Affiliation(s)
- Hélène Lê
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Transgène SA, 400 Boulevard Gonthier d’Andernach, 67400 Illkirch-Graffenstaden, France
| | - Joseph Seitlinger
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Véronique Lindner
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Anne Olland
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Pierre-Emmanuel Falcoz
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Eric Quéméneur
- Transgène SA, 400 Boulevard Gonthier d’Andernach, 67400 Illkirch-Graffenstaden, France
- Correspondence:
| |
Collapse
|
70
|
Guzzeloni V, Veschini L, Pedica F, Ferrero E, Ferrarini M. 3D Models as a Tool to Assess the Anti-Tumor Efficacy of Therapeutic Antibodies: Advantages and Limitations. Antibodies (Basel) 2022; 11:antib11030046. [PMID: 35892706 PMCID: PMC9326665 DOI: 10.3390/antib11030046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are an emerging and very active frontier in clinical oncology, with hundred molecules currently in use or being tested. These treatments have already revolutionized clinical outcomes in both solid and hematological malignancies. However, identifying patients who are most likely to benefit from mAbs treatment is currently challenging and limiting the impact of such therapies. To overcome this issue, and to fulfill the expectations of mAbs therapies, it is urgently required to develop proper culture models capable of faithfully reproducing the interactions between tumor and its surrounding native microenvironment (TME). Three-dimensional (3D) models which allow the assessment of the impact of drugs on tumors within its TME in a patient-specific context are promising avenues to progressively fill the gap between conventional 2D cultures and animal models, substantially contributing to the achievement of personalized medicine. This review aims to give a brief overview of the currently available 3D models, together with their specific exploitation for therapeutic mAbs testing, underlying advantages and current limitations to a broader use in preclinical oncology.
Collapse
Affiliation(s)
- Virginia Guzzeloni
- B-Cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (V.G.); (E.F.)
| | - Lorenzo Veschini
- Academic Centre of Reconstructive Science, Faculty of Dentistry Oral & Craniofacial Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Federica Pedica
- Pathology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Elisabetta Ferrero
- B-Cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (V.G.); (E.F.)
| | - Marina Ferrarini
- B-Cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (V.G.); (E.F.)
- Correspondence:
| |
Collapse
|
71
|
Functional landscapes of POLE and POLD1 mutations in checkpoint blockade-dependent antitumor immunity. Nat Genet 2022; 54:996-1012. [PMID: 35817971 PMCID: PMC10181095 DOI: 10.1038/s41588-022-01108-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 05/26/2022] [Indexed: 12/13/2022]
Abstract
Defects in pathways governing genomic fidelity have been linked to improved response to immune checkpoint blockade therapy (ICB). Pathogenic POLE/POLD1 mutations can cause hypermutation, yet how diverse mutations in POLE/POLD1 influence antitumor immunity following ICB is unclear. Here, we comprehensively determined the effect of POLE/POLD1 mutations in ICB and elucidated the mechanistic impact of these mutations on tumor immunity. Murine syngeneic tumors harboring Pole/Pold1 functional mutations displayed enhanced antitumor immunity and were sensitive to ICB. Patients with POLE/POLD1 mutated tumors harboring telltale mutational signatures respond better to ICB than patients harboring wild-type or signature-negative tumors. A mutant POLE/D1 function-associated signature-based model outperformed several traditional approaches for identifying POLE/POLD1 mutated patients that benefit from ICB. Strikingly, the spectrum of mutational signatures correlates with the biochemical features of neoantigens. Alterations that cause POLE/POLD1 function-associated signatures generate T cell receptor (TCR)-contact residues with increased hydrophobicity, potentially facilitating T cell recognition. Altogether, the functional landscapes of POLE/POLD1 mutations shape immunotherapy efficacy.
Collapse
|
72
|
Peters S, Paz-Ares L, Herbst RS, Reck M. Addressing CPI resistance in NSCLC: targeting TAM receptors to modulate the tumor microenvironment and future prospects. J Immunother Cancer 2022; 10:jitc-2022-004863. [PMID: 35858709 PMCID: PMC9305809 DOI: 10.1136/jitc-2022-004863] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2022] [Indexed: 01/09/2023] Open
Abstract
Lung cancer remains a leading cause of cancer death worldwide, with non-small-cell lung cancer (NSCLC) accounting for the majority of cases. Immune checkpoint inhibitors (CPIs), including those targeting programmed cell death protein-1 and its ligand (PD-1/PD-L1), have revolutionized the treatment landscape for various cancers. Notably, PD-1/PD-L1 inhibitor-based regimens now form the standard first-line therapy for metastatic NSCLC, substantially improving patients' overall survival. Despite the progress made using CPI-based therapies in advanced NSCLC, most patients experience disease progression after an initial response due to resistance. Given the currently limited therapeutic options available for second-line and beyond settings in NSCLC, new treatment approaches are needed to improve long-term survival in these patients. Thus, CPI resistance is an emerging concept in cancer treatment and an active area of clinical research.Among the key mechanisms of CPI resistance is the immunosuppressive tumor microenvironment (TME). Effective CPI therapy is based on shifting immune responses against cancer cells, therefore, manipulating the immunosuppressive TME comprises an important strategy to combat CPI resistance. Several aspects of the TME can contribute to treatment resistance in NSCLC, including through the activation of Tyro3, Axl, MerTK (TAM) receptors which are essential pleiotropic regulators of immune homeostasis. Their roles include negatively modulating the immune response, therefore ectopic expression of TAM receptors in the context of cancer can contribute to the immunosuppressive, protumorigenic TME. Furthermore, TAM receptors represent important candidates to simultaneously target both tumor cells and immune cells in the TME. Clinical development of TAM receptor inhibitors (TAM RIs) is increasingly focused on their ability to rescue the antitumor immune response, thereby shifting the immunosuppressive TME to an immunostimulatory TME. There is a strong biological rationale for combining TAM RIs with a CPI to overcome resistance and improve long-term clinical responses in NSCLC. Combinatorial clinical trials of TAM RIs with CPIs are underway with encouraging preliminary results. This review outlines the key mechanisms of CPI resistance, including the role of the immunosuppressive TME, and discusses the rationale for targeting TAM receptors as a novel, promising therapeutic strategy to overcome CPI resistance in NSCLC.
Collapse
Affiliation(s)
- Solange Peters
- Medical Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Luis Paz-Ares
- Medical Oncology Department, Hospital Universitario 12 de Octubre and CNIO-H12O Lung Cancer Unit, Universidad Complutense and Ciberonc, Madrid, Spain
| | - Roy S Herbst
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, Center for Lung Research, Grosshansdorf, Germany
| |
Collapse
|
73
|
Liu S, Huang F, Ru G, Wang Y, Zhang B, Chen X, Chu L. Mouse Models of Hepatocellular Carcinoma: Classification, Advancement, and Application. Front Oncol 2022; 12:902820. [PMID: 35847898 PMCID: PMC9279915 DOI: 10.3389/fonc.2022.902820] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/01/2022] [Indexed: 11/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the subtype of liver cancer with the highest incidence, which is a heterogeneous malignancy with increasing incidence rate and high mortality. For ethical reasons, it is essential to validate medical clinical trials for HCC in animal models before further consideration on humans. Therefore, appropriate models for the study of the pathogenesis of the disease and related treatment methods are necessary. For tumor research, mouse models are the most commonly used and effective in vivo model, which is closer to the real-life environment, and the repeated experiments performed on it are closer to the real situation. Several mouse models of HCC have been developed with different mouse strains, cell lines, tumor sites, and tumor formation methods. In this review, we mainly introduce some mouse HCC models, including induced model, gene-edited model, HCC transplantation model, and other mouse HCC models, and discuss how to choose the appropriate model according to the purpose of the experiments.
Collapse
Affiliation(s)
- Sha Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Huang
- Cancer Center, Department of Pathology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Guoqing Ru
- Cancer Center, Department of Pathology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Liang Chu,
| |
Collapse
|
74
|
Yuan M, Zhao Y, Arkenau HT, Lao T, Chu L, Xu Q. Signal pathways and precision therapy of small-cell lung cancer. Signal Transduct Target Ther 2022; 7:187. [PMID: 35705538 PMCID: PMC9200817 DOI: 10.1038/s41392-022-01013-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 03/05/2022] [Accepted: 04/29/2022] [Indexed: 12/24/2022] Open
Abstract
Small-cell lung cancer (SCLC) encounters up 15% of all lung cancers, and is characterized by a high rate of proliferation, a tendency for early metastasis and generally poor prognosis. Most of the patients present with distant metastatic disease at the time of clinical diagnosis, and only one-third are eligible for potentially curative treatment. Recently, investigations into the genomic make-up of SCLC show extensive chromosomal rearrangements, high mutational burden and loss-of-function mutations of several tumor suppressor genes. Although the clinical development of new treatments for SCLC has been limited in recent years, a better understanding of oncogenic driver alterations has found potential novel targets that might be suitable for therapeutic approaches. Currently, there are six types of potential treatable signaling pathways in SCLC, including signaling pathways targeting the cell cycle and DNA repair, tumor development, cell metabolism, epigenetic regulation, tumor immunity and angiogenesis. At this point, however, there is still a lack of understanding of their role in SCLC tumor biology and the promotion of cancer growth. Importantly optimizing drug targets, improving drug pharmacology, and identifying potential biomarkers are the main focus and further efforts are required to recognize patients who benefit most from novel therapies in development. This review will focus on the current learning on the signaling pathways, the status of immunotherapy, and targeted therapy in SCLC.
Collapse
Affiliation(s)
- Min Yuan
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China
| | - Yu Zhao
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China
| | | | - Tongnei Lao
- Department of Oncology, Centro Medico BO CHI, Macao, SAR, China
| | - Li Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China.
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China.
| |
Collapse
|
75
|
Sanmamed MF, Berraondo P, Rodriguez-Ruiz ME, Melero I. Charting roadmaps towards novel and safe synergistic immunotherapy combinations. NATURE CANCER 2022; 3:665-680. [PMID: 35764745 DOI: 10.1038/s43018-022-00401-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
Checkpoint inhibitor-based cancer immunotherapy is often combined in the clinic with other immunotherapy strategies, targeted therapies, chemotherapy or standard-of-care treatments to achieve superior therapeutic efficacy. The large number of immunotherapy combinations that are currently undergoing clinical testing necessitate the establishment of faithful criteria to prioritize optimal combinations with evidence of synergy, to determine their safety and optimal sequence of administration and to identify biomarkers of therapy resistance and response. In this review, we focus on recent developments in immunotherapy combinations and reflect on how combinations should be optimized to maximize the impact of immunotherapy in clinical oncology.
Collapse
Affiliation(s)
- Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Maria E Rodriguez-Ruiz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain.
| |
Collapse
|
76
|
Xu D, Wang L, Wieczorek K, Zhang Y, Wang Z, Wang J, Xu B, Singh PK, Wang Y, Zhang X, Wu Y, Smith GJ, Attwood K, Zhang Y, Goodrich DW, Li Q. Single-Cell Analyses of a Novel Mouse Urothelial Carcinoma Model Reveal a Role of Tumor-Associated Macrophages in Response to Anti-PD-1 Therapy. Cancers (Basel) 2022; 14:cancers14102511. [PMID: 35626115 PMCID: PMC9139541 DOI: 10.3390/cancers14102511] [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: 04/10/2022] [Revised: 05/10/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023] Open
Abstract
Approximately 80% of patients with advanced bladder cancer do not respond to immune checkpoint inhibitor (ICI) immunotherapy. Therefore, there is an urgent unmet need to develop clinically relevant preclinical models so that factors governing immunotherapy responses can be studied in immunocompetent mice. We developed a line of mouse triple knockout (TKO: Trp53, Pten, Rb1) urothelial carcinoma organoids transplanted into immunocompetent mice. These bladder tumors recapitulate the molecular phenotypes and heterogeneous immunotherapy responses observed in human bladder cancers. The TKO organoids were characterized in vivo and in vitro and compared to the widely used MB49 murine bladder cancer model. RNAseq analysis of the TKO tumors demonstrated a basal subtype. The TKO xenografts demonstrated the expression of urothelial markers (CK5, CK7, GATA3, and p63), whereas MB49 subcutaneous xenografts did not express urothelial markers. Anti-PD-1 immunotherapy resulted in a mixed pattern of treatment responses for individual tumors. Eight immune cell types were identified (basophils, B cells, dendritic cells, macrophages, monocytes, neutrophils, NK cells, and T cells) in ICI-treated xenografts. Responder xenografts displayed significantly increased immune cell infiltration (15.3%, 742 immune cells/4861 total cells) compared to the non-responder tumors (10.1%, 452 immune cells/4459 total cells, Fisher Exact Test p < 0.0001). Specifically, there were more T cells (1.0% vs. 0.4%, p = 0.002) and macrophages (8.6% vs. 6.4%, p = 0.0002) in responder xenografts than in non-responder xenografts. In conclusion, we have developed a novel preclinical model that exhibits a mixed pattern of response to anti-PD-1 immunotherapy. The higher percentage of macrophage tumor infiltration in responders suggests a potential role for the innate immune microenvironment in regulating ICI treatment responses.
Collapse
Affiliation(s)
- Dongbo Xu
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (D.X.); (L.W.); (K.W.); (Y.W.); (G.J.S.)
| | - Li Wang
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (D.X.); (L.W.); (K.W.); (Y.W.); (G.J.S.)
| | - Kyle Wieczorek
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (D.X.); (L.W.); (K.W.); (Y.W.); (G.J.S.)
| | - Yali Zhang
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (Y.Z.); (J.W.); (K.A.)
| | - Zinian Wang
- Departments of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Jianmin Wang
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (Y.Z.); (J.W.); (K.A.)
| | - Bo Xu
- Departments of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Prashant K. Singh
- Departments of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Yanqing Wang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (Y.W.); (X.Z.); (Y.Z.); (D.W.G.)
| | - Xiaojing Zhang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (Y.W.); (X.Z.); (Y.Z.); (D.W.G.)
| | - Yue Wu
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (D.X.); (L.W.); (K.W.); (Y.W.); (G.J.S.)
| | - Gary J. Smith
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (D.X.); (L.W.); (K.W.); (Y.W.); (G.J.S.)
| | - Kristopher Attwood
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (Y.Z.); (J.W.); (K.A.)
| | - Yuesheng Zhang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (Y.W.); (X.Z.); (Y.Z.); (D.W.G.)
| | - David W. Goodrich
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (Y.W.); (X.Z.); (Y.Z.); (D.W.G.)
| | - Qiang Li
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (D.X.); (L.W.); (K.W.); (Y.W.); (G.J.S.)
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (Y.W.); (X.Z.); (Y.Z.); (D.W.G.)
- Correspondence: ; Tel.: +1-716-845-3389; Fax: +1-716-845-3300
| |
Collapse
|
77
|
Li J, Zhang P, Zhou M, Liu C, Huang Y, Li L. Trauma-Responsive Scaffold Synchronizing Oncolysis Immunization and Inflammation Alleviation for Post-Operative Suppression of Cancer Metastasis. ACS NANO 2022; 16:6064-6079. [PMID: 35344338 DOI: 10.1021/acsnano.1c11562] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor surgery can create an inflammatory trauma to aggravate residual tumor "seed" to colonize pre-metastatic niches (PMNs) "soil" at secondary sites, thereby promoting post-operative metastasis. However, two-pronged strategies for post-surgical elimination of asynchronous "seeds" and "soil" at different regions are currently lacking. Here, we have designed a hydrogel that can be injected into a resection cavity, where it immediately forms a scaffold and gradually degrades responding to enriched reactive oxygen species at adjacent trauma for local delivery and on-demand release of autologous cancer cells succumbing to oncolysis (ACCO) and anti-inflammatory agent. The autologous cell source self-provides a whole array of tumor-associated antigens, and the oncolysis orchestration of a subcellular cascade confers a self-adjuvanting property, together guaranteeing high immunogenicity of the ACCO vaccine that enables specific antitumor immunization. In parallel, inflammation alleviation exerted bidirectional functions to reshape the local immune landscape and resuscitate ACCO, leading to the eradication of residual tumor "seeds" while simultaneously intercepting the "seed-soil" crosstalk to normalize distant lung leading to regression of pre-existing PMN "soil". As a result, regional and metastatic recurrence were completely thwarted. Together, this framework synchronizing oncolysis immunization and inflammation alleviation provides an effective option for post-operative suppression of metastasis.
Collapse
Affiliation(s)
- Junlin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ping Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Minglu Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chendong Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
78
|
Lee SK, Park MJ, Choi JW, Baek JA, Kim SY, Choi HJ, You YK, Jang JW, Sung PS, Bae SH, Yoon SK, Choi JY, Cho ML. Patient-Derived Avatar Mouse Model to Predict the Liver Immune Homeostasis of Long-Term Stable Liver Transplant Patients. Front Immunol 2022; 13:817006. [PMID: 35418987 PMCID: PMC8995467 DOI: 10.3389/fimmu.2022.817006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Although rejection or tolerance can occur in liver transplantation (LT) patients, there are no reliable non-invasive methods for predicting immune homeostasis. In this study, we developed a humanized mouse model to predict liver immune homeostasis in patients who underwent LT. The patient-derived avatar model was developed by injecting peripheral blood mononuclear cells from healthy controls (HCs) or LT patients with stable, rejection, or tolerance into NOD.Cg-PrkdcscidIL2rgtm1Wjl/SzJ (NSG) mice, followed by injection of human hepatic stellate cells and Carbone tetrachloride (CCl4). After 7 weeks, the patient’s T-cell engraftment and liver inflammation in the avatar model were evaluated and compared with the liver histology of LT patients. Changes in liver inflammation following treatment with tacrolimus and/or biguanide derivatives were also examined. The C-X-C Motif Chemokine Receptor 3 (CXCR3)-dependently engrafted patient T cells led to differences in liver inflammation in our model according to the status of LT patients. The livers of avatar models from rejection patients had severe inflammation with more T helper 17 cells and fewer regulatory T cells compared to those of models from tolerance and HCs showing only mild inflammation. Moreover, our model classified stable post-LT patients into severe and mild inflammation groups, which correlated well with liver immunity in these patients. Our models revealed alleviation of inflammation after combination treatment with tacrolimus and biguanide derivatives or monotherapy. Consequently, using our new patient-derived avatar model, we predicted liver immune homeostasis in patients with stable LT without biopsy. Moreover, our avatar model may be useful for preclinical analysis to evaluate treatment responses while reducing risks to patients.
Collapse
Affiliation(s)
- Soon Kyu Lee
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Min-Jung Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jeong Won Choi
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jin-Ah Baek
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Se-Young Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ho Joong Choi
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Young Kyoung You
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jeong Won Jang
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Pil Soo Sung
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Si Hyun Bae
- Division of Hepatology, Department of Internal Medicine, Eunpyeong Se. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung Kew Yoon
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jong Young Choi
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
79
|
Wang S, Zhao Z, Xie J, Ren S, Tang S, Lin F, Qu J, Liu L. Accurate evaluation of the treatment effects of immunotherapy on subcutaneous ovarian cancer in mice with nonlinear optical imaging and algorithmic analysis. BIOMEDICAL OPTICS EXPRESS 2022; 13:2266-2277. [PMID: 35519272 PMCID: PMC9045922 DOI: 10.1364/boe.452569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/05/2022] [Accepted: 03/10/2022] [Indexed: 06/14/2023]
Abstract
Immunotherapy and its evaluation have shown great promise for cancer treatment. Here, a mouse subcutaneous transplantable tumor model was applied to testing therapeutic strategies. The mouse model was treated by regulating anti-PD-L1, anti-CTLA-4, cisplatin and their combined therapy. Biochemistry experiments have found that after immunotherapy, mice have more immune responses, which were manifested by an increase in the content of growth factors and the activation of T cells. Meanwhile, multimodal nonlinear optical microscopy imaging combined with algorithms was used to evaluate the treatment's effectiveness. By detecting the metabolism rate and microstructure in tissue, it was proved that combined therapies including immune checkpoint inhibitors do have a better effect on ovarian tumors. Our discovery of valid treatments for mice with ovarian tumor and provides an evaluation tool via nonlinear optics combined with algorithms offers new insights into therapeutic effect.
Collapse
Affiliation(s)
- Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
- These authors contributed equally
| | - Zewei Zhao
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
- These authors contributed equally
| | - Jiajun Xie
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Sheng Ren
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Shiyao Tang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Fangrui Lin
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| |
Collapse
|
80
|
Philip M, Schietinger A. CD8 + T cell differentiation and dysfunction in cancer. Nat Rev Immunol 2022; 22:209-223. [PMID: 34253904 PMCID: PMC9792152 DOI: 10.1038/s41577-021-00574-3] [Citation(s) in RCA: 423] [Impact Index Per Article: 211.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 02/07/2023]
Abstract
CD8+ T cells specific for cancer cells are detected within tumours. However, despite their presence, tumours progress. The clinical success of immune checkpoint blockade and adoptive T cell therapy demonstrates the potential of CD8+ T cells to mediate antitumour responses; however, most patients with cancer fail to achieve long-term responses to immunotherapy. Here we review CD8+ T cell differentiation to dysfunctional states during tumorigenesis. We highlight similarities and differences between T cell dysfunction and other hyporesponsive T cell states and discuss the spatio-temporal factors contributing to T cell state heterogeneity in tumours. An important challenge is predicting which patients will respond to immunotherapeutic interventions and understanding which T cell subsets mediate the clinical response. We explore our current understanding of what determines T cell responsiveness and resistance to immunotherapy and point out the outstanding research questions.
Collapse
Affiliation(s)
- Mary Philip
- Vanderbilt Center for Immunobiology, Vanderbilt-Ingram Cancer Center, Department of Medicine/Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.,;
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,;
| |
Collapse
|
81
|
Translation of focused ultrasound for blood-brain barrier opening in glioma. J Control Release 2022; 345:443-463. [PMID: 35337938 DOI: 10.1016/j.jconrel.2022.03.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022]
Abstract
Survival outcomes for patients with glioblastoma multiforme (GBM) have remained poor for the past 15 years, reflecting a clear challenge in the development of more effective treatment strategies. The efficacy of systemic therapies for GBM is greatly limited by the presence of the blood-brain barrier (BBB), which prevents drug penetration and accumulation in regions of infiltrative tumour, as represented in a consistent portion of GBM lesions. Focused ultrasound (FUS) - a technique that uses low-frequency ultrasound waves to induce targeted temporary disruption of the BBB - promises to improve survival outcomes by enhancing drug delivery and accumulation to infiltrating tumour regions. In this review we discuss the current state of preclinical investigations using FUS to enhance delivery of systemic therapies to intracranial neoplasms. We highlight critical methodological inconsistencies that are hampering clinical translation of FUS and we provide guiding principles for future preclinical studies. Particularly, we focus our attention on the importance of the selection of clinically relevant animal models and to the standardization of methods for FUS delivery, which will be paramount to the successful clinical translation of this promising technology for treatment in GBM patients. We also discuss how preclinical FUS research can benefit the development of GBM immunotherapies.
Collapse
|
82
|
Preclinical Evaluation of CAR T Cell Function: In Vitro and In Vivo Models. Int J Mol Sci 2022; 23:ijms23063154. [PMID: 35328572 PMCID: PMC8955360 DOI: 10.3390/ijms23063154] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 01/12/2023] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR) T cells is a rapidly emerging modality that engineers T cells to redirect tumor-specific cytotoxicity. CAR T cells have been well characterized for their efficacy against B cell malignancies, and rigorously studied in other types of tumors. Preclinical evaluation of CAR T cell function, including direct tumor killing, cytokine production, and memory responses, is crucial to the development and optimization of CAR T cell therapies. Such comprehensive examinations are usually performed in different types of models. Model establishment should focus on key challenges in the clinical setting and the capability to generate reliable data to indicate CAR T cell therapeutic potency in the clinic. Further, modeling the interaction between CAR T cells and tumor microenvironment provides additional insight for the future endeavors to enhance efficacy, especially against solid tumors. This review will summarize both in vitro and in vivo models for CAR T cell functional evaluation, including how they have evolved with the needs of CAR T cell research, the information they can provide for preclinical assessment of CAR T cell products, and recent technology advances to test CAR T cells in more clinically relevant models.
Collapse
|
83
|
Correia C, Weiskittel TM, Ung CY, Villasboas Bisneto JC, Billadeau DD, Kaufmann SH, Li H. Uncovering Pharmacological Opportunities for Cancer Stem Cells-A Systems Biology View. Front Cell Dev Biol 2022; 10:752326. [PMID: 35359437 PMCID: PMC8962639 DOI: 10.3389/fcell.2022.752326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/10/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer stem cells (CSCs) represent a small fraction of the total cancer cell population, yet they are thought to drive disease propagation, therapy resistance and relapse. Like healthy stem cells, CSCs possess the ability to self-renew and differentiate. These stemness phenotypes of CSCs rely on multiple molecular cues, including signaling pathways (for example, WNT, Notch and Hedgehog), cell surface molecules that interact with cellular niche components, and microenvironmental interactions with immune cells. Despite the importance of understanding CSC biology, our knowledge of how neighboring immune and tumor cell populations collectively shape CSC stemness is incomplete. Here, we provide a systems biology perspective on the crucial roles of cellular population identification and dissection of cell regulatory states. By reviewing state-of-the-art single-cell technologies, we show how innovative systems-based analysis enables a deeper understanding of the stemness of the tumor niche and the influence of intratumoral cancer cell and immune cell compositions. We also summarize strategies for refining CSC systems biology, and the potential role of this approach in the development of improved anticancer treatments. Because CSCs are amenable to cellular transitions, we envision how systems pharmacology can become a major engine for discovery of novel targets and drug candidates that can modulate state transitions for tumor cell reprogramming. Our aim is to provide deeper insights into cancer stemness from a systems perspective. We believe this approach has great potential to guide the development of more effective personalized cancer therapies that can prevent CSC-mediated relapse.
Collapse
Affiliation(s)
- Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Taylor M Weiskittel
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Choong Yong Ung
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | | | - Daniel D Billadeau
- Department of Immunology, Mayo Clinic, Rochester, MN, United States,Division of Oncology Research, Mayo Clinic, Rochester, MN, United States
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States,Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, United States,Division of Oncology Research, Mayo Clinic, Rochester, MN, United States
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States,*Correspondence: Hu Li,
| |
Collapse
|
84
|
Fucikova J, Palova-Jelinkova L, Klapp V, Holicek P, Lanickova T, Kasikova L, Drozenova J, Cibula D, Álvarez-Abril B, García-Martínez E, Spisek R, Galluzzi L. Immunological control of ovarian carcinoma by chemotherapy and targeted anticancer agents. Trends Cancer 2022; 8:426-444. [PMID: 35181272 DOI: 10.1016/j.trecan.2022.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/19/2022] [Indexed: 12/24/2022]
Abstract
At odds with other solid tumors, epithelial ovarian cancer (EOC) is poorly sensitive to immune checkpoint inhibitors (ICIs), largely reflecting active immunosuppression despite CD8+ T cell infiltration at baseline. Accumulating evidence indicates that both conventional chemotherapeutics and targeted anticancer agents commonly used in the clinical management of EOC not only mediate a cytostatic and cytotoxic activity against malignant cells, but also drive therapeutically relevant immunostimulatory or immunosuppressive effects. Here, we discuss such an immunomodulatory activity, with a specific focus on molecular and cellular pathways that can be harnessed to develop superior combinatorial regimens for clinical EOC care.
Collapse
Affiliation(s)
- Jitka Fucikova
- Sotio, Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic.
| | - Lenka Palova-Jelinkova
- Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Vanessa Klapp
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Peter Holicek
- Sotio, Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Tereza Lanickova
- Sotio, Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | | | - Jana Drozenova
- Department of Pathology, Third Faculty of Medicine and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - David Cibula
- Gynecologic Oncology Center, Department of Obstetrics and Gynecology, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Beatriz Álvarez-Abril
- Department of Hematology and Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Elena García-Martínez
- Department of Hematology and Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain; Universidad Católica San Antonio de Murcia, Guadalupe, Spain
| | - Radek Spisek
- Sotio, Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Centre, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
85
|
Mondal P, Bailey KL, Cartwright SB, Band V, Carlson MA. Large Animal Models of Breast Cancer. Front Oncol 2022; 12:788038. [PMID: 35186735 PMCID: PMC8855936 DOI: 10.3389/fonc.2022.788038] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/18/2022] [Indexed: 01/29/2023] Open
Abstract
In this mini review the status, advantages, and disadvantages of large animal modeling of breast cancer (BC) will be discussed. While most older studies of large animal BC models utilized canine and feline subjects, more recently there has been interest in development of porcine BC models, with some early promising results for modeling human disease. Widely used rodent models of BC were briefly reviewed to give context to the work on the large animal BC models. Availability of large animal BC models could provide additional tools for BC research, including availability of human-sized subjects and BC models with greater biologic relevance.
Collapse
Affiliation(s)
- Pinaki Mondal
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, United States,Department of Surgery, VA Medical Center, Omaha, NE, United States
| | - Katie L. Bailey
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sara B. Cartwright
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, United States
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mark A. Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, United States,Department of Surgery, VA Medical Center, Omaha, NE, United States,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States,Center for Advanced Surgical Technology, University of Nebraska Medical Center, Omaha, NE, United States,*Correspondence: Mark A. Carlson,
| |
Collapse
|
86
|
Oshima Y, Sano M, Kajiwara I, Ichimaru Y, Itaya T, Kuramochi T, Hayashi E, Kim J, Kitajima O, Masugi Y, Masamune A, Ijichi H, Ishii Y, Suzuki T. Midazolam exhibits antitumour and anti-inflammatory effects in a mouse model of pancreatic ductal adenocarcinoma. Br J Anaesth 2022; 128:679-690. [PMID: 35120712 DOI: 10.1016/j.bja.2021.12.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/03/2021] [Accepted: 12/18/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Anaesthesia and perioperative management contribute to long-term outcomes of patients with cancer, including pancreatic ductal adenocarcinoma. We assessed the antitumour, anti-inflammatory, and analgesic effects of midazolam on LSL-KrasG12D/+;Trp53flox/flox;Pdx-1cre/+ transgenic mice with pancreatic ductal adenocarcinoma. METHODS Six-week-old transgenic mice were administered midazolam 30 mg kg-1 day-1 p.o. (n=13); midazolam 30 mg kg-1 day-1 with 1-(2-chlorophenyl)-N-methyl-N(1-methylpropyl)-3-isoquinoline carboxamide (PK11195) 3 mg kg-1 day-1 i.p., a peripheral benzodiazepine receptor antagonist (n=10); or vehicle (water; n=14) until the humane endpoint. Cancer-associated pain was evaluated using hunching score and mouse grimace scale. Tumour stage and immuno-inflammatory status were determined histopathologically. Anti-proliferative and apoptotic potentials of midazolam were investigated using mouse pancreatic ductal adenocarcinoma cell lines. RESULTS Midazolam significantly inhibited tumour size and proliferative index of Ki-67 and cyclins in pancreatic ductal adenocarcinoma, which was blocked by administration of PK11195. Local myeloperoxidase+ tumour-associated neutrophils, arginase-1+ M2-like tumour-associated macrophages, and CD11b+Ly-6G+ polymorphonuclear myeloid-derived suppressor cells were reduced by midazolam, which was antagonised by administration of PK11195. Hunching and mouse grimace scale were improved by midazolam, whereas the scores increased with midazolam+PK11195 treatment. Plasma pro-inflammatory cytokines, such as interleukin-6 and CC chemokine ligand (CCL)2, CCL3, and CCL5, were reduced by midazolam, whereas these cytokines increased with PK11195. Midazolam inhibited pancreatic ductal adenocarcinoma proliferation through downregulation of cyclins and cyclin-dependent kinases and induced apoptosis in vitro. CONCLUSIONS These results suggest that midazolam inhibits pancreatic ductal adenocarcinoma proliferation and local infiltration of tumour-associated neutrophils, tumour-associated macrophages, and polymorphonuclear myeloid-derived suppressor cells, thereby inhibiting pancreatic ductal adenocarcinoma progression.
Collapse
Affiliation(s)
- Yukino Oshima
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Makoto Sano
- Division of Medical Research Planning and Development, Nihon University School of Medicine, Tokyo, Japan.
| | - Ichie Kajiwara
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Yoshimi Ichimaru
- Faculty of Pharmaceutical Sciences, Shonan University of Medical Sciences, Yokohama, Japan
| | - Tomoaki Itaya
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Tomoya Kuramochi
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Emiko Hayashi
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Jinsuk Kim
- Division of Medical Research Planning and Development, Nihon University School of Medicine, Tokyo, Japan
| | - Osamu Kitajima
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| | - Yohei Masugi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideaki Ijichi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Clinical Nutrition Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukimoto Ishii
- Division of Medical Research Planning and Development, Nihon University School of Medicine, Tokyo, Japan
| | - Takahiro Suzuki
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
87
|
Griffiths GL, Vasquez C, Escorcia F, Clanton J, Lindenberg L, Mena E, Choyke PL. Translating a radiolabeled imaging agent to the clinic. Adv Drug Deliv Rev 2022; 181:114086. [PMID: 34942275 PMCID: PMC8889912 DOI: 10.1016/j.addr.2021.114086] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/30/2021] [Accepted: 12/16/2021] [Indexed: 02/03/2023]
Abstract
Molecular Imaging is entering the most fruitful, exciting period in its history with many new agents under development, and several reaching the clinic in recent years. While it is unusual for just one laboratory to take an agent from initial discovery through to full clinical approval the steps along the way are important to understand for all interested participants even if one is not involved in the entire process. Here, we provide an overview of these processes beginning at discovery and preclinical validation of a new molecular imaging agent and using as an exemplar a low molecular weight disease-specific targeted positron emission tomography (PET) agent. Compared to standard drug development requirements, molecular imaging agents may benefit from a regulatory standpoint from their low mass administered doses, they nonetheless still need to go through a series of well-defined steps before they can be considered for Phase 1 human testing. After outlining the discovery and preclinical validation approaches, we will also discuss the nuances of Phase 1, Phase 2 and Phase 3 studies that may culminate in an FDA general use approval. Finally, some post-approval aspects of novel molecular imaging agents are considered.
Collapse
Affiliation(s)
- Gary L. Griffiths
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD
| | - Crystal Vasquez
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| | - Freddy Escorcia
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| | | | - Liza Lindenberg
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| | - Esther Mena
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| | - Peter L. Choyke
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| |
Collapse
|
88
|
Sun J, Min YQ, Li Y, Sun X, Deng F, Wang H, Ning YJ. Animal Model of Severe Fever With Thrombocytopenia Syndrome Virus Infection. Front Microbiol 2022; 12:797189. [PMID: 35087498 PMCID: PMC8787146 DOI: 10.3389/fmicb.2021.797189] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/29/2021] [Indexed: 12/30/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS), an emerging life-threatening infectious disease caused by SFTS bunyavirus (SFTSV; genus Bandavirus, family Phenuiviridae, order Bunyavirales), has been a significant medical problem. Currently, there are no licensed vaccines or specific therapeutic agents available and the viral pathogenesis remains largely unclear. Developing appropriate animal models capable of recapitulating SFTSV infection in humans is crucial for both the study of the viral pathogenic processes and the development of treatment and prevention strategies. Here, we review the current progress in animal models for SFTSV infection by summarizing susceptibility of various potential animal models to SFTSV challenge and the clinical manifestations and histopathological changes in these models. Together with exemplification of studies on SFTSV molecular mechanisms, vaccine candidates, and antiviral drugs, in which animal infection models are utilized, the strengths and limitations of the existing SFTSV animal models and some important directions for future research are also discussed. Further exploration and optimization of SFTSV animal models and the corresponding experimental methods will be undoubtedly valuable for elucidating the viral infection and pathogenesis and evaluating vaccines and antiviral therapies.
Collapse
Affiliation(s)
- Jiawen Sun
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan-Qin Min
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yunjie Li
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xiulian Sun
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Fei Deng
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Hualin Wang
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yun-Jia Ning
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
89
|
Pan B, Wei X, Xu X. Patient-derived xenograft models in hepatopancreatobiliary cancer. Cancer Cell Int 2022; 22:41. [PMID: 35090441 PMCID: PMC8796540 DOI: 10.1186/s12935-022-02454-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 01/04/2022] [Indexed: 12/20/2022] Open
Abstract
Animal models are crucial tools for evaluating the biological progress of human cancers and for the preclinical investigation of anticancer drugs and cancer prevention. Various animals are widely used in hepatopancreatobiliary cancer research, and mouse models are the most popular. Generally, genetic tools, graft transplantation, and chemical and physical measures are adopted to generate sundry mouse models of hepatopancreatobiliary cancer. Graft transplantation is commonly used to study tumour progression. Over the past few decades, subcutaneous or orthotopic cell-derived tumour xenograft models (CDX models) have been developed to simulate distinct tumours in patients. However, two major limitations exist in CDX models. One model poorly simulates the microenvironment of tumours in humans, such as the vascular, lymphatic and immune environments. The other model loses genetic heterogeneity compared with the corresponding primary tumour. Increased efforts have focused on developing better models for hepatopancreatobiliary cancer research. Hepatopancreatobiliary cancer is considered a tumour with high molecular heterogeneity, making precision medicine challenging in cancer treatment. Developing a new animal model that can better mimic tumour tissue and more accurately predict the efficacy of anticancer treatments is urgent. For the past several years, the patient-derived xenograft model (PDX model) has emerged as a promising tool for translational research. It can retain the genetic and histological stability of their originating tumour at limited passages and shed light on precision cancer medicine. In this review, we summarize the methodology, advantages/disadvantages and applications of PDX models in hepatopancreatobiliary cancer research.
Collapse
|
90
|
Moquin-Beaudry G, Benabdallah B, Maggiorani D, Le O, Li Y, Colas C, Raggi C, Ellezam B, M'Callum MA, Dal Soglio D, Guimond JV, Paganelli M, Haddad E, Beauséjour C. Autologous humanized mouse models of iPSC-derived tumors enable characterization and modulation of cancer-immune cell interactions. CELL REPORTS METHODS 2022; 2:100153. [PMID: 35474871 PMCID: PMC9017190 DOI: 10.1016/j.crmeth.2021.100153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/03/2021] [Accepted: 12/21/2021] [Indexed: 01/21/2023]
Abstract
Modeling the tumor-immune cell interactions in humanized mice is complex and limits drug development. Here, we generated easily accessible tumor models by transforming either primary skin fibroblasts or induced pluripotent stem cell-derived cell lines injected in immune-deficient mice reconstituted with human autologous immune cells. Our results showed that fibroblastic, hepatic, or neural tumors were all efficiently infiltrated and partially or totally rejected by autologous immune cells in humanized mice. Characterization of tumor-immune infiltrates revealed high expression levels of the dysfunction markers Tim3 and PD-1 in T cells and an enrichment in regulatory T cells, suggesting rapid establishment of immunomodulatory phenotypes. Inhibition of PD-1 by Nivolumab in humanized mice resulted in increased immune cell infiltration and a slight decrease in tumor growth. We expect that these versatile and accessible cancer models will facilitate preclinical studies and the evaluation of autologous cancer immunotherapies across a range of different tumor cell types.
Collapse
Affiliation(s)
- Gaël Moquin-Beaudry
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Pharmacologie et Physiologie, Montréal, QC, Canada
| | - Basma Benabdallah
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Damien Maggiorani
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Oanh Le
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Yuanyi Li
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Chloé Colas
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Microbiologie, Immunologie et Infectiologie, Montréal, QC, Canada
| | - Claudia Raggi
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Benjamin Ellezam
- Département de Neurosciences, Montréal, QC, Canada
- Département de Pathologie, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Marie-Agnès M'Callum
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Biologie Moléculaire, Montréal, QC, Canada
| | - Dorothée Dal Soglio
- Département de Pathologie et Biologie Moléculaire, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
- Département de Pathologie, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Jean V. Guimond
- CIUSSS du Centre-Sud-de-l’Ile-de-Montréal, Montréal, QC, Canada
| | - Massimiliano Paganelli
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Biologie Moléculaire, Montréal, QC, Canada
- Division of Gastroenterology, Hepatology and Nutrition and Pediatric Liver Transplantation Program at CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
- Département de Pédiatrie, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Elie Haddad
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Microbiologie, Immunologie et Infectiologie, Montréal, QC, Canada
- Département de Pédiatrie, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Christian Beauséjour
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Pharmacologie et Physiologie, Montréal, QC, Canada
| |
Collapse
|
91
|
Miao YR, Xia M, Luo M, Luo T, Yang M, Guo AY. ImmuCellAI-mouse: a tool for comprehensive prediction of mouse immune cell abundance and immune microenvironment depiction. Bioinformatics 2022; 38:785-791. [PMID: 34636837 DOI: 10.1093/bioinformatics/btab711] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/22/2021] [Accepted: 10/08/2021] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Immune cells are important components of the immune system and are crucial for disease initiation, progression, prognosis and survival. Although several computational methods have been designed for predicting the abundance of immune cells, very few tools are applicable to mouse. Given that, mouse is the most widely used animal model in biomedical research, there is an urgent need to develop a precise algorithm for predicting mouse immune cells. RESULTS We developed a tool named Immune Cell Abundance Identifier for mouse (ImmuCellAI-mouse), for estimating the abundance of 36 immune cell (sub)types from gene expression data in a hierarchical strategy of three layers. Reference expression profiles and robust marker gene sets of immune cell types were curated. The abundance of cells in three layers was predicted separately by calculating the ssGSEA enrichment score of the expression deviation profile per cell type. Benchmark results showed high accuracy of ImmuCellAI-mouse in predicting most immune cell types, with correlation coefficients between predicted value and real cell proportion of most cell types being larger than 0.8. We applied ImmuCellAI-mouse to a mouse breast tumor dataset and revealed the dynamic change of immune cell infiltration during treatment, which is consistent with the findings of the original study but with more details. We also constructed an online server for ImmuCellAI-mouse, on which users can upload expression matrices for analysis. ImmuCellAI-mouse will be a useful tool for studying the immune microenvironment, cancer immunology and immunotherapy in mouse models, providing an indispensable supplement for human disease studies. AVAILABILITY AND IMPLEMENTATION Software is available at http://bioinfo.life.hust.edu.cn/ImmuCellAI-mouse/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ya-Ru Miao
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mengxuan Xia
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mei Luo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tao Luo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mei Yang
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - An-Yuan Guo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
92
|
D'Amico F, Barone M, Tavella T, Rampelli S, Brigidi P, Turroni S. Host microbiomes in tumor precision medicine: how far are we? Curr Med Chem 2022; 29:3202-3230. [PMID: 34986765 DOI: 10.2174/0929867329666220105121754] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/13/2021] [Accepted: 11/22/2021] [Indexed: 11/22/2022]
Abstract
The human gut microbiome has received a crescendo of attention in recent years, due to the countless influences on human pathophysiology, including cancer. Research on cancer and anticancer therapy is constantly looking for new hints to improve the response to therapy while reducing the risk of relapse. In this scenario, the gut microbiome and the plethora of microbial-derived metabolites are considered a new opening in the development of innovative anticancer treatments for a better prognosis. This narrative review summarizes the current knowledge on the role of the gut microbiome in the onset and progression of cancer, as well as in response to chemo-immunotherapy. Recent findings regarding the tumor microbiome and its implications for clinical practice are also commented on. Current microbiome-based intervention strategies (i.e., prebiotics, probiotics, live biotherapeutics and fecal microbiota transplantation) are then discussed, along with key shortcomings, including a lack of long-term safety information in patients who are already severely compromised by standard treatments. The implementation of bioinformatic tools applied to microbiomics and other omics data, such as machine learning, has an enormous potential to push research in the field, enabling the prediction of health risk and therapeutic outcomes, for a truly personalized precision medicine.
Collapse
Affiliation(s)
- Federica D'Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Monica Barone
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Teresa Tavella
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Simone Rampelli
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Patrizia Brigidi
- Microbiome Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| |
Collapse
|
93
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2022; 172:xiii-xxiii. [PMID: 36064230 PMCID: PMC10087864 DOI: 10.1016/s0091-679x(22)00139-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
94
|
Avdoshina DV, Kondrashova AS, Belikova MG, Bayurova EO. Murine Models of Chronic Viral Infections and Associated Cancers. Mol Biol 2022; 56:649-667. [PMID: 36217336 PMCID: PMC9534466 DOI: 10.1134/s0026893322050028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/07/2022]
Abstract
Viruses are now recognized as bona fide etiologic factors of human cancer. Carcinogenic viruses include Epstein– Barr virus (EBV), high-risk human papillomaviruses (HPVs), hepatitis B virus (HBV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), human immunodeficiency virus type 1 (HIV-1, indirectly), and several candidate human cancer viruses. It is estimated that 15% of all human tumors worldwide are caused by viruses. Tumor viruses establish long-term persistent infections in humans, and cancer is an accidental side effect of viral replication strategies. Viruses are usually not complete carcinogens, supporting the concept that cancer results from the accumulation of multiple cooperating events, in which human cancer viruses display different, often opposing roles. The laboratory mouse Mus musculus is one of the best in vivo experimental systems for modeling human pathology, including viral infections and cancer. However, mice are unsusceptible to infection with the known carcinogenic viruses. Many murine models were developed to overcome this limitation and to address various aspects of virus-associated carcinogenesis, from tumors resulting from xenografts of human tissues and cells, including cancerous and virus infected, to genetically engineered mice susceptible to viral infections and associated cancer. The review considers the main existing models, analyzes their advantages and drawbacks, describes their applications, outlines the prospects of their further development.
Collapse
Affiliation(s)
- D. V. Avdoshina
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia
| | - A. S. Kondrashova
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia
| | - M. G. Belikova
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia ,Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia ,Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| | - E. O. Bayurova
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia ,Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| |
Collapse
|
95
|
Ni S, Chen R, Hu K. Experimental murine models of brainstem gliomas. Drug Discov Today 2021; 27:1218-1235. [PMID: 34954326 DOI: 10.1016/j.drudis.2021.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/16/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022]
Abstract
As an intractable central nervous system (CNS) tumor, brainstem gliomas (BGs) are one of the leading causes of pediatric death by brain tumors. Owing to the risk of surgical resection and the little improvement in survival time after radiotherapy and chemotherapy, there is an urgent need to find reliable model systems to better understand the regional pathogenesis of the brainstem and improve treatment strategies. In this review, we outline the evolution of BG murine models, and discuss both their advantages and limitations in drug discovery.
Collapse
Affiliation(s)
- Shuting Ni
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rujing Chen
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kaili Hu
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
96
|
Graveline R, Haida M, Dumont C, Poulin D, Poitout-Belissent F, Samadfam R, Kronenberg S, Regenass-Lechner F, Prell R, Piche MS. Development of a nonhuman primate challenge model to evaluate CD8 + T cell responses to an adenovirus-based vaccine expressing SIV proteins upon repeat-dose treatment with checkpoint inhibitors. MAbs 2021; 14:1979447. [PMID: 34923919 PMCID: PMC8726661 DOI: 10.1080/19420862.2021.1979447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Targeting immune checkpoint receptors expressed in the T cell synapse induces active and long-lasting antitumor immunity in preclinical tumor models and oncology patients. However, traditional nonhuman primate (NHP) studies in healthy animals have thus far demonstrated little to no pharmacological activity or toxicity for checkpoint inhibitors (CPIs), likely due to a quiescent immune system. We developed a NHP vaccine challenge model in Mauritius cynomolgus monkey (MCMs) that elicits a strong CD8+ T cell response to assess both pharmacology and safety within the same animal. MHC I-genotyped MCMs were immunized with three replication incompetent adenovirus serotype 5 (Adv5) encoding Gag, Nef and Pol simian immunodeficiency virus (SIV) proteins administered 4 weeks apart. Immunized animals received the anti-PD-L1 atezolizumab or an immune checkpoint-targeting bispecific antibody (mAbX) in early development. After a single immunization, Adv5-SIVs induced T-cell activation as assessed by the expression of several co-stimulatory and co-inhibitory molecules, proliferation, and antigen-specific T-cell response as measured by a Nef-dependent interferon-γ ELIspot and tetramer analysis. Administration of atezolizumab increased the number of Ki67+ CD8+ T cells, CD8+ T cells co-expressing TIM3 and LAG3 and the number of CD4+ T cells co-expressing 4–1BB, BTLA, and TIM3 two weeks after vaccination. Both atezolizumab and mAbX extended the cytolytic activity of the SIV antigen-specific CD8+ T cell up to 8 weeks. Taken together, this vaccine challenge model allowed the combined study of pharmacology and safety parameters for a new immunomodulatory protein-based therapeutic targeting CD8+ T cells in an NHP model.
Collapse
Affiliation(s)
| | - Morad Haida
- Immunology, Charles River Laboratories, Senneville, Canada
| | | | - Dominic Poulin
- Immunology, Charles River Laboratories, Senneville, Canada
| | | | - Rana Samadfam
- Immunology, Charles River Laboratories, Senneville, Canada
| | - Sven Kronenberg
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Franziska Regenass-Lechner
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Rodney Prell
- Safety Assessment, Development Sciences, Genentech, South San Francisco, CA, USA
| | | |
Collapse
|
97
|
Freund E, Bekeschus S. Gas Plasma-Oxidized Liquids for Cancer Treatment: Preclinical Relevance, Immuno-Oncology, and Clinical Obstacles. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2021. [DOI: 10.1109/trpms.2020.3029982] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
98
|
Schetters STT, Rodriguez E, Kruijssen LJW, Crommentuijn MHW, Boon L, Van den Bossche J, Den Haan JMM, Van Kooyk Y. Monocyte-derived APCs are central to the response of PD1 checkpoint blockade and provide a therapeutic target for combination therapy. J Immunother Cancer 2021; 8:jitc-2020-000588. [PMID: 32690667 PMCID: PMC7371367 DOI: 10.1136/jitc-2020-000588] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2020] [Indexed: 12/23/2022] Open
Abstract
Background PD1 immune checkpoint blockade (αPD1 ICB) has shown unparalleled success in treating many types of cancer. However, response to treatment does not always lead to tumor rejection. While αPD1 ICB relies on cytotoxic CD8+ T cells, antigen-presenting cells (APCs) at the tumor site are also needed for costimulation of tumor-infiltrating lymphocytes (TILs). It is still unclear how these APCs develop and function before and during αPD1 ICB or how they are associated with tumor rejection. Methods Here, we used B16 mouse melanoma and MC38 colorectal carcinoma tumor models, which show differential responses to αPD1 ICB. The immune composition of ICB insensitive B16 and sensitive MC38 were extensively investigated using multi-parameter flow cytometry and unsupervised clustering and trajectory analyses. We additionally analyzed existing single cell RNA sequencing data of the myeloid compartment of patients with melanoma undergoing αPD1 ICB. Lastly, we investigated the effect of CD40 agonistic antibody on the tumor-infiltrating monocyte-derived cells during αPD1 ICB. Results We show that monocyte-derived dendritic cells (moDCs) express high levels of costimulatory molecules and are correlated with effector TILs in the tumor microenvironment (TME) after αPD1 ICB only in responding mouse tumor models. Tumor-resident moDCs showed distinct differentiation from monocytes in both mouse and human tumors. We further confirmed significant enrichment of tumor-resident differentiated moDCs in patients with melanoma responding to αPD1 ICB therapy compared with non-responding patients. Moreover, moDCs could be targeted by agonistic anti-CD40 antibody, supporting moDC differentiation, effector T-cell expansion and anti-tumor immunity. Conclusion The combined analysis of myeloid and lymphoid populations in the TME during successful and non-successful PD1 ICB led to the discovery of monocyte-to-DC differentiation linked to expanding T-cell populations. This differentiation was found in patients during ICB, which was significantly higher during successful ICB. The finding of tumor-infiltrating monocytes and differentiating moDCs as druggable target for rational combination therapy opens new avenues of anti-tumor therapy design.
Collapse
Affiliation(s)
- Sjoerd T T Schetters
- Molecular Cell Biology and Immunology, Amsterdam Institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC - Location VUMC, Amsterdam, The Netherlands
| | - Ernesto Rodriguez
- Molecular Cell Biology and Immunology, Amsterdam Institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC - Location VUMC, Amsterdam, The Netherlands
| | - Laura J W Kruijssen
- Molecular Cell Biology and Immunology, Amsterdam Institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC - Location VUMC, Amsterdam, The Netherlands
| | - Matheus H W Crommentuijn
- Molecular Cell Biology and Immunology, Amsterdam Institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC - Location VUMC, Amsterdam, The Netherlands
| | - Louis Boon
- Polpharma Biologics, Utrecht, The Netherlands
| | - Jan Van den Bossche
- Molecular Cell Biology and Immunology, Amsterdam Institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC - Location VUMC, Amsterdam, The Netherlands
| | - Joke M M Den Haan
- Molecular Cell Biology and Immunology, Amsterdam Institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC - Location VUMC, Amsterdam, The Netherlands
| | - Yvette Van Kooyk
- Molecular Cell Biology and Immunology, Amsterdam Institute for Infection and Immunity, Cancer Center Amsterdam, Amsterdam UMC - Location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
99
|
Application of LDH assay for therapeutic efficacy evaluation of ex vivo tumor models. Sci Rep 2021; 11:18571. [PMID: 34535719 PMCID: PMC8448883 DOI: 10.1038/s41598-021-97894-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022] Open
Abstract
The current standard preclinical oncology models are not able to fully recapitulate therapeutic targets and clinically relevant disease biology, evidenced by the 90% attrition rate of new therapies in clinical trials. Three-dimensional (3D) culture systems have the potential to enhance the relevance of preclinical models. However, the limitations of currently available cellular assays to accurately evaluate therapeutic efficacy in these models are hindering their widespread adoption. We assessed the compatibility of the lactate dehydrogenase (LDH) assay in 3D spheroid cultures against other commercially available readout methods. We developed a standardized protocol to apply the LDH assay to ex vivo cultures, considering the impact of culture growth dynamics. We show that accounting for growth rates and background release levels of LDH are sufficient to make the LDH assay a suitable methodology for longitudinal monitoring and endpoint assessment of therapeutic efficacy in both cell line-derived xenografts (xenospheres) and patient-derived explant cultures. This method has the added value of being non-destructive and not dependent on reagent penetration or manipulation of the parent material. The establishment of reliable readout methods for complex 3D culture systems will further the utility of these tumor models in preclinical and co-clinical drug development studies.
Collapse
|
100
|
Bule P, Aguiar SI, Aires-Da-Silva F, Dias JNR. Chemokine-Directed Tumor Microenvironment Modulation in Cancer Immunotherapy. Int J Mol Sci 2021; 22:9804. [PMID: 34575965 PMCID: PMC8464715 DOI: 10.3390/ijms22189804] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022] Open
Abstract
Chemokines are a large family of small chemotactic cytokines that coordinates immune cell trafficking. In cancer, they have a pivotal role in the migration pattern of immune cells into the tumor, thereby shaping the tumor microenvironment immune profile, often towards a pro-tumorigenic state. Furthermore, chemokines can directly target non-immune cells in the tumor microenvironment, including cancer, stromal and vascular endothelial cells. As such, chemokines participate in several cancer development processes such as angiogenesis, metastasis, cancer cell proliferation, stemness and invasiveness, and are therefore key determinants of disease progression, with a strong influence in patient prognosis and response to therapy. Due to their multifaceted role in the tumor immune response and tumor biology, the chemokine network has emerged as a potential immunotherapy target. Under the present review, we provide a general overview of chemokine effects on several tumoral processes, as well as a description of the currently available chemokine-directed therapies, highlighting their potential both as monotherapy or in combination with standard chemotherapy or other immunotherapies. Finally, we discuss the most critical challenges and prospects of developing targeted chemokines as therapeutic options.
Collapse
Affiliation(s)
| | | | | | - Joana Nunes Ribeiro Dias
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal; (P.B.); (S.I.A.); (F.A.-D.-S.)
| |
Collapse
|