1
|
Li X, Jian J, Zhang A, Xiang JM, Huang J, Chen Y. The role of immune cells and immune related genes in the tumor microenvironment of papillary thyroid cancer and their significance for immunotherapy. Sci Rep 2024; 14:18125. [PMID: 39103463 PMCID: PMC11300445 DOI: 10.1038/s41598-024-69187-9] [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: 03/07/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common pathological type of thyroid cancer (THCA) and shows a better prognosis than other types. However, further research is needed to determine the risk of PTC. We herein used the CIBERSORT algorithm to analyze the gene-expression profile obtained from TCGA, estimated the infiltration ratio of 22 immune cell types in tumor tissues and normal tissues, analyzed the differential expression of immune-related genes, and identified immune cells and immune-related genes related to clinical progress and prognosis. We uncovered 12 immune cell types and nine immune-related genes that were closely correlated with TNM staging, and two immune cell types (activated NK cells and γδT cells) and one immune-related gene (CD40LG) that were associated with prognosis. After evaluation, four immune cell types could be used to determine low-risk PTC, with six immune cell types and six immune-related genes closely associated with high-risk PTC. The type and quantity of infiltrating immune cells in the microenvironment of PTC, as well as immune-related genes, appear to be closely related to tumor progression and can therefore be used as important indicators for the evaluation of patient prognosis. We posit that the study of immune cells and immune-related genes in the tumor microenvironment will facilitate the determination of low-risk PTC more accurately, and that this will greatly promote the development of high-risk PTC immunotherapy.
Collapse
Affiliation(s)
- Xumei Li
- Department of Pathology, Chongqing Changshou District Maternal and Child Health Hospital, Chongqing, China
| | - Jie Jian
- Department of Pathology, Chongqing Changshou District Maternal and Child Health Hospital, Chongqing, China
| | - Anzhi Zhang
- Department of Pathology, Jiaxing University Affiliated Women and Children Hospital (Jiaxing Maternity and Child Health Care Hospital), Jiaxing University, Jiaxing, China
| | - Jiang Ming Xiang
- Department of Surgery, Chongqing Changshou District Maternal and Child Health Hospital, Chongqing, China
| | - Jingjing Huang
- Department of Surgery, Chongqing Changshou District Maternal and Child Health Hospital, Chongqing, China
| | - Yanlin Chen
- Department of Pathology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
A phase I study of an adenoviral vector delivering a MUC1/CD40-ligand fusion protein in patients with advanced adenocarcinoma. Nat Commun 2022; 13:6453. [PMID: 36307410 PMCID: PMC9616917 DOI: 10.1038/s41467-022-33834-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 10/05/2022] [Indexed: 12/25/2022] Open
Abstract
Cancer vaccines as immunotherapy for solid tumours are currently in development with promising results. We report a phase 1 study of Ad-sig-hMUC1/ecdCD40L (NCT02140996), an adenoviral-vector vaccine encoding the tumour-associated antigen MUC1 linked to CD40 ligand, in patients with advanced adenocarcinoma. The primary objective of this study is safety and tolerability. We also study the immunome in vaccinated patients as a secondary outcome. This trial, while not designed to determine clinical efficacy, reports an exploratory endpoint of overall response rate. The study meets its pre-specified primary endpoint demonstrating safety and tolerability in a cohort of 21 patients with advanced adenocarcinomas (breast, lung and ovary). The maximal dose of the vaccine is 1 ×1011 viral particles, with no dose limiting toxicities. All drug related adverse events are of low grades, most commonly injection site reactions in 15 (71%) patients. Using exploratory high-dimensional analyses, we find both quantitative and relational changes in the cancer immunome after vaccination. Our data highlights the utility of high-dimensional analyses in understanding and predicting effective immunotherapy, underscoring the importance of immune competency in cancer prognosis.
Collapse
|
3
|
Biegert GWG, Rosewell Shaw A, Suzuki M. Current development in adenoviral vectors for cancer immunotherapy. Mol Ther Oncolytics 2021; 23:571-581. [PMID: 34938857 DOI: 10.1016/j.omto.2021.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Adenoviruses are well characterized and thus easily modified to generate oncolytic vectors that directly lyse tumor cells and can be "armed" with transgenes to promote lysis, antigen presentation, and immunostimulation. Oncolytic adenoviruses (OAds) are safe, versatile, and potent immunostimulants in patients. Since transgene expression is restricted to the tumor, adenoviral transgenes overcome the toxicities and short half-life of systemically administered cytokines, immune checkpoint blockade molecules, and bispecific T cell engagers. While OAds expressing immunostimulatory molecules ("armed" OAds) have demonstrated anti-tumor potential in preclinical solid tumor models, the efficacy has not translated into significant clinical outcomes as a monotherapy. However, OAds synergize with established standards of care and novel immunotherapeutic agents, providing a multifaceted means to address complexities associated with solid tumors. Critically, armed OAds revitalize endogenous and adoptively transferred immune cells while simultaneously enhancing their anti-tumor function. To properly evaluate these novel vectors and reduce the gap in the cycle between bench-to-bedside and back, improving model systems must be a priority. The future of OAds will involve a multidimensional approach that provides immunostimulatory molecules, immune checkpoint blockade, and/or immune engagers in concert with endogenous and exogenous immune cells to initiate durable and comprehensive anti-tumor responses.
Collapse
Affiliation(s)
- Greyson Willis Grossman Biegert
- Department of Medicine, Section of Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| | - Amanda Rosewell Shaw
- Department of Medicine, Section of Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| | - Masataka Suzuki
- Department of Medicine, Section of Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
4
|
Irenaeus S, Hellström V, Wenthe J, Krause J, Sundin A, Ahlström H, Tufveson G, Tötterman TH, Loskog A, Ullenhag GJ. Intratumoral immunostimulatory AdCD40L gene therapy in patients with advanced solid tumors. Cancer Gene Ther 2020; 28:1188-1197. [PMID: 33318679 DOI: 10.1038/s41417-020-00271-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/07/2020] [Accepted: 11/23/2020] [Indexed: 11/09/2022]
Abstract
AdCD40L is a replication-deficient virus carrying the gene for CD40 ligand which has previously been evaluated in patients with urothelial cancer and malignant melanoma. Herein, we present the results of repeated intratumoral injections of AdCD40L in seven patients with metastatic solid cancer. One patient who developed urothelial cancer derived from a renal transplant was treated with repeated injections of AdCD40L alone. The remaining patients suffered from cholangiocarcinoma, kidney, breast, rectal, or ovarian cancer and received AdCD40L repeatedly (4x) in combination with cyclophosphamide. The treatment was safe and generally well-tolerated. Two patients had clinical benefit of the treatment and one of them was accepted for re-treatment. Circulating proinflammatory cytokines were commonly increased after treatment, but save for TNFα, significances were not reached which could be due to the low number of patients. Similar to earlier findings in AdCD40L-treated melanoma patients, IL8 plasma levels were high in the present study. In conclusion, gene therapy by repeated intratumoral AdCD40L injections alone, or in combination with cyclophosphamide, is feasible and safe in patients with solid cancers. The potential of intratumoral CD40L gene transfer as treatment of cancer was illustrated by the clinical improvement in two out of seven patients.
Collapse
Affiliation(s)
- Sandra Irenaeus
- Department of Immunology, Genetics and Pathology, Science of Life Laboratory, Uppsala University, Dag Hammarskjolds vag 20, 751 85, Uppsala, Sweden.,Department of Oncology, Uppsala University Hospital, 751 85, Uppsala, Sweden
| | - Vivan Hellström
- Department of Surgical Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | - Jessica Wenthe
- Department of Immunology, Genetics and Pathology, Science of Life Laboratory, Uppsala University, Dag Hammarskjolds vag 20, 751 85, Uppsala, Sweden
| | - Johan Krause
- Department of Surgical Sciences, Uppsala University, 751 85, Uppsala, Sweden.,Division of Radiology, Uppsala University Hospital, 751 85, Uppsala, Sweden
| | - Anders Sundin
- Department of Surgical Sciences, Uppsala University, 751 85, Uppsala, Sweden.,Division of Radiology, Uppsala University Hospital, 751 85, Uppsala, Sweden
| | - Håkan Ahlström
- Department of Surgical Sciences, Uppsala University, 751 85, Uppsala, Sweden.,Division of Radiology, Uppsala University Hospital, 751 85, Uppsala, Sweden
| | - Gunnar Tufveson
- Department of Surgical Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | - Thomas H Tötterman
- Department of Immunology, Genetics and Pathology, Science of Life Laboratory, Uppsala University, Dag Hammarskjolds vag 20, 751 85, Uppsala, Sweden
| | - Angelica Loskog
- Department of Immunology, Genetics and Pathology, Science of Life Laboratory, Uppsala University, Dag Hammarskjolds vag 20, 751 85, Uppsala, Sweden
| | - Gustav J Ullenhag
- Department of Immunology, Genetics and Pathology, Science of Life Laboratory, Uppsala University, Dag Hammarskjolds vag 20, 751 85, Uppsala, Sweden. .,Department of Oncology, Uppsala University Hospital, 751 85, Uppsala, Sweden.
| |
Collapse
|
5
|
Kochneva GV, Sivolobova GF, Tkacheva AV, Gorchakov AA, Kulemzin SV. Combination of Oncolytic Virotherapy and CAR T/NK Cell Therapy for the Treatment of Cancer. Mol Biol 2020; 54:3-16. [DOI: 10.1134/s0026893320010100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 03/19/2019] [Indexed: 12/28/2022]
|
6
|
Governa V, Brittoli A, Mele V, Pinamonti M, Terracciano L, Muenst S, Iezzi G, Spagnoli GC, Zajac P, Trella E. A replication-incompetent CD154/40L recombinant vaccinia virus induces direct and macrophage-mediated antitumor effects in vitro and in vivo. Oncoimmunology 2019; 8:e1568162. [PMID: 31069131 PMCID: PMC6492963 DOI: 10.1080/2162402x.2019.1568162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/30/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022] Open
Abstract
CD40 triggering may result in antitumor effects of potentially high clinical relevance. To gain insights important for patient selection and to identify adequate targeting techniques, we investigated CD40 expression in human cancer tissues and generated a replication-incompetent recombinant vaccinia virus expressing CD40 ligand (rVV40L). Its effects were explored in vitro and in vivo upon direct CD40 targeting on malignant cells or macrophage activation. CD40 expression was analyzed by immunohistochemistry in tumor and stromal cells in a multi-tumor array including 836 specimens from 27 different tumor types. Established tumor cell lines were used to explore the capacity of rVV40L to induce malignant cell apoptosis and modulate functional profiles of polarized macrophages. CD40 expression was detectable in significantly higher numbers of stromal as compared to malignant cells in lung and breast cancers. CD40 ligation following rVV40L infection induced apoptosis in CD40(+) cancer cells, but only in the presence of intact specific signal transduction chain. Importantly, rVV40L infection promoted the induction of TNF-α-dependent antitumor activity of M1-like macrophages directed against CD40(-) targets. CD40-activated M1-like macrophages also displayed enhanced ability to CXCL10-dependently recruit CD8+ T cells and to efficiently present cancer cell intracellular antigens through cross-priming. Moreover, rVV-driven CD40L expression partially “re-educated” M2-like macrophages, as suggested by detectable CXCL10 and IL-12 production. Most importantly, we observed that intra-tumoral injection of rVV40L-infected human macrophages inhibits progression of human CD40(-) tumors in vivo. First evidences of anticancer activity of rVV40L strongly encourage further evaluations.
Collapse
Affiliation(s)
- Valeria Governa
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Alvaro Brittoli
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Valentina Mele
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Maurizio Pinamonti
- Unit of Pathology, Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Luigi Terracciano
- Institute of Pathology, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Simone Muenst
- Institute of Pathology, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Giandomenica Iezzi
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Department of Surgery, Ente Ospedaliero Cantonale and Università della Svizzera Italiana, Lugano, Switzerland
| | - Giulio Cesare Spagnoli
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Consiglio Nazionale delle Ricerche, Institute of Translational Pharmacology, Rome, Italy
| | - Paul Zajac
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Emanuele Trella
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
7
|
Irenaeus SMM, Nielsen D, Ellmark P, Yachnin J, Deronic A, Nilsson A, Norlén P, Veitonmäki N, Wennersten CS, Ullenhag GJ. First-in-human study with intratumoral administration of a CD40 agonistic antibody, ADC-1013, in advanced solid malignancies. Int J Cancer 2019; 145:1189-1199. [PMID: 30664811 DOI: 10.1002/ijc.32141] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/17/2018] [Accepted: 01/02/2019] [Indexed: 12/20/2022]
Abstract
Agonistic CD40 antibodies activate dendritic cells and can expand and activate tumor-specific T cells. Our purpose was to assess the CD40 agonistic antibody ADC-1013 in the clinical setting including intratumoral administration since preclinical studies have indicated that intratumoral is better than intravenous administration. A Phase I, open label, multicenter study was conducted in patients with advanced solid tumors who had received established treatments. A modified 3 + 3 dose-escalation was applied (every other week dosing). Twenty-three patients were treated with ADC-1013 intratumorally (dosing from 22.5 μg/kg up to 400 μg/kg) or intravenously (dosing at 75 μg/kg). The pharmacodynamic effects observed in the patients were further verified in an hCD40tg mouse model. Adverse events were mostly Common Terminology Criteria for Adverse Events (CTCAE) Grades 1 or 2 and transient. The serum concentration ADC-1013 and cytokine release (MCP-1, TNFα and IL-6) were more pronounced in patients receiving injections in deep metastases compared to patients receiving injections in superficial metastases. Treatment with ADC-1013 resulted in a marked decrease in B cell levels in peripheral blood after 24 h while remaining B cells significantly increased their expression of the cell surface activation marker CD86. Activation of antigen-presenting cells and subsequent activation of T cells were demonstrated in hCD40tg mice. Moreover, ADC-1013 treatment in this mouse model acted synergistically with a PD-1 inhibitor. The results from the first-in-human study of ADC-1013 indicate that intratumoral administration of ADC-1013 into superficial lesions is well tolerated at clinically relevant doses and associated with pharmacodynamic responses.
Collapse
Affiliation(s)
- Sandra M M Irenaeus
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Dorte Nielsen
- Department of Oncology, Herlev-Gentofte Hospital, Herlev, Denmark
| | - Peter Ellmark
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Jeffrey Yachnin
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Adnan Deronic
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | - Per Norlén
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | | | - Gustav J Ullenhag
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
8
|
Guedan S, Alemany R. CAR-T Cells and Oncolytic Viruses: Joining Forces to Overcome the Solid Tumor Challenge. Front Immunol 2018; 9:2460. [PMID: 30405639 PMCID: PMC6207052 DOI: 10.3389/fimmu.2018.02460] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/04/2018] [Indexed: 12/28/2022] Open
Abstract
Adoptive transfer of chimeric antigen receptor (CAR)-modified T cells has resulted in unprecedented rates of long-lasting complete responses in patients with leukemia and lymphoma. However, despite the impressive results in patients with hematologic malignancies, CAR-T cells have showed limited effect against solid cancers. New approaches will need to simultaneously overcome the multiple challenges that CAR-T cells encounter in solid tumors, including the immunosuppressive tumor microenvironment and heterogeneity of antigen expression. Oncolytic viruses are lytic and immunogenic anti-cancer agents with the potential to synergize with CAR-T cells for the treatment of solid tumors. In addition, viruses can be further modified to deliver therapeutic transgenes selectively to the tumor microenvironment, which could enhance the effector functions of tumor-specific T cells. This review summarizes the major limitations of CAR-T cells in solid tumors and discusses the potential role for oncolytic viruses as partners for CAR-T cells in the fight against cancer.
Collapse
Affiliation(s)
- Sonia Guedan
- Department of Hematology and Oncology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Ramon Alemany
- ProCure Program, IDIBELL-Institut Catala d'Oncologia, L'Hospitalet de Llobregat, Spain
| |
Collapse
|
9
|
Park GT, Choi KC. Advanced new strategies for metastatic cancer treatment by therapeutic stem cells and oncolytic virotherapy. Oncotarget 2018; 7:58684-58695. [PMID: 27494901 PMCID: PMC5295462 DOI: 10.18632/oncotarget.11017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 05/29/2016] [Indexed: 01/14/2023] Open
Abstract
The field of therapeutic stem cell and oncolytic virotherapy for cancer treatment has rapidly expanded over the past decade. Oncolytic viruses constitute a promising new class of anticancer agent because of their ability to selectively infect and destroy tumor cells. Engineering of viruses to express anticancer genes and specific cancer targeting molecules has led to the use of these systems as a novel platform of metastatic cancer therapy. In addition, stem cells have a cancer specific migratory capacity, which is available for metastatic cancer targeting. Prodrug activating enzyme or anticancer cytokine expressing stem cells successfully inhibited the proliferation of cancer cells. Preclinical models have clearly demonstrated anticancer activity of these two platforms against a number of different cancer types and metastatic cancer. Several systems using therapeutic stem cells or oncolytic virus have entered clinical trials, and promising results have led to late stage clinical development. Consequently, metastatic cancer therapies using stem cells and oncolytic viruses are extremely promising. The following review will focus on the metastatic cancer targeting mechanism of therapeutic stem cells and oncolytic viruses, and potential challenges ahead for advancing the field.
Collapse
Affiliation(s)
- Geon-Tae Park
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.,TheraCell Bio & Science, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
10
|
Ajina A, Maher J. Prospects for combined use of oncolytic viruses and CAR T-cells. J Immunother Cancer 2017; 5:90. [PMID: 29157300 PMCID: PMC5696728 DOI: 10.1186/s40425-017-0294-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/17/2017] [Indexed: 12/18/2022] Open
Abstract
With the approval of talimogene laherparepvec (T-VEC) for inoperable locally advanced or metastatic malignant melanoma in the USA and Europe, oncolytic virotherapy is now emerging as a viable therapeutic option for cancer patients. In parallel, following the favourable results of several clinical trials, adoptive cell transfer using chimeric antigen receptor (CAR)-redirected T-cells is anticipated to enter routine clinical practice for the management of chemotherapy-refractory B-cell malignancies. However, CAR T-cell therapy for patients with advanced solid tumours has proved far less successful. This Review draws upon recent advances in the design of novel oncolytic viruses and CAR T-cells and provides a comprehensive overview of the synergistic potential of combination oncolytic virotherapy with CAR T-cell adoptive cell transfer for the management of solid tumours, drawing particular attention to the methods by which recombinant oncolytic viruses may augment CAR T-cell trafficking into the tumour microenvironment, mitigate or reverse local immunosuppression and enhance CAR T-cell effector function and persistence.
Collapse
Affiliation(s)
- Adam Ajina
- Department of Oncology, Royal Free London NHS Foundation Trust, London, UK
| | - John Maher
- King’s College London, CAR Mechanics Group, School of Cancer and Pharmaceutical Sciences, Guy’s Hospital Campus, Great Maze Pond, London, SE1 9RT UK
- Department of Clinical Immunology and Allergy, King’s College Hospital NHS Foundation Trust, London, UK
- Department of Immunology, Eastbourne Hospital, East Sussex, UK
| |
Collapse
|
11
|
Trella E, Raafat N, Mengus C, Traunecker E, Governa V, Heidtmann S, Heberer M, Oertli D, Spagnoli GC, Zajac P. CD40 ligand-expressing recombinant vaccinia virus promotes the generation of CD8(+) central memory T cells. Eur J Immunol 2015; 46:420-31. [PMID: 26561341 DOI: 10.1002/eji.201545554] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 10/05/2015] [Accepted: 11/05/2015] [Indexed: 01/17/2023]
Abstract
Central memory CD8(+) T cells (TCM ) play key roles in the protective immunity against infectious agents, cancer immunotherapy, and adoptive treatments of malignant and viral diseases. CD8(+) TCM cells are characterized by specific phenotypes, homing, and proliferative capacities. However, CD8(+) TCM -cell generation is challenging, and usually requires CD4(+) CD40L(+) T-cell "help" during the priming of naïve CD8(+) T cells. We have generated a replication incompetent CD40 ligand-expressing recombinant vaccinia virus (rVV40L) to promote the differentiation of human naïve CD8(+) T cells into TCM specific for viral and tumor-associated antigens. Soluble CD40 ligand recombinant protein (sCD40L), and vaccinia virus wild-type (VV WT), alone or in combination, were used as controls. Here, we show that, in the absence of CD4(+) T cells, a single "in vitro" stimulation of naïve CD8(+) T cells by rVV40L-infected nonprofessional CD14(+) antigen presenting cells promotes the rapid generation of viral or tumor associated antigen-specific CD8(+) T cells displaying TCM phenotypic and functional properties. These observations demonstrate the high ability of rVV40L to fine tune CD8(+) mediated immune responses, and strongly support the use of similar reagents for clinical immunization and adoptive immunotherapy purposes.
Collapse
Affiliation(s)
- Emanuele Trella
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Nermin Raafat
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland.,Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Chantal Mengus
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Valeria Governa
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Michael Heberer
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Daniel Oertli
- Department of Surgery, University Hospital, Basel, Switzerland
| | - Giulio C Spagnoli
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Paul Zajac
- Institute of Surgical Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
12
|
8th International Conference on Oncolytic Virus Therapeutics 2014 • April 10–13, 2014Lincoln College & Examination Schools • Oxford, United Kingdom. Hum Gene Ther 2014. [DOI: 10.1089/hum.2014.2538.abstracts] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
13
|
Xu W, Xu Y, Wei Y, Tan Y, Zhao H, Zhao W, Wu J. Self-complementary adeno-associated virus 5-mediated gene transduction of a novel CD40L mutant confers direct antitumor effects in lung carcinoma. Mol Med Rep 2014; 11:482-8. [PMID: 25352298 DOI: 10.3892/mmr.2014.2765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 09/24/2014] [Indexed: 11/06/2022] Open
Abstract
CD40 ligand (CD40L) gene therapy offers a potentially useful option for lung cancer due to its multiple antitumor activities. However, membrane-bound CD40L may be proteolytically cleaved to form soluble CD40L (sCD40L), which results in adverse effects. In a previous study by our group, it was demonstrated that recombinant self-complementary adeno-associated virus 5 (scAAV5) efficiently delivered genes to lung cancer cells. In the present study, an scAAV5 expressing a non-cleavable human CD40L mutant (scAAV5-CD40L-M) was generated and its direct antitumor effects in lung cancer were evaluated. Transduction with scAAV5-CD40L-M resulted in effective expression of CD40L on the cell surface with low levels of cleaved sCD40L, which significantly reduced the percentage of viable cells and promoted caspase-3-dependent apoptosis of CD40-positive lung carcinoma A549 cells, compared with scAAV5-CD40L transduction (P<0.05). Furthermore, treatment with scAAV5-CD40L-M exerted a significant antitumor effect against CD40-positive A549 xenografts by inducing apoptosis (P<0.05) with few side effects. Gene therapy using an scAAV5 vector expressing non-cleavable human CD40L mutant may therefore have direct antitumor effects against CD40-positive lung cancers. These tumoricidal effects of scAAV5-CD40L-M treatment make it a promising therapeutic technique for the treatment of lung cancer.
Collapse
Affiliation(s)
- Wei Xu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yuanyuan Xu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yunyan Wei
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yaoxi Tan
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hongye Zhao
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Weihong Zhao
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jianqing Wu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
14
|
Zhu D, Chen C, Purwanti YI, Du S, Lam DH, Wu C, Zeng J, Toh HC, Wang S. Induced Pluripotent Stem Cell-Derived Neural Stem Cells Transduced with Baculovirus Encoding CD40 Ligand for Immunogene Therapy in Mouse Models of Breast Cancer. Hum Gene Ther 2014; 25:747-58. [DOI: 10.1089/hum.2013.160] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Detu Zhu
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Can Chen
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Yovita Ida Purwanti
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Shouhui Du
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Dang Hoang Lam
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Chunxiao Wu
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | - Jieming Zeng
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| | | | - Shu Wang
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Institute of Bioengineering and Nanotechnology, Singapore 138669
| |
Collapse
|
15
|
Purwanti YI, Chen C, Lam DH, Wu C, Zeng J, Fan W, Wang S. Antitumor effects of CD40 ligand-expressing endothelial progenitor cells derived from human induced pluripotent stem cells in a metastatic breast cancer model. Stem Cells Transl Med 2014; 3:923-35. [PMID: 24972599 DOI: 10.5966/sctm.2013-0140] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Given their intrinsic ability to home to tumor sites, endothelial progenitor cells (EPCs) are attractive as cellular vehicles for targeted cancer gene therapy. However, collecting sufficient EPCs is one of the challenging issues critical for effective clinical translation of this new approach. In this study, we sought to explore whether human induced pluripotent stem (iPS) cells could be used as a reliable and accessible cell source to generate human EPCs suitable for cancer treatment. We used an embryoid body formation method to derive CD133(+)CD34(+) EPCs from human iPS cells. The generated EPCs expressed endothelial markers such as CD31, Flk1, and vascular endothelial-cadherin without expression of the CD45 hematopoietic marker. After intravenous injection, the iPS cell-derived EPCs migrated toward orthotopic and lung metastatic tumors in the mouse 4T1 breast cancer model but did not promote tumor growth and metastasis. To investigate their therapeutic potential, the EPCs were transduced with baculovirus encoding the potent T cell costimulatory molecule CD40 ligand. The systemic injection of the CD40 ligand-expressing EPCs stimulated the secretion of both tumor necrosis factor-α and interferon-γ and increased the caspase 3/7 activity in the lungs with metastatic tumors, leading to prolonged survival of the tumor bearing mice. Therefore, our findings suggest that human iPS cell-derived EPCs have the potential to serve as tumor-targeted cellular vehicles for anticancer gene therapy.
Collapse
Affiliation(s)
- Yovita Ida Purwanti
- Department of Biological Sciences, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Singapore; Program of Innovative Cancer Therapeutics, Department of Surgery, First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| | - Can Chen
- Department of Biological Sciences, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Singapore; Program of Innovative Cancer Therapeutics, Department of Surgery, First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| | - Dang Hoang Lam
- Department of Biological Sciences, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Singapore; Program of Innovative Cancer Therapeutics, Department of Surgery, First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| | - Chunxiao Wu
- Department of Biological Sciences, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Singapore; Program of Innovative Cancer Therapeutics, Department of Surgery, First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| | - Jieming Zeng
- Department of Biological Sciences, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Singapore; Program of Innovative Cancer Therapeutics, Department of Surgery, First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| | - Weimin Fan
- Department of Biological Sciences, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Singapore; Program of Innovative Cancer Therapeutics, Department of Surgery, First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| | - Shu Wang
- Department of Biological Sciences, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Singapore; Program of Innovative Cancer Therapeutics, Department of Surgery, First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
16
|
Ferrer-Miralles N, Rodríguez-Carmona E, Corchero JL, García-Fruitós E, Vázquez E, Villaverde A. Engineering protein self-assembling in protein-based nanomedicines for drug delivery and gene therapy. Crit Rev Biotechnol 2013; 35:209-21. [DOI: 10.3109/07388551.2013.833163] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
17
|
Targeting of the tumor necrosis factor receptor superfamily for cancer immunotherapy. ISRN ONCOLOGY 2013; 2013:371854. [PMID: 23840967 PMCID: PMC3693168 DOI: 10.1155/2013/371854] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 05/11/2013] [Indexed: 12/17/2022]
Abstract
The tumor necrosis factor (TNF) ligand and cognate TNF receptor superfamilies constitute an important regulatory axis that is pivotal for immune homeostasis and correct execution of immune responses. TNF ligands and receptors are involved in diverse biological processes ranging from the selective induction of cell death in potentially dangerous and superfluous cells to providing costimulatory signals that help mount an effective immune response. This diverse and important regulatory role in immunity has sparked great interest in the development of TNFL/TNFR-targeted cancer immunotherapeutics. In this review, I will discuss the biology of the most prominent proapoptotic and co-stimulatory TNF ligands and review their current status in cancer immunotherapy.
Collapse
|
18
|
Robak T. Emerging Monoclonal Antibodies and Related Agents for the Treatment of Chronic Lymphocytic Leukemia. Future Oncol 2013; 9:69-91. [DOI: 10.2217/fon.12.157] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Tadeusz Robak
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Ul. Ciolkowskiego 2, Poland and Copernicus Memorial Hospital, 93-510 Lodz, Ul. Ciolkowskiego 2, Poland
| |
Collapse
|