1
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
2
|
Malika N, Zhansaya A, Kasym M, Kanat T, Yerlan R, Kanatbek M. Analysis of Antibody Induction by Macrophages Treated Ex Vivo with Human Proteins in Mice. Rep Biochem Mol Biol 2023; 11:694-701. [PMID: 37131900 PMCID: PMC10149125 DOI: 10.52547/rbmb.11.4.694] [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: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 05/04/2023]
Abstract
Background Macrophages are essential cellular components in various body tissues and tumor microenvironments. The high infiltration of macrophages into the tumor microenvironment determines the importance of ex vivo treatment of personalized macrophages with recombinant cytotoxic T-lymphocyte-associated protein 4 (rCTLA-4), programmed death-ligand 1 (rPD-L1), and programmed cell death protein 1 (rPD-1) proteins to block immune checkpoints. Methods We investigated the development of humoral immunity against CTLA-4, PD-L1, and PD-1 receptors by introducing macrophages treated ex vivo with the corresponding proteins into mice. Peritoneal macrophages from BALB/c mice were cultured in medium containing recombinant human CTLA-4, PD-L1, and PD-1 proteins. Macrophages processing recombinant proteins were analyzed via immunofluorescence staining using antibodies against CTLA-4, PD-L1, and PD-1. The treated macrophages were administered intraperitoneally to mice to induce anti-CTLA-4, anti-PD-L1, and anti-PD-1 antibodies. The antibody titer in vaccinated mice was determined via enzyme-linked immunosorbent assays, followed by statistical analysis of the results. The specificity of the antibodies was determined via immunofluorescence staining in MCF7 cells. Results The ex vivo treatment of macrophages with rCTLA-4, rPD-L1, and rPD-1 induced the formation of specific antibodies in vaccinated mice. The various rPD-L1 and rPD-1 concentrations used to treat macrophages had no significant effect on the specific antibody titers, while the anti-rCTLA-4 titer was dependent on the protein concentration in the culture medium. Immunofluorescence analysis revealed that anti-CTLA-4 and PD-L1 antibodies reacted with MCF7 cells. Conclusion The ex vivo treatment of macrophages with rCTLA-4, rPD-L1, and rPD-1 can help induce humoral immunity and develop new approaches for cancer immunotherapy.
Collapse
Affiliation(s)
- Nurtleu Malika
- Republican State Enterprise National Center for Biotechnology, Ministry of Healthcare of the Republic of Kazakhstan, Nur-Sultan, 010000, Kazakhstan
- L.N. Gumilyov Eurasian National University, Satpayev st., 2, Nur-Sultan, 010008, Kazakhstan.
| | - Adish Zhansaya
- Republican State Enterprise National Center for Biotechnology, Ministry of Healthcare of the Republic of Kazakhstan, Nur-Sultan, 010000, Kazakhstan
- L.N. Gumilyov Eurasian National University, Satpayev st., 2, Nur-Sultan, 010008, Kazakhstan.
| | - Mukanov Kasym
- Republican State Enterprise National Center for Biotechnology, Ministry of Healthcare of the Republic of Kazakhstan, Nur-Sultan, 010000, Kazakhstan
| | - Tursunov Kanat
- Republican State Enterprise National Center for Biotechnology, Ministry of Healthcare of the Republic of Kazakhstan, Nur-Sultan, 010000, Kazakhstan
- Corresponding author: Tursunov Kanat; Tel: +77 172-707527; E-mail:
| | - Ramankulov Yerlan
- Republican State Enterprise National Center for Biotechnology, Ministry of Healthcare of the Republic of Kazakhstan, Nur-Sultan, 010000, Kazakhstan
| | - Mukantayev Kanatbek
- Republican State Enterprise National Center for Biotechnology, Ministry of Healthcare of the Republic of Kazakhstan, Nur-Sultan, 010000, Kazakhstan
| |
Collapse
|
3
|
Yew CHT, Gurumoorthy N, Nordin F, Tye GJ, Wan Kamarul Zaman WS, Tan JJ, Ng MH. Integrase deficient lentiviral vector: prospects for safe clinical applications. PeerJ 2022; 10:e13704. [PMID: 35979475 PMCID: PMC9377332 DOI: 10.7717/peerj.13704] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/19/2022] [Indexed: 01/17/2023] Open
Abstract
HIV-1 derived lentiviral vector is an efficient transporter for delivering desired genetic materials into the targeted cells among many viral vectors. Genetic material transduced by lentiviral vector is integrated into the cell genome to introduce new functions, repair defective cell metabolism, and stimulate certain cell functions. Various measures have been administered in different generations of lentiviral vector systems to reduce the vector's replicating capabilities. Despite numerous demonstrations of an excellent safety profile of integrative lentiviral vectors, the precautionary approach has prompted the development of integrase-deficient versions of these vectors. The generation of integrase-deficient lentiviral vectors by abrogating integrase activity in lentiviral vector systems reduces the rate of transgenes integration into host genomes. With this feature, the integrase-deficient lentiviral vector is advantageous for therapeutic implementation and widens its clinical applications. This short review delineates the biology of HIV-1-erived lentiviral vector, generation of integrase-deficient lentiviral vector, recent studies involving integrase-deficient lentiviral vectors, limitations, and prospects for neoteric clinical use.
Collapse
Affiliation(s)
- Chee-Hong Takahiro Yew
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Universiti Kebangsaan Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - Narmatha Gurumoorthy
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Universiti Kebangsaan Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Universiti Kebangsaan Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | | | - Jun Jie Tan
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM), Bertam, Kepala Batas, Pulau Pinang, Malaysia
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Universiti Kebangsaan Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Bialek-Waldmann JK, Domning S, Esser R, Glienke W, Mertens M, Aleksandrova K, Arseniev L, Kumar S, Schneider A, Koenig J, Theobald SJ, Tsay HC, Cornelius ADA, Bonifacius A, Eiz-Vesper B, Figueiredo C, Schaudien D, Talbot SR, Bleich A, Spineli LM, von Kaisenberg C, Clark C, Blasczyk R, Heuser M, Ganser A, Köhl U, Farzaneh F, Stripecke R. Induced dendritic cells co-expressing GM-CSF/IFN-α/tWT1 priming T and B cells and automated manufacturing to boost GvL. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:621-641. [PMID: 34095345 PMCID: PMC8142053 DOI: 10.1016/j.omtm.2021.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/03/2021] [Indexed: 12/13/2022]
Abstract
Acute myeloid leukemia (AML) patients with minimal residual disease and receiving allogeneic hematopoietic stem cell transplantation (HCT) have poor survival. Adoptive administration of dendritic cells (DCs) presenting the Wilms tumor protein 1 (WT1) leukemia-associated antigen can potentially stimulate de novo T and B cell development to harness the graft-versus-leukemia (GvL) effect after HCT. We established a simple and fast genetic modification of monocytes for simultaneous lentiviral expression of a truncated WT1 antigen (tWT1), granulocyte macrophage-colony-stimulating factor (GM-CSF), and interferon (IFN)-α, promoting their self-differentiation into potent “induced DCs” (iDCtWT1). A tricistronic integrase-defective lentiviral vector produced under good manufacturing practice (GMP)-like conditions was validated. Transduction of CD14+ monocytes isolated from peripheral blood, cord blood, and leukapheresis material effectively induced their self-differentiation. CD34+ cell-transplanted Nod.Rag.Gamma (NRG)- and Nod.Scid.Gamma (NSG) mice expressing human leukocyte antigen (HLA)-A∗0201 (NSG-A2)-immunodeficient mice were immunized with autologous iDCtWT1. Both humanized mouse models showed improved development and maturation of human T and B cells in the absence of adverse effects. Toward clinical use, manufacturing of iDCtWT1 was up scaled and streamlined using the automated CliniMACS Prodigy system. Proof-of-concept clinical-scale runs were feasible, and the 38-h process enabled standardized production and high recovery of a cryopreserved cell product with the expected identity characteristics. These results advocate for clinical trials testing iDCtWT1 to boost GvL and eradicate leukemia.
Collapse
Affiliation(s)
- Julia K Bialek-Waldmann
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Sabine Domning
- Molecular Medicine Group, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, Kings College London, London, UK
| | - Ruth Esser
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany
| | - Wolfgang Glienke
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany
| | - Mira Mertens
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany
| | | | - Lubomir Arseniev
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany
| | - Suresh Kumar
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Schneider
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Johannes Koenig
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, 30625 Hannover, Germany
| | - Sebastian J Theobald
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, 30625 Hannover, Germany
| | - Hsin-Chieh Tsay
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Angela D A Cornelius
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Constanca Figueiredo
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | - Steven R Talbot
- Institute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany
| | - Loukia M Spineli
- Department of Obstetrics, Gynecology and Reproductive Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Constantin von Kaisenberg
- Department of Obstetrics, Gynecology and Reproductive Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Caren Clark
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
| | - Ulrike Köhl
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany.,Fraunhofer Institute for Cell Therapy and Immunology IZI and University of Leipzig, 04103 Leipzig, Germany
| | - Farzin Farzaneh
- Molecular Medicine Group, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, Kings College London, London, UK
| | - Renata Stripecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, 30625 Hannover, Germany
| |
Collapse
|
5
|
Jung S, Appleton E, Ali M, Church GM, Del Sol A. A computer-guided design tool to increase the efficiency of cellular conversions. Nat Commun 2021; 12:1659. [PMID: 33712564 PMCID: PMC7954801 DOI: 10.1038/s41467-021-21801-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Human cell conversion technology has become an important tool for devising new cell transplantation therapies, generating disease models and testing gene therapies. However, while transcription factor over-expression-based methods have shown great promise in generating cell types in vitro, they often endure low conversion efficiency. In this context, great effort has been devoted to increasing the efficiency of current protocols and the development of computational approaches can be of great help in this endeavor. Here we introduce a computer-guided design tool that combines a computational framework for prioritizing more efficient combinations of instructive factors (IFs) of cellular conversions, called IRENE, with a transposon-based genomic integration system for efficient delivery. Particularly, IRENE relies on a stochastic gene regulatory network model that systematically prioritizes more efficient IFs by maximizing the agreement of the transcriptional and epigenetic landscapes between the converted and target cells. Our predictions substantially increased the efficiency of two established iPSC-differentiation protocols (natural killer cells and melanocytes) and established the first protocol for iPSC-derived mammary epithelial cells with high efficiency.
Collapse
Affiliation(s)
- Sascha Jung
- Computational Biology Group, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Bizkaia Technology Park, Derio, Spain
| | - Evan Appleton
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Muhammad Ali
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Maastricht University School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- GC Therapeutics, Inc, Cambridge, MA, USA
| | - Antonio Del Sol
- Computational Biology Group, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Bizkaia Technology Park, Derio, Spain.
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.
| |
Collapse
|
6
|
Fenton SE, Saleiro D, Platanias LC. Type I and II Interferons in the Anti-Tumor Immune Response. Cancers (Basel) 2021; 13:1037. [PMID: 33801234 PMCID: PMC7957896 DOI: 10.3390/cancers13051037] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
The interferons (IFNs) are essential components of the immune response against infections and malignancies. IFNs are potent promoters of the anti-tumor response, but there is also evidence that feedback mechanisms regulated by IFNs negatively control immune responses to avoid hyper-activation and limit inflammation. This balance of responses plays an important role in cancer surveillance, immunoediting and response to anticancer therapeutic approaches. Here we review the roles of both type I and type II IFNs on the control of the immune response against malignancies in the context of effects on both malignant cells and cells of the immune system in the tumor microenvironment.
Collapse
Affiliation(s)
- Sarah E. Fenton
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA; (S.E.F.); (D.S.)
- Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Diana Saleiro
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA; (S.E.F.); (D.S.)
- Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA; (S.E.F.); (D.S.)
- Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
7
|
Eiz-Vesper B, Schmetzer HM. Antigen-Presenting Cells: Potential of Proven und New Players in Immune Therapies. Transfus Med Hemother 2020; 47:429-431. [PMID: 33442337 PMCID: PMC7768096 DOI: 10.1159/000512729] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Helga Maria Schmetzer
- Med III, Department for Hematopoietic Transplantations, University Hospital of Munich − Grosshadern, Munich, Germany
| |
Collapse
|
8
|
Cunningham S, Hackstein H. Recent Advances in Good Manufacturing Practice-Grade Generation of Dendritic Cells. Transfus Med Hemother 2020; 47:454-463. [PMID: 33442340 DOI: 10.1159/000512451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/11/2020] [Indexed: 12/23/2022] Open
Abstract
Dendritic cells (DCs) are pivotal regulators of immune responses, specialized in antigen presentation and bridging the gap between the innate and adaptive immune system. Due to these key features, DCs have become a pillar of the continuously growing field of cellular therapies. Here we review recent advances in good manufacturing practice strategies and their individual specificities in relation to DC production for clinical applications. These take into account both small-scale experimental approaches as well as automated systems for patient care.
Collapse
Affiliation(s)
- Sarah Cunningham
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
9
|
Lapenta C, Gabriele L, Santini SM. IFN-Alpha-Mediated Differentiation of Dendritic Cells for Cancer Immunotherapy: Advances and Perspectives. Vaccines (Basel) 2020; 8:vaccines8040617. [PMID: 33086492 PMCID: PMC7711454 DOI: 10.3390/vaccines8040617] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 01/08/2023] Open
Abstract
The past decade has seen tremendous developments in novel cancer therapies through targeting immune-checkpoint molecules. However, since increasing the presentation of tumor antigens remains one of the major issues for eliciting a strong antitumor immune response, dendritic cells (DC) still hold a great potential for the development of cancer immunotherapy. A considerable body of evidence clearly demonstrates the importance of the interactions of type I IFN with the immune system for the generation of a durable antitumor response through its effects on DC. Actually, highly active DC can be rapidly generated from blood monocytes in vitro in the presence of IFN-α (IFN-DC), suitable for therapeutic vaccination of cancer patients. Here we review how type I IFN can promote the ex vivo differentiation of human DC and orientate DC functions towards the priming and expansion of protective antitumor immune responses. New epigenetic elements of control on activation of the type I IFN signal will be highlighted. We also review a few clinical trials exploiting IFN-DC in cancer vaccination and discuss how IFN-DC could be exploited for the design of effective strategies of cancer immunotherapy as a monotherapy or in combination with immune-checkpoint inhibitors or immunomodulatory drugs.
Collapse
|
10
|
Cox MC, Lapenta C, Santini SM. Advances and perspectives of dendritic cell-based active immunotherapies in follicular lymphoma. Cancer Immunol Immunother 2020; 69:913-925. [PMID: 32322910 DOI: 10.1007/s00262-020-02577-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/11/2020] [Indexed: 12/13/2022]
Abstract
Follicular lymphoma (FL) is a remarkably immune-responsive malignancy, which is still considered incurable. As, standard immunochemotherapy is complex, toxic and not curative, improvement in FL care is now a crucial topic in hemato-oncology. Recently, we and others have shown that dendritic cell (DC)-based therapies allow a specific immune response associated with sustained lymphoma regression in a proportion of low-tumor burden FL patients. Importantly, the rate of objective clinical response (33-50%) and of sustained remission is remarkably higher compared to similar studies in solid tumors, corroborating the assumption of the immune responsiveness of FL. Our experimental intra-tumoral strategy combined injection with rituximab and interferon-α-derived dendritic cells (IFN-DC), a novel DC population particularly efficient in biasing T-helper response toward the Th1 type and in the cross-priming of CD8 + T cells. Noteworthy, intra-tumoral injection of DC is a new therapeutic option based on the assumption that following the induction of cancer-cell immunogenic death, unloaded DC would phagocytize in vivo the tumor associated antigens and give rise to a specific immune response. This approach allows the design of easy and inexpensive schedules. On the other hand, advanced and straightforward methods to produce clinical-grade antigenic formulations are currently under development. Both unloaded DC strategies and DC-vaccines are suited for combination with radiotherapy, immune checkpoint inhibitors, immunomodulators and metronomic chemotherapy. In fact, studies in animal models have already shown impressive results, while early-phase combination trials are ongoing. Here, we summarize the recent advances and the future perspectives of DC-based therapies in the treatment of FL patients.
Collapse
Affiliation(s)
- Maria Christina Cox
- Department of Haematology, King's College Hospital NHS Foundation Trust and Sant'Andrea University Hospital, Rome, Italy
| | - Caterina Lapenta
- Dipartimento Di Oncologia e Medicina Molecolare, Istituto Superiore Di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Stefano M Santini
- Dipartimento Di Oncologia e Medicina Molecolare, Istituto Superiore Di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|