1
|
Li C, Yu X, Han X, Lian C, Wang Z, Shao S, Shao F, Wang H, Ma S, Liu J. Innate immune cells in tumor microenvironment: A new frontier in cancer immunotherapy. iScience 2024; 27:110750. [PMID: 39280627 PMCID: PMC11399700 DOI: 10.1016/j.isci.2024.110750] [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: 09/18/2024] Open
Abstract
Innate immune cells, crucial in resisting infections and initiating adaptive immunity, play diverse and significant roles in tumor development. These cells, including macrophages, granulocytes, dendritic cells (DCs), innate lymphoid cells, and innate-like T cells, are pivotal in the tumor microenvironment (TME). Innate immune cells are crucial components of the TME, based on which various immunotherapy strategies have been explored. Immunotherapy strategies, such as novel immune checkpoint inhibitors, STING/CD40 agonists, macrophage-based surface backpack anchoring, ex vivo polarization approaches, DC-based tumor vaccines, and CAR-engineered innate immune cells, aim to enhance their anti-tumor potential and counteract cancer-induced immunosuppression. The proximity of innate immune cells to tumor cells in the TME also makes them excellent drug carriers. In this review, we will first provide a systematic overview of innate immune cells within the TME and then discuss innate cell-based therapeutic strategies. Furthermore, the research obstacles and perspectives within the field will also be addressed.
Collapse
Affiliation(s)
- Changhui Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyu Yu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyan Han
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Chen Lian
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Zijin Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Fangwei Shao
- National Key Laboratory of Biobased Transportation Fuel Technology, ZJU-UIUC Institute, Zhejiang University, Hangzhou 310027, China
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou 310002, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
2
|
Taghiloo S, Asgarian-Omran H. Cross-talk between leukemic and immune cells at the tumor microenvironment in chronic lymphocytic leukemia: An update review. Eur J Haematol 2024; 113:4-15. [PMID: 38698678 DOI: 10.1111/ejh.14224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is a mature-type B cell malignancy correlated with significant changes and defects in both the innate and adaptive arms of the immune system, together with a high dependency on the tumor microenvironment. Overall, the tumor microenvironment (TME) in CLL provides a supportive niche for leukemic cells to grow and survive, and interactions between CLL cells and the TME can contribute to disease progression and treatment resistance. Therefore, the increasing knowledge of the complicated interaction between immune cells and tumor cells, which is responsible for immune evasion and cancer progression, has provided an opportunity for the development of new therapeutic approaches. In this review, we outline tumor microenvironment-driven contributions to the licensing of immune escape mechanisms in CLL patients.
Collapse
Affiliation(s)
- Saeid Taghiloo
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Asgarian-Omran
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Gastrointestinal Cancer Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
3
|
Silva FS, Barros-Lima A, Souza-Barros M, Crespo-Neto JA, Santos VGR, Pereira DS, Alves-Hanna FS, Magalhães-Gama F, Faria JAQA, Costa AG. A dual-role for IL-10: From leukemogenesis to the tumor progression in acute lymphoblastic leukemia. Cytokine 2023; 171:156371. [PMID: 37725872 DOI: 10.1016/j.cyto.2023.156371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer in the world, and accounts for 25% of all childhood cancers among children under 15 years of age. Longitudinal studies have shown that children with ALL are born with a deregulated immune response that, together with postnatal environmental exposures, favor the onset of the disease. In this context, IL-10, a key cytokine in the regulation of the immune response, presents itself as a paradoxical mediator, initially influencing the development of ALL through the regulation of inflammatory processes and later on the progression of malignancy, with the increase of this molecule in the leukemia microenvironment. According to the literature, this cytokine plays a critical role in the natural history of the disease and plays an important role in two different though complex scenarios. Thus, in this review, we explore the dual role of IL-10 in ALL, and describe its biological characteristics, immunological mechanisms and genetics, as well as its impact on the leukemia microenvironment and its clinical implications.
Collapse
Affiliation(s)
- Flavio Souza Silva
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Amanda Barros-Lima
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Mateus Souza-Barros
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Juniel Assis Crespo-Neto
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | | | - Daniele Sá Pereira
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil; Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| | - Fabíola Silva Alves-Hanna
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Fábio Magalhães-Gama
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou - Fundação Oswaldo Cruz (FIOCRUZ) Minas, Belo Horizonte, Brazil
| | - Jerusa Araújo Quintão Arantes Faria
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Allyson Guimarães Costa
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil; Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil; Escola de Enfermagem de Manaus, UFAM, Manaus, Brazil.
| |
Collapse
|
4
|
Gamal W, Sahakian E, Pinilla-Ibarz J. The role of Th17 cells in chronic lymphocytic leukemia: friend or foe? Blood Adv 2023; 7:2401-2417. [PMID: 36574293 PMCID: PMC10238851 DOI: 10.1182/bloodadvances.2022008985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/18/2022] [Indexed: 12/28/2022] Open
Abstract
T helper 17 (Th17) cells have a prominent role in autoimmune diseases. In contrast, the nature of these cells in cancer is controversial, with either pro- or antitumorigenic activities depending on various cancer settings. Chronic lymphocytic leukemia (CLL), a B-cell malignancy, is characterized by an imbalance in T-cell immune responses that contributes to disease progression and increased mortality. Many clinical reports indicate an increase in Th17 cells and/or interleukin 17 serum cytokine levels in patients with CLL compared with healthy individuals, which correlates with various prognostic markers and significant changes in the tumor microenvironment. The exact mechanisms by which Th17 cells might contribute to CLL progression remain poorly investigated. In this review, we provide an updated presentation of the clinical information related to the significance of Th17 cells in CLL and their interaction with the complex leukemic microenvironment, including various mediators, immune cells, and nonimmune cells. We also address the available data regarding the effects of CLL-targeted therapies on Th17 cells and the potential of using these cells in adoptive cell therapies. Having a sound understanding of the role played by Th17 cells in CLL is crucial for designing novel therapies that can achieve immune homeostasis and maximize clinical benefits.
Collapse
Affiliation(s)
- Wael Gamal
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Eva Sahakian
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Javier Pinilla-Ibarz
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
5
|
Caro AA, Deschoemaeker S, Allonsius L, Coosemans A, Laoui D. Dendritic Cell Vaccines: A Promising Approach in the Fight against Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14164037. [PMID: 36011029 PMCID: PMC9406463 DOI: 10.3390/cancers14164037] [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: 07/27/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022] Open
Abstract
Simple Summary With an overall 5-year survival of only 20% for advanced-stage ovarian cancer patients, enduring and effective therapies are a highly unmet clinical need. Current standard-of-care therapies are able to improve progression-free survival; however, patients still relapse. Moreover, immunotherapy has not resulted in clear patient benefits so far. In this situation, dendritic cell vaccines can serve as a potential therapeutic addition against ovarian cancer. In the current review, we provide an overview of the different dendritic cell subsets and the roles they play in ovarian cancer. We focus on the advancements in dendritic cell vaccination against ovarian cancer and highlight the key outcomes and pitfalls associated with currently used strategies. Finally, we address future directions that could be taken to improve the dendritic cell vaccination outcomes in ovarian cancer. Abstract Ovarian cancer (OC) is the deadliest gynecological malignancy in developed countries and is the seventh-highest cause of death in women diagnosed with cancer worldwide. Currently, several therapies are in use against OC, including debulking surgery, chemotherapy, as well as targeted therapies. Even though the current standard-of-care therapies improve survival, a vast majority of OC patients relapse. Additionally, immunotherapies have only resulted in meager patient outcomes, potentially owing to the intricate immunosuppressive nexus within the tumor microenvironment. In this scenario, dendritic cell (DC) vaccination could serve as a potential addition to the therapeutic options available against OC. In this review, we provide an overview of current therapies in OC, focusing on immunotherapies. Next, we highlight the potential of using DC vaccines in OC by underscoring the different DC subsets and their functions in OC. Finally, we provide an overview of the advances and pitfalls of current DC vaccine strategies in OC while providing future perspectives that could improve patient outcomes.
Collapse
Affiliation(s)
- Aarushi Audhut Caro
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium
| | - Sofie Deschoemaeker
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Lize Allonsius
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium
| | - Damya Laoui
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Correspondence: ; Tel.: +32-2-6291969
| |
Collapse
|
6
|
Kopalli SR, Annamneedi VP, Koppula S. Potential Natural Biomolecules Targeting JAK/STAT/SOCS Signaling in the Management of Atopic Dermatitis. Molecules 2022; 27:molecules27144660. [PMID: 35889539 PMCID: PMC9319717 DOI: 10.3390/molecules27144660] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/29/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease caused by the dysregulation of cytokines and other immune mediators. JAK/STAT is a classical signal transduction pathway involved in various biological processes, and its dysregulation contributes to the key aspects of AD pathogenesis. Suppressor of cytokine signaling (SOCS) proteins negatively regulate the immune-related inflammatory responses mediated by the JAK/STAT pathway. JAK/STAT-mediated production of cytokines including IL-4, IL-13, IL-31, and TSLP inhibits the expression of important skin barrier proteins and triggers pruritus in AD. The expression of SOCS proteins regulates the JAK-mediated cytokines and facilitates maintaining the skin barrier disruptions seen in AD. STATs are crucial in dendritic-cell-activated Th2 cell differentiation in the skin, releasing inflammatory cytokines, indicating that AD is a Th2-mediated skin disorder. SOCS proteins aid in balancing Th1/Th2 cells and, moreover, regulate the onset and maintenance of Th2-mediated allergic responses by reducing the Th2 cell activation and differentiation. SOCS proteins play a pivotal role in inflammatory cytokine-signaling events that act via the JAK/STAT pathway. Therapies relying on natural products and derived biomolecules have proven beneficial in AD when compared with the synthetic regimen. In this review, we focused on the available literature on the potential natural-product-derived biomolecules targeting JAK/STAT/SOCS signaling, mainly emphasizing the SOCS family of proteins (SOCS1, SOCS3, and SOCS5) acting as negative regulators in modulating JAK/STAT-mediated responses in AD pathogenesis and other inflammatory disorders.
Collapse
Affiliation(s)
| | - Venkata Prakash Annamneedi
- Convergence Science Research Center, College of Pharmacy and Institute of Chronic Diseases, Sahmyook University, Seoul 01795, Korea;
| | - Sushruta Koppula
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27381, Korea
- Correspondence:
| |
Collapse
|
7
|
Busà R, Bulati M, Badami E, Zito G, Maresca DC, Conaldi PG, Ercolano G, Ianaro A. Tissue-Resident Innate Immune Cell-Based Therapy: A Cornerstone of Immunotherapy Strategies for Cancer Treatment. Front Cell Dev Biol 2022; 10:907572. [PMID: 35757002 PMCID: PMC9221069 DOI: 10.3389/fcell.2022.907572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/03/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer immunotherapy has led to impressive advances in cancer treatment. Unfortunately, in a high percentage of patients is difficult to consistently restore immune responses to eradicate established tumors. It is well accepted that adaptive immune cells, such as B lymphocytes, CD4+ helper T lymphocytes, and CD8+ cytotoxic T-lymphocytes (CTLs), are the most effective cells able to eliminate tumors. However, it has been recently reported that innate immune cells, including natural killer cells (NK), dendritic cells (DC), macrophages, myeloid-derived suppressor cells (MDSCs), and innate lymphoid cells (ILCs), represent important contributors to modulating the tumor microenvironment and shaping the adaptive tumor response. In fact, their role as a bridge to adaptive immunity, make them an attractive therapeutic target for cancer treatment. Here, we provide a comprehensive overview of the pleiotropic role of tissue-resident innate immune cells in different tumor contexts. In addition, we discuss how current and future therapeutic approaches targeting innate immune cells sustain the adaptive immune system in order to improve the efficacy of current tumor immunotherapies.
Collapse
Affiliation(s)
- Rosalia Busà
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), Palermo, Italy
| | - Matteo Bulati
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), Palermo, Italy
| | - Ester Badami
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), Palermo, Italy
- Ri.MED Foundation, Palermo, Italy
| | - Giovanni Zito
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), Palermo, Italy
| | | | - Pier Giulio Conaldi
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), Palermo, Italy
| | - Giuseppe Ercolano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
- *Correspondence: Giuseppe Ercolano,
| | - Angela Ianaro
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| |
Collapse
|
8
|
Pinho MP, Lepski GA, Rehder R, Chauca-Torres NE, Evangelista GCM, Teixeira SF, Flatow EA, de Oliveira JV, Fogolin CS, Peres N, Arévalo A, Alves V, Barbuto JAM, Bergami-Santos PC. Near-Complete Remission of Glioblastoma in a Patient Treated with an Allogenic Dendritic Cell-Based Vaccine: The Role of Tumor-Specific CD4+T-Cell Cytokine Secretion Pattern in Predicting Response and Recurrence. Int J Mol Sci 2022; 23:5396. [PMID: 35628206 PMCID: PMC9141410 DOI: 10.3390/ijms23105396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/19/2022] [Accepted: 05/03/2022] [Indexed: 01/27/2023] Open
Abstract
Immunotherapy has brought hope to the fight against glioblastoma, but its efficacy remains unclear. We present the case of CST, a 25-year-old female patient with a large right-hemisphere glioblastoma treated with a dendritic-tumor cell fusion vaccine. CST showed a near-complete tumor response, with a marked improvement in her functional status and simultaneous increases in tumor-specific CD8+ and CD4+ T cells. Two months before recurrence, the frequency of tumor-specific T cells decreased, while that of IL-17 and CD4+ T cells increased. CST passed away 15 months after enrollment. In this illustrative case, the tumor-specific CD4+ T-cell numbers and phenotype behaved as treatment efficacy biomarkers, highlighting the key role of the latter in glioblastoma immunotherapy.
Collapse
Affiliation(s)
- Mariana P. Pinho
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Guilherme A. Lepski
- Hospital das Clínicas HCFMUSP, LIM26, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil; (G.A.L.); (A.A.)
- Department of Neurosurgery, Eberhard-Karls University, 72074 Tuebingen, Germany
| | | | - Nadia E. Chauca-Torres
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Gabriela C. M. Evangelista
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Sarah F. Teixeira
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Elizabeth A. Flatow
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Jaqueline V. de Oliveira
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Carla S. Fogolin
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| | - Nataly Peres
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
- Laboratory of Experimental Surgery (LIM-26), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Analía Arévalo
- Hospital das Clínicas HCFMUSP, LIM26, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil; (G.A.L.); (A.A.)
| | - Venâncio Alves
- Department of Pathology, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246-903, Brazil;
| | - José A. M. Barbuto
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Patricia C. Bergami-Santos
- Department of Immunology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (M.P.P.); (N.E.C.-T.); (G.C.M.E.); (S.F.T.); (E.A.F.); (J.V.d.O.); (C.S.F.); (N.P.)
| |
Collapse
|
9
|
Zhou Y, Li K, Zou X, Hua Z, Wang H, Bian W, Wang H, Chen F, Dai T. LncRNA DHRS4-AS1 ameliorates hepatocellular carcinoma by suppressing proliferation and promoting apoptosis via miR-522-3p/SOCS5 axis. Bioengineered 2021; 12:10862-10877. [PMID: 34666613 PMCID: PMC8809963 DOI: 10.1080/21655979.2021.1994719] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Recent years have seen much effect in revealing the pathological association between lncRNA and HCC. Herein, we identified lncRNA DHRS4-AS1 as a potential tumor suppressor in HCC. Firstly, it was discovered that DHRS4-AS1 was significantly down-regulated in HCC tissues compared to normal tissues based on the database TCGA. It was also detected in a lower-than-usual expression quantity in HCC tissues we collected and HCC cell lines. Kaplan-Meier survival analysis revealed that high expression of DHRS4-AS1 contributed to higher overall survival rate of HCC patients.DHRS4-AS1 expression was significantly correlated to tumor size (P = 0.02) and TNM stage (P = 0.045). CCK-8, BrdU and colony-forming assays collectively demonstrated that overexpression of DHRS4-AS1 significantly restrained HCC cell proliferation. In vivo xenograft animal experiment showed that DHRS4-AS1 could efficiently preclude the tumor growth of HCC. Further investigation performed using flow cytometry and western blot showed that DHRS4-AS1 exerted its effects by accelerating cell apoptosis and capturing cell cycle in G0/G1 phase. Our study subsequently lucubrated that miR-522-3p was a negative target of DHRS4-AS1. Increased expression level of miR-522-3p was examined in HCC tissues and cell lines. Similarly, miR-522-3p mimics could reverse the inhibitory effect on HCC brought by DHRS4-AS1. SOCS5 was then discovered as a down-stream target of miR-522-3p, which suggested that SOCS5 participated in DHRS4-AS1/miR-522-3p axis to collectively mediate the development of HCC. Our study provides lncRNA DHRS4-AS1/miR-522-3p/SOCS5 axis as a novel target for HCC therapeutic strategy with potentiality.
Collapse
Affiliation(s)
- Yongping Zhou
- Department of Hepatobiliary, Wuxi No.2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu, China
| | - Kuan Li
- Department of Hepatobiliary Surgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Xuexia Zou
- Department of Operation Room, Wuxi No.2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu, China
| | - Zhiyuan Hua
- Department of Hepatobiliary, Wuxi No.2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu, China
| | - Hao Wang
- Department of Hepatobiliary, Wuxi No.2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu, China
| | - Wuyang Bian
- Department of Hepatobiliary, Wuxi No.2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu, China
| | - Hong Wang
- Department of Hepatobiliary, Wuxi No.2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu, China
| | - Fangming Chen
- Department of Imagine, Wuxi No.2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu, China
| | - Tu Dai
- Department of Hepatobiliary, Wuxi No.2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu, China
| |
Collapse
|
10
|
Subtil B, Cambi A, Tauriello DVF, de Vries IJM. The Therapeutic Potential of Tackling Tumor-Induced Dendritic Cell Dysfunction in Colorectal Cancer. Front Immunol 2021; 12:724883. [PMID: 34691029 PMCID: PMC8527179 DOI: 10.3389/fimmu.2021.724883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is the third most diagnosed malignancy and the second leading cause of cancer-related deaths worldwide. Locally advanced and metastatic disease exhibit resistance to therapy and are prone to recurrence. Despite significant advances in standard of care and targeted (immuno)therapies, the treatment effects in metastatic CRC patients have been modest. Untreatable cancer metastasis accounts for poor prognosis and most CRC deaths. The generation of a strong immunosuppressive tumor microenvironment (TME) by CRC constitutes a major hurdle for tumor clearance by the immune system. Dendritic cells (DCs), often impaired in the TME, play a critical role in the initiation and amplification of anti-tumor immune responses. Evidence suggests that tumor-mediated DC dysfunction is decisive for tumor growth and metastasis initiation, as well as for the success of immunotherapies. Unravelling and understanding the complex crosstalk between CRC and DCs holds promise for identifying key mechanisms involved in tumor progression and spread that can be exploited for therapy. The main goal of this review is to provide an overview of the current knowledge on the impact of CRC-driven immunosuppression on DCs phenotype and functionality, and its significance for disease progression, patient prognosis, and treatment response. Moreover, present knowledge gaps will be highlighted as promising opportunities to further understand and therapeutically target DC dysfunction in CRC. Given the complexity and heterogeneity of CRC, future research will benefit from the use of patient-derived material and the development of in vitro organoid-based co-culture systems to model and study DCs within the CRC TME.
Collapse
Affiliation(s)
- Beatriz Subtil
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alessandra Cambi
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Daniele V. F. Tauriello
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
11
|
RNF2 ablation reprograms the tumor-immune microenvironment and stimulates durable NK and CD4 + T-cell-dependent antitumor immunity. NATURE CANCER 2021; 2:1018-1038. [PMID: 35121884 PMCID: PMC8809507 DOI: 10.1038/s43018-021-00263-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/25/2021] [Indexed: 02/08/2023]
Abstract
Expanding the utility of immune-based cancer treatments is a clinical challenge due to tumor-intrinsic factors that suppress the immune response. Here we report the identification of tumoral ring finger protein 2 (RNF2), the core subunit of polycomb repressor complex 1, as a negative regulator of antitumor immunity in various human cancers, including breast cancer. In syngeneic murine models of triple-negative breast cancer, we found that deleting genes encoding the polycomb repressor complex 1 subunits Rnf2, BMI1 proto-oncogene, polycomb ring finger (Bmi1), or the downstream effector of Rnf2, remodeling and spacing factor 1 (Rsf1), was sufficient by itself to induce durable tumor rejection and establish immune memory by enhancing infiltration and activation of natural killer and CD4+ T cells, but not CD8+ T cells, into the tumor and enabled their cooperativity. These findings uncover an epigenetic reprogramming of the tumor-immune microenvironment, which fosters durable antitumor immunity and memory.
Collapse
|
12
|
Keewan E, Matlawska-Wasowska K. The Emerging Role of Suppressors of Cytokine Signaling (SOCS) in the Development and Progression of Leukemia. Cancers (Basel) 2021; 13:4000. [PMID: 34439155 PMCID: PMC8393695 DOI: 10.3390/cancers13164000] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
Cytokines are pleiotropic signaling molecules that execute an essential role in cell-to-cell communication through binding to cell surface receptors. Receptor binding activates intracellular signaling cascades in the target cell that bring about a wide range of cellular responses, including induction of cell proliferation, migration, differentiation, and apoptosis. The Janus kinase and transducers and activators of transcription (JAK/STAT) signaling pathways are activated upon cytokines and growth factors binding with their corresponding receptors. The SOCS family of proteins has emerged as a key regulator of cytokine signaling, and SOCS insufficiency leads to constitutive activation of JAK/STAT signaling and oncogenic transformation. Dysregulation of SOCS expression is linked to various solid tumors with invasive properties. However, the roles of SOCS in hematological malignancies, such as leukemia, are less clear. In this review, we discuss the recent advances pertaining to SOCS dysregulation in leukemia development and progression. We also highlight the roles of specific SOCS in immune cells within the tumor microenvironment and their possible involvement in anti-tumor immunity. Finally, we discuss the epigenetic, genetic, and post-transcriptional modifications of SOCS genes during tumorigenesis, with an emphasis on leukemia.
Collapse
Affiliation(s)
- Esra’a Keewan
- Department of Pediatrics, Division of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA;
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA
| | - Ksenia Matlawska-Wasowska
- Department of Pediatrics, Division of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA;
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
13
|
Salah A, Wang H, Li Y, Ji M, Ou WB, Qi N, Wu Y. Insights Into Dendritic Cells in Cancer Immunotherapy: From Bench to Clinical Applications. Front Cell Dev Biol 2021; 9:686544. [PMID: 34262904 PMCID: PMC8273339 DOI: 10.3389/fcell.2021.686544] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/11/2021] [Indexed: 01/05/2023] Open
Abstract
Dendritic cells (DCs) are efficient antigen-presenting cells (APCs) and potent activators of naïve T cells. Therefore, they act as a connective ring between innate and adaptive immunity. DC subsets are heterogeneous in their ontogeny and functions. They have proven to potentially take up and process tumor-associated antigens (TAAs). In this regard, researchers have developed strategies such as genetically engineered or TAA-pulsed DC vaccines; these manipulated DCs have shown significant outcomes in clinical and preclinical models. Here, we review DC classification and address how DCs are skewed into an immunosuppressive phenotype in cancer patients. Additionally, we present the advancements in DCs as a platform for cancer immunotherapy, emphasizing the technologies used for in vivo targeting of endogenous DCs, ex vivo generated vaccines from peripheral blood monocytes, and induced pluripotent stem cell-derived DCs (iPSC-DCs) to boost antitumoral immunity.
Collapse
Affiliation(s)
- Ahmed Salah
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Hao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.,Hangzhou Biaomo Biosciences Co., Ltd., Hangzhou, China.,Asia Stem Cell Therapies Co., Limited, Shanghai, China
| | - Yanqin Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Meng Ji
- Hangzhou Biaomo Biosciences Co., Ltd., Hangzhou, China
| | - Wen-Bin Ou
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nianmin Qi
- Hangzhou Biaomo Biosciences Co., Ltd., Hangzhou, China.,Asia Stem Cell Therapies Co., Limited, Shanghai, China
| | - Yuehong Wu
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
14
|
Berzaghi R, Tornaas S, Lode K, Hellevik T, Martinez-Zubiaurre I. Ionizing Radiation Curtails Immunosuppressive Effects From Cancer-Associated Fibroblasts on Dendritic Cells. Front Immunol 2021; 12:662594. [PMID: 34177901 PMCID: PMC8221608 DOI: 10.3389/fimmu.2021.662594] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/24/2021] [Indexed: 01/07/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) participate actively in tumor development and affect treatment responses, by among other mechanisms, promoting an immunosuppressive tumor microenvironment. In contrast to normal fibroblasts, reactive CAFs secrete a myriad of immunomodulatory soluble factors at high levels, i.e. growth factors, cytokines, and chemokines, which directly influence tumor immunity and inflammation. CAFs have been identified as important players in tumor radioresistance. However, knowledge on the immunomodulatory functions of CAFs during/after radiotherapy is still lacking. In this study, we investigated the effects of ionizing radiation on CAF-mediated regulation of dendritic cells (DCs). CAFs were obtained from freshly operated lung cancer tissues, while DCs were procured from peripheral blood of healthy donors. Experimental settings comprised both co-cultures and incubations with conditioned medium from control and irradiated CAFs. Functional assays to study DC differentiation/activation consisted on cytokine release, expression of cell-surface markers, antigen uptake, migration rates, T cell priming, and DC-signaling analysis. We demonstrate that CAFs induce a tolerogenic phenotype in DCs by promoting down-regulation of: i) signature DC markers (CD14, CD1a, CD209); ii) activation markers (CD80, CD86, CD40, and HLA-DR) and iii) functional properties (migration, antigen uptake, and CD4+ T cell priming). Notably, some of these effects were lost in conditioned medium from CAFs irradiated at fractionated medium-dose regimens (3x6 Gy). However, the expression of relevant CAF-derived regulatory agents like thymic stromal lymphopoietin (TSLP) or tryptophan 2,3-dioxygenase (TDO2) was unchanged upon irradiation. This study demonstrates that CAFs interfere with DC immune functions and unveil that certain radiation regimens may reverse CAF-mediated immunosuppressive effects.
Collapse
Affiliation(s)
- Rodrigo Berzaghi
- Department of Clinical Medicine, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Stian Tornaas
- Department of Clinical Medicine, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Kristin Lode
- Department of Clinical Medicine, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Turid Hellevik
- Department of Radiation Oncology, University Hospital of Northern Norway, Tromsø, Norway
| | - Inigo Martinez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
15
|
Tu Z, Schmoellerl J, Mariani O, Zheng Y, Hu Y, Vincent-Salomon A, Karnoub AE. The LINC01119-SOCS5 axis as a critical theranostic in triple-negative breast cancer. NPJ Breast Cancer 2021; 7:69. [PMID: 34059683 PMCID: PMC8166834 DOI: 10.1038/s41523-021-00259-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 04/09/2021] [Indexed: 02/04/2023] Open
Abstract
The development of triple-negative breast cancer (TNBC) is critically regulated by certain tumor-microenvironment-associated cells called mesenchymal stem/stromal cells (MSCs), which we and others have shown promote TNBC progression by activating pro-malignant signaling in neighboring cancer cells. Characterization of these cascades would better our understanding of TNBC biology and bring about therapeutics that eliminate the morbidity and mortality associated with advanced disease. Here, we focused on the emerging class of RNAs called long non-coding RNAs or lncRNAs and utilized a MSC-supported TNBC progression model to identify specific family members of functional relevance to TNBC pathogenesis. Indeed, although some have been described to play functional roles in TNBC, activities of lncRNAs as mediators of tumor-microenvironment-driven TNBC development remain to be fully explored. We report that MSCs stimulate robust expression of LINC01119 in TNBC cells, which in turn induces suppressor of cytokine signaling 5 (SOCS5), leading to accelerated cancer cell growth and tumorigenesis. We show that LINC01119 and SOCS5 exhibit tight correlation across multiple breast cancer gene sets and that they are highly enriched in TNBC patient cohorts. Importantly, we present evidence that the LINC01119-SOCS5 axis represents a powerful prognostic indicator of adverse outcomes in TNBC patients, and demonstrate that its repression severely impairs cancer cell growth. Altogether, our findings identify LINC01119 as a major driver of TNBC development and delineate critical non-coding RNA theranostics of potential translational utility in the management of advanced TNBC, a class of tumors in most need of effective and targeted therapy.
Collapse
Affiliation(s)
- Zhenbo Tu
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Johannes Schmoellerl
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Yurong Zheng
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yi Hu
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Antoine E Karnoub
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
16
|
Targeting CD38 is lethal to Breg-like chronic lymphocytic leukemia cells and Tregs, but restores CD8+ T-cell responses. Blood Adv 2021; 4:2143-2157. [PMID: 32421811 DOI: 10.1182/bloodadvances.2019001091] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 04/09/2020] [Indexed: 01/03/2023] Open
Abstract
Patients with chronic lymphocytic leukemia (CLL) are characterized by monoclonal expansion of CD5+CD23+CD27+CD19+κ/λ+ B lymphocytes and are clinically noted to have profound immune suppression. In these patients, it has been recently shown that a subset of B cells possesses regulatory functions and secretes high levels of interleukin 10 (IL-10). Our investigation identified that CLL cells with a CD19+CD24+CD38hi immunophenotype (B regulatory cell [Breg]-like CLL cells) produce high amounts of IL-10 and transforming growth factor β (TGF-β) and are capable of transforming naive T helper cells into CD4+CD25+FoxP3+ T regulatory cells (Tregs) in an IL-10/TGF-β-dependent manner. A strong correlation between the percentage of CD38+ CLL cells and Tregs was observed. CD38hi Tregs comprised more than 50% of Tregs in peripheral blood mononuclear cells (PBMCs) in patients with CLL. Anti-CD38 targeting agents resulted in lethality of both Breg-like CLL and Treg cells via apoptosis. Ex vivo, use of anti-CD38 monoclonal antibody (mAb) therapy was associated with a reduction in IL-10 and CLL patient-derived Tregs, but an increase in interferon-γ and proliferation of cytotoxic CD8+ T cells with an activated phenotype, which showed an improved ability to lyse patient-autologous CLL cells. Finally, effects of anti-CD38 mAb therapy were validated in a CLL-patient-derived xenograft model in vivo, which showed decreased percentage of Bregs, Tregs, and PD1+CD38hiCD8+ T cells, but increased Th17 and CD8+ T cells (vs vehicle). Altogether, our results demonstrate that targeting CD38 in CLL can modulate the tumor microenvironment; skewing T-cell populations from an immunosuppressive to immune-reactive milieu, thus promoting immune reconstitution for enhanced anti-CLL response.
Collapse
|
17
|
Liu HM, Guo CL, Zhang YF, Chen JF, Liang ZP, Yang LH, Ma YP. Leonurine-Repressed miR-18a-5p/SOCS5/JAK2/STAT3 Axis Activity Disrupts CML malignancy. Front Pharmacol 2021; 12:657724. [PMID: 33935775 PMCID: PMC8087248 DOI: 10.3389/fphar.2021.657724] [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: 01/23/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022] Open
Abstract
Leonurine, an active natural alkaloid compound isolated from Herba leonuri, has been reported to exhibit promising anticancer activity in solid tumors. The aim of this study was to explore whether leonurine is able to inhibit chronic myeloid leukemia (CML) malignancy. Here, we found that leonurine dose dependently inhibited the proliferation, migration, colony formation and promoted apoptosis of CML cells. Furthermore, leonurine markedly reduced CML xenograft growth in vivo. Mechanically, leonurine upregulated SOCS5 expression, thus leading JAK2/STAT3 signaling suppression. Silencing of SOCS5 by its siRNA abrogated the effect of leonurine on CML cells, demonstrating that SOCS5 mediates the anti-leukemia effect of leonurine. Notably, we observed that miR-18a-5p was remarkably increased in CML cells. Treating CML cells with leonurine significantly decreased miR-18a-5p expression. Moreover, we found miR-18a-5p repressed SOCS5 by directly targeting its 3′-UTR. miR-18a-5p downregulation induced by leonurine reduced the biological activity of CML cells by relieving miR-18a-5p repression of SOCS5 expression. Taken together, leonurine exerts significant anti-leukemia efficacy in CML by regulating miR-18a-5p/SOCS5/JAK2/STAT3 axis.
Collapse
Affiliation(s)
- Hui-Min Liu
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chun-Ling Guo
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yao-Fang Zhang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jian-Fang Chen
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhi-Peng Liang
- Basic Laboratory of Internal Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lin-Hua Yang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan-Ping Ma
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
18
|
Mysona DP, Tran L, Bai S, dos Santos B, Ghamande S, Chan J, She JX. Tumor-intrinsic and -extrinsic (immune) gene signatures robustly predict overall survival and treatment response in high grade serous ovarian cancer patients. Am J Cancer Res 2021; 11:181-199. [PMID: 33520368 PMCID: PMC7840710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 09/14/2020] [Indexed: 06/12/2023] Open
Abstract
In the present study, we developed a transcriptomic signature capable of predicting prognosis and response to primary therapy in high grade serous ovarian cancer (HGSOC). Proportional hazard analysis was performed on individual genes in the TCGA RNAseq data set containing 229 HGSOC patients. Ridge regression analysis was performed to select genes and develop multigenic models. Survival analysis identified 120 genes whose expression levels were associated with overall survival (OS) (HR = 1.49-2.46 or HR = 0.48-0.63). Ridge regression modeling selected 38 of the 120 genes for development of the final Ridge regression models. The consensus model based on plurality voting by 68 individual Ridge regression models classified 102 (45%) as low, 23 (10%) as moderate and 104 patients (45%) as high risk. The median OS was 31 months (HR = 7.63, 95% CI = 4.85-12.0, P < 1.0-10) and 77 months (HR = ref) in the high and low risk groups, respectively. The gene signature had two components: intrinsic (proliferation, metastasis, autophagy) and extrinsic (immune evasion). Moderate/high risk patients had more partial and non-responses to primary therapy than low risk patients (odds ratio = 4.54, P < 0.001). We concluded that the overall survival and response to primary therapy in ovarian cancer is best assessed using a combination of gene signatures. A combination of genes which combines both tumor intrinsic and extrinsic functions has the best prediction. Validation studies are warranted in the future.
Collapse
Affiliation(s)
- David P Mysona
- University of North CarolinaChapel Hill, NC 27517, USA
- Jinfiniti Precision Medicine, Inc.Augusta, GA 30907, USA
| | - Lynn Tran
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
| | - Shan Bai
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
| | | | - Sharad Ghamande
- Department of OBGYN, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
| | - John Chan
- Palo Alto Medical Foundation Research InstitutePalo Alto, CA 94301, USA
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
- Department of OBGYN, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
| |
Collapse
|
19
|
Griggio V, Perutelli F, Salvetti C, Boccellato E, Boccadoro M, Vitale C, Coscia M. Immune Dysfunctions and Immune-Based Therapeutic Interventions in Chronic Lymphocytic Leukemia. Front Immunol 2020; 11:594556. [PMID: 33312177 PMCID: PMC7708380 DOI: 10.3389/fimmu.2020.594556] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/14/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy characterized by a wide range of tumor-induced alterations, which affect both the innate and adaptive arms of the immune response, and accumulate during disease progression. In recent years, the development of targeted therapies, such as the B-cell receptor signaling inhibitors and the Bcl-2 protein inhibitor venetoclax, has dramatically changed the treatment landscape of CLL. Despite their remarkable anti-tumor activity, targeted agents have some limitations, which include the development of drug resistance mechanisms and the inferior efficacy observed in high-risk patients. Therefore, additional treatments are necessary to obtain deeper responses and overcome drug resistance. Allogeneic hematopoietic stem cell transplantation (HSCT), which exploits immune-mediated graft-versus-leukemia effect to eradicate tumor cells, currently represents the only potentially curative therapeutic option for CLL patients. However, due to its potential toxicities, HSCT can be offered only to a restricted number of younger and fit patients. The growing understanding of the complex interplay between tumor cells and the immune system, which is responsible for immune escape mechanisms and tumor progression, has paved the way for the development of novel immune-based strategies. Despite promising preclinical observations, results from pilot clinical studies exploring the safety and efficacy of novel immune-based therapies have been sometimes suboptimal in terms of long-term tumor control. Therefore, further advances to improve their efficacy are needed. In this context, possible approaches include an earlier timing of immunotherapy within the treatment sequencing, as well as the possibility to improve the efficacy of immunotherapeutic agents by administering them in combination with other anti-tumor drugs. In this review, we will provide a comprehensive overview of main immune defects affecting patients with CLL, also describing the complex networks leading to immune evasion and tumor progression. From the therapeutic standpoint, we will go through the evolution of immune-based therapeutic approaches over time, including i) agents with broad immunomodulatory effects, such as immunomodulatory drugs, ii) currently approved and next-generation monoclonal antibodies, and iii) immunotherapeutic strategies aiming at activating or administering immune effector cells specifically targeting leukemic cells (e.g. bi-or tri-specific antibodies, tumor vaccines, chimeric antigen receptor T cells, and checkpoint inhibitors).
Collapse
Affiliation(s)
- Valentina Griggio
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Francesca Perutelli
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Chiara Salvetti
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elia Boccellato
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Mario Boccadoro
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Candida Vitale
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Marta Coscia
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
20
|
Del Prete A, Sozio F, Barbazza I, Salvi V, Tiberio L, Laffranchi M, Gismondi A, Bosisio D, Schioppa T, Sozzani S. Functional Role of Dendritic Cell Subsets in Cancer Progression and Clinical Implications. Int J Mol Sci 2020; 21:ijms21113930. [PMID: 32486257 PMCID: PMC7312661 DOI: 10.3390/ijms21113930] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DCs) constitute a complex network of cell subsets with common functions but also with many divergent aspects. All dendritic cell subsets share the ability to prime T cell response and to undergo a complex trafficking program related to their stage of maturation and function. For these reasons, dendritic cells are implicated in a large variety of both protective and detrimental immune responses, including a crucial role in promoting anti-tumor responses. Although cDC1s are the most potent subset in tumor antigen cross-presentation, they are not sufficient to induce full-strength anti-tumor cytotoxic T cell response and need close interaction and cooperativity with the other dendritic cell subsets, namely cDC2s and pDCs. This review will take into consideration different aspects of DC biology, including the functional role of dendritic cell subsets in both fostering and suppressing tumor growth, the mechanisms underlying their recruitment into the tumor microenvironment, as well as the prognostic value and the potentiality of dendritic cell therapeutic targeting. Understanding the specificity of dendritic cell subsets will allow to gain insights on role of these cells in pathological conditions and to design new selective promising therapeutic approaches.
Collapse
Affiliation(s)
- Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Francesca Sozio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Ilaria Barbazza
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Laura Tiberio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Mattia Laffranchi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Angela Gismondi
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy;
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Tiziana Schioppa
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Silvano Sozzani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy;
- Correspondence: ; Tel.: +39-06-4434-0632
| |
Collapse
|
21
|
Immune Response Dysfunction in Chronic Lymphocytic Leukemia: Dissecting Molecular Mechanisms and Microenvironmental Conditions. Int J Mol Sci 2020; 21:ijms21051825. [PMID: 32155826 PMCID: PMC7084946 DOI: 10.3390/ijms21051825] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 01/05/2023] Open
Abstract
Representing the major cause of morbidity and mortality for chronic lymphocytic leukemia (CLL) patients, immunosuppression is a common feature of the disease. Effectors of the innate and the adaptive immune response show marked dysfunction and skewing towards the generation of a tolerant environment that favors disease expansion. Major deregulations are found in the T lymphocyte compartment, with inhibition of CD8+ cytotoxic and CD4+ activated effector T cells, replaced by exhausted and more tolerogenic subsets. Likewise, differentiation of monocytes towards a suppressive M2-like phenotype is induced at the expense of pro-inflammatory sub-populations. Thanks to their B-regulatory phenotype, leukemic cells play a central role in driving immunosuppression, progressively inhibiting immune responses. A number of signaling cascades triggered by soluble mediators and cell–cell contacts contribute to immunomodulation in CLL, fostered also by local environmental conditions, such as hypoxia and derived metabolic acidosis. Specifically, molecular pathways modulating T-cell activity in CLL, spanning from the best known cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed cell death 1 (PD-1) to the emerging T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibition motif domains (TIGIT)/CD155 axes, are attracting increasing research interest and therapeutic relevance also in the CLL field. On the other hand, in the microenvironment, the B cell receptor (BCR), which is undoubtedly the master regulator of leukemic cell behavior, plays an important role in orchestrating immune responses, as well. Lastly, local conditions of hypoxia, typical of the lymphoid niche, have major effects both on CLL cells and on non-leukemic immune cells, partly mediated through adenosine signaling, for which novel specific inhibitors are currently under development. In summary, this review will provide an overview of the molecular and microenvironmental mechanisms that modify innate and adaptive immune responses of CLL patients, focusing attention on those that may have therapeutic implications.
Collapse
|
22
|
O'Brien LJ, Guillerey C, Radford KJ. Can Dendritic Cell Vaccination Prevent Leukemia Relapse? Cancers (Basel) 2019; 11:cancers11060875. [PMID: 31234526 PMCID: PMC6627518 DOI: 10.3390/cancers11060875] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/14/2019] [Accepted: 06/20/2019] [Indexed: 01/02/2023] Open
Abstract
Leukemias are clonal proliferative disorders arising from immature leukocytes in the bone marrow. While the advent of targeted therapies has improved survival in certain subtypes, relapse after initial therapy is a major problem. Dendritic cell (DC) vaccination has the potential to induce tumor-specific T cells providing long-lasting, anti-tumor immunity. This approach has demonstrated safety but limited clinical success until recently, as DC vaccination faces several barriers in both solid and hematological malignancies. Importantly, vaccine-mediated stimulation of protective immune responses is hindered by the aberrant production of immunosuppressive factors by cancer cells which impede both DC and T cell function. Leukemias present the additional challenge of severely disrupted hematopoiesis owing to both cytogenic defects in hematopoietic progenitors and an abnormal hematopoietic stem cell niche in the bone marrow; these factors accentuate systemic immunosuppression and DC malfunction. Despite these obstacles, several recent clinical trials have caused great excitement by extending survival in Acute Myeloid Leukemia (AML) patients through DC vaccination. Here, we review the phenotype and functional capacity of DCs in leukemia and approaches to harness DCs in leukemia patients. We describe the recent clinical successes in AML and detail the multiple new strategies that might enhance prognosis in AML and other leukemias.
Collapse
Affiliation(s)
- Liam J O'Brien
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| | - Camille Guillerey
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| | - Kristen J Radford
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
23
|
Sharma ND, Nickl CK, Kang H, Ornatowski W, Brown R, Ness SA, Loh ML, Mullighan CG, Winter SS, Hunger SP, Cannon JL, Matlawska‐Wasowska K. Epigenetic silencing of SOCS5 potentiates JAK-STAT signaling and progression of T-cell acute lymphoblastic leukemia. Cancer Sci 2019; 110:1931-1946. [PMID: 30974024 PMCID: PMC6549933 DOI: 10.1111/cas.14021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 04/03/2019] [Accepted: 04/08/2019] [Indexed: 01/12/2023] Open
Abstract
Activating mutations in cytokine receptors and transcriptional regulators govern aberrant signal transduction in T-cell lineage acute lymphoblastic leukemia (T-ALL). However, the roles played by suppressors of cytokine signaling remain incompletely understood. We examined the regulatory roles of suppressor of cytokine signaling 5 (SOCS5) in T-ALL cellular signaling networks and leukemia progression. We found that SOCS5 was differentially expressed in primary T-ALL and its expression levels were lowered in HOXA-deregulated leukemia harboring KMT2A gene rearrangements. Here, we report that SOCS5 expression is epigenetically regulated by DNA methyltransferase-3A-mediated DNA methylation and methyl CpG binding protein-2-mediated histone deacetylation. We show that SOCS5 negatively regulates T-ALL cell growth and cell cycle progression but has no effect on apoptotic cell death. Mechanistically, SOCS5 silencing induces activation of JAK-STAT signaling, and negatively regulates interleukin-7 and interleukin-4 receptors. Using a human T-ALL murine xenograft model, we show that genetic inactivation of SOCS5 accelerates leukemia engraftment and progression, and leukemia burden. We postulate that SOCS5 is epigenetically deregulated in T-ALL and serves as an important regulator of T-ALL cell proliferation and leukemic progression. Our results link aberrant downregulation of SOCS5 expression to the enhanced activation of the JAK-STAT and cytokine receptor-signaling cascade in T-ALL.
Collapse
Affiliation(s)
- Nitesh D. Sharma
- Department of PediatricsUniversity of New Mexico Health Sciences CenterAlbuquerqueNM
| | - Christian K. Nickl
- Department of PediatricsUniversity of New Mexico Health Sciences CenterAlbuquerqueNM
| | - Huining Kang
- Department of Internal MedicineUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Wojciech Ornatowski
- Department of PathologyUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Roger Brown
- Department of Internal MedicineUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Scott A. Ness
- Department of Internal MedicineUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Mignon L. Loh
- Department of PediatricsBenioff Children's HospitalUniversity of California at San FranciscoSan FranciscoCA
| | | | - Stuart S. Winter
- Children's Minnesota Research Institute and Cancer and Blood Disorders ProgramChildren's MinnesotaMinneapolisMN
| | - Stephen P. Hunger
- Department of Pediatrics and the Center for Childhood Cancer ResearchChildren's Hospital of PhiladelphiaPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPA
| | - Judy L. Cannon
- Department of PathologyUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | | |
Collapse
|
24
|
Hadadi L, Hafezi M, Amirzargar AA, Sharifian RA, Abediankenari S, Asgarian-Omran H. Dysregulated Expression of Tim-3 and NKp30 Receptors on NK Cells of Patients with Chronic Lymphocytic Leukemia. Oncol Res Treat 2019; 42:202-208. [PMID: 30870839 DOI: 10.1159/000497208] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 01/23/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND In this study, the expression pattern of NKp30 and T cell immunoglobulin and mucin domain-containing molecule-3 (Tim-3), as candidates for activating and inhibitory receptors of NK cells, were evaluated in patients with chronic lymphocytic leukemia (CLL). PATIENTS AND METHODS 24 CLL patients and 19 healthy controls were enrolled. Fresh peripheral blood was collected from all subjects and stained with fluorochrome-conjugated antibodies. The frequency of CD56+/CD3-/NKp30+ and CD56+/CD3-/Tim-3+ cells was determined by multicolor flow cytometry. RESULTS Our results revealed that Tim-3 is significantly upregulated on natural killer (NK) cells of CLL patients in comparison to healthy controls. NK cells of CLL patients showed lower expression of NKp30-activating receptor compared to controls. Tim-3 expression pattern on NK cells of CLL patients was correlated with poor prognostic factors including low hemoglobin level, high absolute lymphocyte count, and high serum C-reactive protein level. CONCLUSION Dysregulated expression of Tim-3 and NKp30 receptors confirms the exhaustion state of NK cells in CLL. Our data introduce Tim-3 as a promising biomarker and potential target for immunotherapy of CLL.
Collapse
Affiliation(s)
- Leila Hadadi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Morteza Hafezi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Amirzargar
- Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramazan Ali Sharifian
- Clinic of Hematology and Oncology, Vali-Asr Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeid Abediankenari
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Asgarian-Omran
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran, .,Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran,
| |
Collapse
|
25
|
Patente TA, Pinho MP, Oliveira AA, Evangelista GCM, Bergami-Santos PC, Barbuto JAM. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front Immunol 2019; 9:3176. [PMID: 30719026 PMCID: PMC6348254 DOI: 10.3389/fimmu.2018.03176] [Citation(s) in RCA: 239] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/24/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DC) are professional antigen presenting cells, uniquely able to induce naïve T cell activation and effector differentiation. They are, likewise, involved in the induction and maintenance of immune tolerance in homeostatic conditions. Their phenotypic and functional heterogeneity points to their great plasticity and ability to modulate, according to their microenvironment, the acquired immune response and, at the same time, makes their precise classification complex and frequently subject to reviews and improvement. This review will present general aspects of the DC physiology and classification and will address their potential and actual uses in the management of human disease, more specifically cancer, as therapeutic and monitoring tools. New combination treatments with the participation of DC will be also discussed.
Collapse
Affiliation(s)
- Thiago A Patente
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariana P Pinho
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aline A Oliveira
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gabriela C M Evangelista
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patrícia C Bergami-Santos
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - José A M Barbuto
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Discipline of Molecular Medicine, Department of Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Bruns H, Jitschin S, Gamali S, Saul D, Böttcher M, Mackensen A, Jitschin R, Mougiakakos D. A novel immunoregulatory function of beta-2-microglobulin as a promoter of myeloid derived suppressor cell induction. Leukemia 2019; 33:1282-1287. [PMID: 30635624 DOI: 10.1038/s41375-018-0345-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/28/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Heiko Bruns
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Simon Jitschin
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Sara Gamali
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Domenica Saul
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Martin Böttcher
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Regina Jitschin
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany.
| |
Collapse
|
27
|
Haseeb M, Anwar MA, Choi S. Molecular Interactions Between Innate and Adaptive Immune Cells in Chronic Lymphocytic Leukemia and Their Therapeutic Implications. Front Immunol 2018; 9:2720. [PMID: 30542344 PMCID: PMC6277854 DOI: 10.3389/fimmu.2018.02720] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/05/2018] [Indexed: 12/15/2022] Open
Abstract
Innate immunity constitutes the first line of host defense against various anomalies in humans, and it also guides the adaptive immune response. The function of innate immune components and adaptive immune components are interlinked in hematological malignancies including chronic lymphocytic leukemia (CLL), and molecular interactions between innate and adaptive immune components are crucial for the development, progression and the therapeutic outcome of CLL. In this leukemia, genetic mutations in B cells and B cell receptors (BCR) are key driving factors along with evasion of cytotoxic T lymphocytes and promotion of regulatory T cells. Similarly, the release of various cytokines from CLL cells triggers the protumor phenotype in macrophages that further edges the CLL cells. Moreover, under the influence of various cytokines, dendritic cells are unable to mature and trigger T cell mediated antitumor response. The phenotypes of these cells are ultimately controlled by respective signaling pathways, the most notables are BCR, Wnt, Notch, and NF-κB, and their activation affects the cytokine profile that controls the pathogenesis of CLL, and challenge its treatment. There are several novel substances for CLL under clinical development, including kinase inhibitors, antibodies, and immune-modulators that offer new hopes. DC-based vaccines and CAR T cell therapy are promising tools; however, further studies are required to precisely dissect the molecular interactions among various molecular entities. In this review, we systematically discuss the involvement, common targets and therapeutic interventions of various cells for the better understanding and therapy of CLL.
Collapse
Affiliation(s)
- Muhammad Haseeb
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Muhammad Ayaz Anwar
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
28
|
Sanchez-Mejias A, Kwon J, Chew XH, Siemens A, Sohn HS, Jing G, Zhang B, Yang H, Tay Y. A novel SOCS5/miR-18/miR-25 axis promotes tumorigenesis in liver cancer. Int J Cancer 2018; 144:311-321. [DOI: 10.1002/ijc.31857] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 07/14/2018] [Accepted: 08/15/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Avencia Sanchez-Mejias
- Cancer Science Institute of Singapore, Centre for Translational Medicine; National University of Singapore; Singapore 117599 Singapore
| | - Junsu Kwon
- Cancer Science Institute of Singapore, Centre for Translational Medicine; National University of Singapore; Singapore 117599 Singapore
| | - Xiao Hong Chew
- Cancer Science Institute of Singapore, Centre for Translational Medicine; National University of Singapore; Singapore 117599 Singapore
| | - Angela Siemens
- Cancer Science Institute of Singapore, Centre for Translational Medicine; National University of Singapore; Singapore 117599 Singapore
| | - Hye Seon Sohn
- Cancer Science Institute of Singapore, Centre for Translational Medicine; National University of Singapore; Singapore 117599 Singapore
| | - Guo Jing
- Cancer Science Institute of Singapore, Centre for Translational Medicine; National University of Singapore; Singapore 117599 Singapore
| | - Bin Zhang
- Cancer Science Institute of Singapore, Centre for Translational Medicine; National University of Singapore; Singapore 117599 Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, Centre for Translational Medicine; National University of Singapore; Singapore 117599 Singapore
| | - Yvonne Tay
- Cancer Science Institute of Singapore, Centre for Translational Medicine; National University of Singapore; Singapore 117599 Singapore
- Department of Biochemistry; Yong Loo Lin School of Medicine, National University of Singapore; Singapore 117597 Singapore
| |
Collapse
|
29
|
van Attekum MH, Eldering E, Kater AP. Chronic lymphocytic leukemia cells are active participants in microenvironmental cross-talk. Haematologica 2017; 102:1469-1476. [PMID: 28775118 PMCID: PMC5685246 DOI: 10.3324/haematol.2016.142679] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
The importance of the tumor microenvironment in chronic lymphocytic leukemia is
widely accepted. Nevertheless, the understanding of the complex interplay
between the various types of bystander cells and chronic lymphocytic leukemia
cells is incomplete. Numerous studies have indicated that bystander cells
provide chronic lymphocytic leukemia-supportive functions, but it has also
become clear that chronic lymphocytic leukemia cells actively engage in the
formation of a supportive tumor microenvironment through several cross-talk
mechanisms. In this review, we describe how chronic lymphocytic leukemia cells
participate in this interplay by inducing migration and tumor-supportive
differentiation of bystander cells. Furthermore, chronic lymphocytic
leukemia-mediated alterations in the interactions between bystander cells are
discussed. Upon bystander cell interaction, chronic lymphocytic leukemia cells
secrete cytokines and chemokines such as migratory factors [chemokine
(C-C motif) ligand 22 and chemokine (CC motif) ligand 2], which result
in further recruitment of T cells but also of monocyte-derived cells. Within the
tumor microenvironment, chronic lymphocytic leukemia cells induce
differentiation towards a tumor-supportive M2 phenotype of monocyte-derived
cells and suppress phagocytosis, but also induce increased numbers of supportive
regulatory T cells. Like other tumor types, the differentiation of stromal cells
towards supportive cancer-associated fibroblasts is critically dependent on
chronic lymphocytic leukemia-derived factors such as exosomes and
platelet-derived growth factor. Lastly, both chronic lymphocytic leukemia and
bystander cells induce a tolerogenic tumor microenvironment; chronic lymphocytic
leukemia-secreted cytokines, such as interleukin-10, suppress cytotoxic T-cell
functions, while chronic lymphocytic leukemia-associated monocyte-derived cells
contribute to suppression of T-cell function by producing the immune checkpoint
factor, programmed cell death-ligand 1. Deeper understanding of the active
involvement and cross-talk of chronic lymphocytic leukemia cells in shaping the
tumor microenvironment may offer novel clues for designing therapeutic
strategies.
Collapse
Affiliation(s)
- Martijn Ha van Attekum
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, the Netherlands.,Department of Hematology, Academic Medical Center, University of Amsterdam, the Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, the Netherlands.,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Academic Medical Center, University of Amsterdam, the Netherlands
| | - Arnon P Kater
- Department of Hematology, Academic Medical Center, University of Amsterdam, the Netherlands .,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Academic Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|