1
|
Zaninelli S, Panna S, Tettamanti S, Melita G, Doni A, D’Autilia F, Valgardsdottir R, Gotti E, Rambaldi A, Golay J, Introna M. Functional Activity of Cytokine-Induced Killer Cells Enhanced by CAR-CD19 Modification or by Soluble Bispecific Antibody Blinatumomab. Antibodies (Basel) 2024; 13:71. [PMID: 39311376 PMCID: PMC11417890 DOI: 10.3390/antib13030071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 09/26/2024] Open
Abstract
Strategies to increase the anti-tumor efficacy of cytokine-induced killer cells (CIKs) include genetic modification with chimeric antigen receptors (CARs) or the addition of soluble T-cell engaging bispecific antibodies (BsAbs). Here, CIKs were modified using a transposon system integrating two distinct anti-CD19 CARs (CAR-MNZ and CAR-BG2) or combined with soluble CD3xCD19 BsAb blinatumomab (CIK + Blina). CAR-MNZ bearing the CD28-OX40-CD3ζ signaling modules, and CAR-BG2, designed on the Tisagenlecleucel CAR sequence (Kymriah®), carrying the 4-1BB and CD3ζ signaling elements, were employed. After transfection and CIK expansion, cells expressed CAR-CD19 to a similar extent (35.9% CAR-MNZ and 17.7% CAR-BG2). In vitro evaluations demonstrated robust proliferation and cytotoxicity (~50% cytotoxicity) of CARCIK-MNZ, CARCIK-BG2, and CIK + Blina against CD19+ target cells, suggesting similar efficacy. All effectors formed an increased number of synapses, activated NFAT and NFkB, and secreted IL-2 and IFN-ɣ upon encountering targets. CIK + Blina displayed strongest NFAT and IFN-ɣ induction, whereas CARCIK-BG2 demonstrated superior synapse formation. All the effectors have shown therapeutic activity in vivo against the CD19+ Daudi tumor model, with CARCIK cells showing a more durable response compared to CIK + Blina, likely due to the short half-life of Blina in this model.
Collapse
Affiliation(s)
- Silvia Zaninelli
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Silvia Panna
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Sarah Tettamanti
- M. Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Giusi Melita
- M. Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Andrea Doni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, 20089 Milano, Italy
| | - Francesca D’Autilia
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, 20089 Milano, Italy
| | - Rut Valgardsdottir
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Elisa Gotti
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII Hospital, 24127 Bergamo, Italy
- Department of Oncology and Hematology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Josée Golay
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Martino Introna
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| |
Collapse
|
2
|
Ma Y, Lv H, Xing F, Xiang W, Wu Z, Feng Q, Wang H, Yang W. Cancer stem cell-immune cell crosstalk in the tumor microenvironment for liver cancer progression. Front Med 2024; 18:430-445. [PMID: 38600350 DOI: 10.1007/s11684-023-1049-z] [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: 06/09/2023] [Accepted: 11/15/2023] [Indexed: 04/12/2024]
Abstract
Crosstalk between cancer cells and the immune microenvironment is determinant for liver cancer progression. A tumor subpopulation called liver cancer stem cells (CSCs) significantly accounts for the initiation, metastasis, therapeutic resistance, and recurrence of liver cancer. Emerging evidence demonstrates that the interaction between liver CSCs and immune cells plays a crucial role in shaping an immunosuppressive microenvironment and determining immunotherapy responses. This review sheds light on the bidirectional crosstalk between liver CSCs and immune cells for liver cancer progression, as well as the underlying molecular mechanisms after presenting an overview of liver CSCs characteristic and their microenvironment. Finally, we discuss the potential application of liver CSCs-targeted immunotherapy for liver cancer treatment.
Collapse
Affiliation(s)
- Yue Ma
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Hongwei Lv
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
| | - Fuxue Xing
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Wei Xiang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Zixin Wu
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Qiyu Feng
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Hongyang Wang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - Wen Yang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| |
Collapse
|
3
|
Zhao H, Ling Y, He J, Dong J, Mo Q, Wang Y, Zhang Y, Yu H, Tang C. Potential targets and therapeutics for cancer stem cell-based therapy against drug resistance in hepatocellular carcinoma. Drug Resist Updat 2024; 74:101084. [PMID: 38640592 DOI: 10.1016/j.drup.2024.101084] [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: 02/26/2024] [Revised: 03/22/2024] [Accepted: 04/06/2024] [Indexed: 04/21/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common digestive malignancyin the world, which is frequently diagnosed at late stage with a poor prognosis. For most patients with advanced HCC, the therapeutic options arelimiteddue to cancer occurrence of drug resistance. Hepatic cancer stem cells (CSCs) account for a small subset of tumor cells with the ability of self-renewal and differentiationin HCC. It is widely recognized that the presence of CSCs contributes to primary and acquired drug resistance. Therefore, hepatic CSCs-targeted therapy is considered as a promising strategy to overcome drug resistance and improve therapeutic outcome in HCC. In this article, we review drug resistance in HCC and provide a summary of potential targets for CSCs-based therapy. In addition, the development of CSCs-targeted therapeuticsagainst drug resistance in HCC is summarized in both preclinical and clinical trials. The in-depth understanding of CSCs-related drug resistance in HCC will favor optimization of the current therapeutic strategies and gain encouraging therapeutic outcomes.
Collapse
Affiliation(s)
- Hongxing Zhao
- Department of Radiology, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China
| | - Yuhang Ling
- Central Laboratory, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China; Huzhou Key Laboratory of Translational Medicine, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China
| | - Jie He
- Department of Hepatology, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China
| | - Jinling Dong
- Department of Hepatology, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China
| | - Qinliang Mo
- Department of Hepatopancreatobiliary Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China
| | - Yao Wang
- Department of Hepatopancreatobiliary Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China
| | - Ying Zhang
- Central Laboratory, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China; Department of Hepatology, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China
| | - Hongbin Yu
- Department of General Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China
| | - Chengwu Tang
- Huzhou Key Laboratory of Translational Medicine, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China; Department of Hepatopancreatobiliary Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province, China.
| |
Collapse
|
4
|
Chen YC, Gowda K, Amin S, Schell TD, Sharma AK, Robertson GP. Pharmacological agents targeting drug-tolerant persister cells in cancer. Pharmacol Res 2024; 203:107163. [PMID: 38569982 DOI: 10.1016/j.phrs.2024.107163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/05/2024] [Accepted: 03/29/2024] [Indexed: 04/05/2024]
Abstract
Current cancer therapy can be effective, but the development of drug resistant disease is the usual outcome. These drugs can eliminate most of the tumor burden but often fail to eliminate the rare, "Drug Tolerant Persister" (DTP) cell subpopulations in residual tumors, which can be referred to as "Persister" cells. Therefore, novel therapeutic agents specifically targeting or preventing the development of drug-resistant tumors mediated by the remaining persister cells subpopulations are needed. Since approximately ninety percent of cancer-related deaths occur because of the eventual development of drug resistance, identifying, and dissecting the biology of the persister cells is essential for the creation of drugs to target them. While there remains uncertainty surrounding all the markers identifying DTP cells in the literature, this review summarizes the drugs and therapeutic approaches that are available to target the persister cell subpopulations expressing the cellular markers ATP-binding cassette sub-family B member 5 (ABCB5), CD133, CD271, Lysine-specific histone demethylase 5 (KDM5), and aldehyde dehydrogenase (ALDH). Persister cells expressing these markers were selected as the focus of this review because they have been found on cells surviving following drug treatments that promote recurrent drug resistant cancer and are associated with stem cell-like properties, including self-renewal, differentiation, and resistance to therapy. The limitations and obstacles facing the development of agents targeting these DTP cell subpopulations are detailed, with discussion of potential solutions and current research areas needing further exploration.
Collapse
Affiliation(s)
- Yu-Chi Chen
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Krishne Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Shantu Amin
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Todd D Schell
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Arun K Sharma
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA, USA; The Pennsylvania State University Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
5
|
MacLean MR, Walker OL, Arun RP, Fernando W, Marcato P. Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways. Int J Mol Sci 2024; 25:4102. [PMID: 38612911 PMCID: PMC11012648 DOI: 10.3390/ijms25074102] [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: 02/28/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation within tumors that promote cancer progression, metastasis, and recurrence due to their self-renewal capacity and resistance to conventional therapies. CSC-specific markers and signaling pathways highly active in CSCs have emerged as a promising strategy for improving patient outcomes. This review provides a comprehensive overview of the therapeutic targets associated with CSCs of solid tumors across various cancer types, including key molecular markers aldehyde dehydrogenases, CD44, epithelial cellular adhesion molecule, and CD133 and signaling pathways such as Wnt/β-catenin, Notch, and Sonic Hedgehog. We discuss a wide array of therapeutic modalities ranging from targeted antibodies, small molecule inhibitors, and near-infrared photoimmunotherapy to advanced genetic approaches like RNA interference, CRISPR/Cas9 technology, aptamers, antisense oligonucleotides, chimeric antigen receptor (CAR) T cells, CAR natural killer cells, bispecific T cell engagers, immunotoxins, drug-antibody conjugates, therapeutic peptides, and dendritic cell vaccines. This review spans developments from preclinical investigations to ongoing clinical trials, highlighting the innovative targeting strategies that have been informed by CSC-associated pathways and molecules to overcome therapeutic resistance. We aim to provide insights into the potential of these therapies to revolutionize cancer treatment, underscoring the critical need for a multi-faceted approach in the battle against cancer. This comprehensive analysis demonstrates how advances made in the CSC field have informed significant developments in novel targeted therapeutic approaches, with the ultimate goal of achieving more effective and durable responses in cancer patients.
Collapse
Affiliation(s)
- Maya R. MacLean
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Olivia L. Walker
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Raj Pranap Arun
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Wasundara Fernando
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Biology, Acadia University, Wolfville, NS B4P 2R6, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Nova Scotia Health Authority, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
6
|
Yi SY, Wei MZ, Zhao L. Targeted immunotherapy to cancer stem cells: A novel strategy of anticancer immunotherapy. Crit Rev Oncol Hematol 2024; 196:104313. [PMID: 38428702 DOI: 10.1016/j.critrevonc.2024.104313] [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: 10/14/2023] [Revised: 02/04/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
Cancer is a major disease that endangers human health. Cancer drug resistance and relapse are the two main causes contributing to cancer treatment failure. Cancer stem cells (CSCs) are a small fraction of tumor cells that are responsible for tumorigenesis, metastasis, relapse, and resistance to conventional anticancer therapies. Therefore, CSCs are considered to be the root of cancer recurrence, metastasis, and drug resistance. Novel anticancer strategies need to face this new challenge and explore their efficacy against CSCs. Recently, immunotherapy has made rapid advances in cancer treatment, and its potential against CSCs is also an interesting area of research. Meanwhile, immunotherapy strategies are novel therapeutic modalities with promising results in targeting CSCs. In this review, we summarize the targeting of CSCs by various immunotherapy strategies such as monoclonal antibodies(mAb), tumor vaccines, immune checkpoint inhibitors, and chimeric antigen receptor-T cells(CAR-T) in pre-clinical and clinical studies. This review provides new insights into the application of these immunotherapeutic approaches to potential anti-tumor therapies in the future.
Collapse
Affiliation(s)
- Shan-Yong Yi
- Department of Oncology of the Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zheng Zhou, Henan Province 450007, China.
| | - Mei-Zhuo Wei
- Department of Oncology of the Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zheng Zhou, Henan Province 450007, China
| | - Ling Zhao
- Department of Oncology of the Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zheng Zhou, Henan Province 450007, China.
| |
Collapse
|
7
|
Guo X, Wu Y, Xue Y, Xie N, Shen G. Revolutionizing cancer immunotherapy: unleashing the potential of bispecific antibodies for targeted treatment. Front Immunol 2023; 14:1291836. [PMID: 38106416 PMCID: PMC10722299 DOI: 10.3389/fimmu.2023.1291836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/08/2023] [Indexed: 12/19/2023] Open
Abstract
Recent progressions in immunotherapy have transformed cancer treatment, providing a promising strategy that activates the immune system of the patient to find and eliminate cancerous cells. Bispecific antibodies, which engage two separate antigens or one antigen with two distinct epitopes, are of tremendous concern in immunotherapy. The bi-targeting idea enabled by bispecific antibodies (BsAbs) is especially attractive from a medical standpoint since most diseases are complex, involving several receptors, ligands, and signaling pathways. Several research look into the processes in which BsAbs identify different cancer targets such angiogenesis, reproduction, metastasis, and immune regulation. By rerouting cells or altering other pathways, the bispecific proteins perform effector activities in addition to those of natural antibodies. This opens up a wide range of clinical applications and helps patients with resistant tumors respond better to medication. Yet, further study is necessary to identify the best conditions where to use these medications for treating tumor, their appropriate combination partners, and methods to reduce toxicity. In this review, we provide insights into the BsAb format classification based on their composition and symmetry, as well as the delivery mode, focus on the action mechanism of the molecule, and discuss the challenges and future perspectives in BsAb development.
Collapse
Affiliation(s)
- Xiaohan Guo
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yi Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ying Xue
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Guobo Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
8
|
Koukourakis IM, Platoni K, Kouloulias V, Arelaki S, Zygogianni A. Prostate Cancer Stem Cells: Biology and Treatment Implications. Int J Mol Sci 2023; 24:14890. [PMID: 37834336 PMCID: PMC10573523 DOI: 10.3390/ijms241914890] [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: 09/06/2023] [Revised: 09/30/2023] [Accepted: 10/01/2023] [Indexed: 10/15/2023] Open
Abstract
Stem cells differentiate into mature organ/tissue-specific cells at a steady pace under normal conditions, but their growth can be accelerated during the process of tissue healing or in the context of certain diseases. It is postulated that the proliferation and growth of carcinomas are sustained by the presence of a vital cellular compartment resembling stem cells residing in normal tissues: 'stem-like cancer cells' or cancer stem cells (CSCs). Mutations in prostate stem cells can lead to the formation of prostate cancer. Prostate CSCs (PCSCs) have been identified and partially characterized. These express surface markers include CD44, CD133, integrin α2β1, and pluripotency factors like OCT4, NANOG, and SOX2. Several signaling pathways are also over-activated, including Notch, PTEN/Akt/PI3K, RAS-RAF-MEK-ERK and HH. Moreover, PCSCs appear to induce resistance to radiotherapy and chemotherapy, while their presence has been linked to aggressive cancer behavior and higher relapse rates. The development of treatment policies to target PCSCs in tumors is appealing as radiotherapy and chemotherapy, through cancer cell killing, trigger tumor repopulation via activated stem cells. Thus, blocking this reactive stem cell mobilization may facilitate a positive outcome through cytotoxic treatment.
Collapse
Affiliation(s)
- Ioannis M. Koukourakis
- Radiation Oncology Unit, 1st Department of Radiology, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens (NKUOA), 11528 Athens, Greece; (I.M.K.); (A.Z.)
| | - Kalliopi Platoni
- Medical Physics Unit, 2nd Department of Radiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens (NKUOA), 12462 Athens, Greece
| | - Vassilis Kouloulias
- Radiation Oncology Unit, 2nd Department of Radiology, School of Medicine, National and Kapodistrian University of Athens (NKUOA), 12462 Athens, Greece;
| | - Stella Arelaki
- Translational Functional Cancer Genomics, National Center for Tumor Diseases, German Cancer Research Center, 69120 Heidelberg, Germany;
| | - Anna Zygogianni
- Radiation Oncology Unit, 1st Department of Radiology, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens (NKUOA), 11528 Athens, Greece; (I.M.K.); (A.Z.)
| |
Collapse
|
9
|
An J, Hu X, Liu F. Current understanding of cancer stem cells: Immune evasion and targeted immunotherapy in gastrointestinal malignancies. Front Oncol 2023; 13:1114621. [PMID: 36910604 PMCID: PMC9996315 DOI: 10.3389/fonc.2023.1114621] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
As a relatively rare population of cancer cells existing in the tumor microenvironment, cancer stem cells (CSCs) possess properties of immune privilege to evade the attack of immune system, regulated by the microenvironment of CSCs, the so-called CSCs niche. The bidirectional interaction of CSCs with tumor microenvironment (TME) components favors an immunosuppressive shelter for CSCs' survival and maintenance. Gastrointestinal cancer stem cells (GCSCs) are broadly regarded to be intimately involved in tumor initiation, progression, metastasis and recurrence, with elevated tumor resistance to conventional therapies, which pose a major hindrance to the clinical efficacy for treated patients with gastrointestinal malignancies. Thus, a multitude of efforts have been made to combat and eradicate GCSCs within the tumor mass. Among diverse methods of targeting CSCs in gastrointestinal malignancies, immunotherapy represents a promising strategy. And the better understanding of GCSCs immunomodulation and immunoresistance mechanisms is beneficial to guide and design novel GCSCs-specific immunotherapies with enhanced immune response and clinical efficacy. In this review, we have gathered available and updated information to present an overview of the immunoevasion features harbored by cancer stem cells, and we focus on the description of immune escape strategies utilized by CSCs and microenvironmental regulations underlying CSCs immuno-suppression in the context of gastrointestinal malignancies. Importantly, this review offers deep insights into recent advances of CSC-targeting immunotherapeutic approaches in gastrointestinal cancers.
Collapse
Affiliation(s)
- Junyi An
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohua Hu
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Liu
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Cancer Stem Cells in Hepatocellular Carcinoma: Intrinsic and Extrinsic Molecular Mechanisms in Stemness Regulation. Int J Mol Sci 2022; 23:ijms232012327. [PMID: 36293184 PMCID: PMC9604119 DOI: 10.3390/ijms232012327] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/18/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
Hepatocellular carcinoma (HCC) remains the most predominant type of liver cancer with an extremely poor prognosis due to its late diagnosis and high recurrence rate. One of the culprits for HCC recurrence and metastasis is the existence of cancer stem cells (CSCs), which are a small subset of cancer cells possessing robust stem cell properties within tumors. CSCs play crucial roles in tumor heterogeneity constitution, tumorigenesis, tumor relapse, metastasis, and resistance to anti-cancer therapies. Elucidation of how these CSCs maintain their stemness features is essential for the development of CSCs-based therapy. In this review, we summarize the present knowledge of intrinsic molecules and signaling pathways involved in hepatic CSCs, especially the CSC surface markers and associated signaling in regulating the stemness characteristics and the heterogeneous subpopulations within the CSC pool. In addition, we recapitulate the effects of crucial extrinsic cellular components in the tumor microenvironment, including stromal cells and immune cells, on the modulation of hepatic CSCs. Finally, we synopsize the currently valuable CSCs-targeted therapy strategies based on intervention in these intrinsic and extrinsic molecular mechanisms, in the hope of shedding light on better clinical management of HCC patients.
Collapse
|
11
|
Liao F, Zhang J, Hu Y, Najafabadi AH, Moon JJ, Wicha MS, Kaspo B, Whitfield J, Chang AE, Li Q. Efficacy of an ALDH peptide-based dendritic cell vaccine targeting cancer stem cells. Cancer Immunol Immunother 2022; 71:1959-1973. [DOI: 10.1007/s00262-021-03129-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 12/08/2021] [Indexed: 11/29/2022]
|
12
|
Sahoo OS, Pethusamy K, Srivastava TP, Talukdar J, Alqahtani MS, Abbas M, Dhar R, Karmakar S. The metabolic addiction of cancer stem cells. Front Oncol 2022; 12:955892. [PMID: 35957877 PMCID: PMC9357939 DOI: 10.3389/fonc.2022.955892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer stem cells (CSC) are the minor population of cancer originating cells that have the capacity of self-renewal, differentiation, and tumorigenicity (when transplanted into an immunocompromised animal). These low-copy number cell populations are believed to be resistant to conventional chemo and radiotherapy. It was reported that metabolic adaptation of these elusive cell populations is to a large extent responsible for their survival and distant metastasis. Warburg effect is a hallmark of most cancer in which the cancer cells prefer to metabolize glucose anaerobically, even under normoxic conditions. Warburg's aerobic glycolysis produces ATP efficiently promoting cell proliferation by reprogramming metabolism to increase glucose uptake and stimulating lactate production. This metabolic adaptation also seems to contribute to chemoresistance and immune evasion, a prerequisite for cancer cell survival and proliferation. Though we know a lot about metabolic fine-tuning in cancer, what is still in shadow is the identity of upstream regulators that orchestrates this process. Epigenetic modification of key metabolic enzymes seems to play a decisive role in this. By altering the metabolic flux, cancer cells polarize the biochemical reactions to selectively generate "onco-metabolites" that provide an added advantage for cell proliferation and survival. In this review, we explored the metabolic-epigenetic circuity in relation to cancer growth and proliferation and establish the fact how cancer cells may be addicted to specific metabolic pathways to meet their needs. Interestingly, even the immune system is re-calibrated to adapt to this altered scenario. Knowing the details is crucial for selective targeting of cancer stem cells by choking the rate-limiting stems and crucial branch points, preventing the formation of onco-metabolites.
Collapse
Affiliation(s)
- Om Saswat Sahoo
- Department of Biotechnology, National Institute of technology, Durgapur, India
| | - Karthikeyan Pethusamy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | - Joyeeta Talukdar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, United Kingdom
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
- Computers and communications Department, College of Engineering, Delta University for Science and Technology, Gamasa, Egypt
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
13
|
Singh D, Khan MA, Siddique HR. Specific targeting of cancer stem cells by immunotherapy: A possible stratagem to restrain cancer recurrence and metastasis. Biochem Pharmacol 2022; 198:114955. [PMID: 35181312 DOI: 10.1016/j.bcp.2022.114955] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSCs), the tumor-initiating cells playing a crucial role in cancer progression, recurrence, and metastasis, have the intrinsic property of self-renewal and therapy resistance. The tumorigenic properties of these cells include generation of cellular heterogeneity and immuno-suppressive tumor microenvironment (TME), conferring them the capability to resist a variety of anti-cancer therapeutics. Further, CSCs possess several unique immunological properties that help them escape recognition by the innate and adaptive immune system and shape a TME into a pro-tumorigenic and immunosuppressive landscape. In this context, immunotherapy is considered one of the best therapeutic options for eliminating CSCs to halt cancer recurrence and metastasis. In this review, we discuss the various immunomodulatory properties of CSCs and the interaction of CSCs with the immune system enabling immune evasion. In addition, we also highlight the present research update on immunotherapeutic targeting of CSCs and the possible further scope of research on this topic. We believe that a deeper understanding of CSCs' immunological properties and the crosstalk between CSCs and the immune system can develop better innovative immune-therapeutics and enhance the efficacy of current therapy-resistant cancer treatments.
Collapse
Affiliation(s)
- Deepti Singh
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Mohammad Afsar Khan
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
14
|
Cancer stem cells in hepatocellular carcinoma - from origin to clinical implications. Nat Rev Gastroenterol Hepatol 2022; 19:26-44. [PMID: 34504325 DOI: 10.1038/s41575-021-00508-3] [Citation(s) in RCA: 218] [Impact Index Per Article: 109.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2021] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive disease with a poor clinical outcome. The cancer stem cell (CSC) model states that tumour growth is powered by a subset of tumour stem cells within cancers. This model explains several clinical observations in HCC (as well as in other cancers), including the almost inevitable recurrence of tumours after initial successful chemotherapy and/or radiotherapy, as well as the phenomena of tumour dormancy and treatment resistance. The past two decades have seen a marked increase in research on the identification and characterization of liver CSCs, which has encouraged the design of novel diagnostic and treatment strategies for HCC. These studies revealed novel aspects of liver CSCs, including their heterogeneity and unique immunobiology, which are suggestive of opportunities for new research directions and potential therapies. In this Review, we summarize the present knowledge of liver CSC markers and the regulators of stemness in HCC. We also comprehensively describe developments in the liver CSC field with emphasis on experiments utilizing single-cell transcriptomics to understand liver CSC heterogeneity, lineage-tracing and cell-ablation studies of liver CSCs, and the influence of the CSC niche and tumour microenvironment on liver cancer stemness, including interactions between CSCs and the immune system. We also discuss the potential application of liver CSC-based therapies for treatment of HCC.
Collapse
|
15
|
Esmaeili SA, Sahranavard S, Salehi A, Bagheri V. Selectively targeting cancer stem cells: Current and novel therapeutic strategies and approaches in the effective eradication of cancer. IUBMB Life 2021; 73:1045-1059. [PMID: 34184810 DOI: 10.1002/iub.2524] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022]
Abstract
Cancer stem cells (CSCs) are a subgroup of cells in malignant cancers, which possess self-renewal capacity, tumor-initiating capability, and pluripotency, as well as being responsible for tumor maintenance, metastasis, relapse, and chemoresistance. The treatment modalities previously established for cancer included surgery, chemotherapy, and radiotherapy. The majority of tumor cells of non-CSCs could be eradicated using conventional chemotherapy and radiotherapy. Therefore, novel and promising therapeutic strategies that selectively target CSCs are of great importance. In this review, we described different therapeutic strategies such as immunotherapy, metabolism-based therapeutic strategies, and additional potential therapeutic approaches (targeting microRNAs [miRNAs], histone deacetylase, and DNA methyl transferase) against CSCs. Taken together, due to the inefficiency of anticancer single therapies, targeting CSCs through their metabolism and using immunotherapy and miRNAs besides classical chemo- and radiotherapy may exert better therapeutic effects.
Collapse
Affiliation(s)
- Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shamim Sahranavard
- Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Astireh Salehi
- Biology Department, Islamic Azad University, Sanandaj, Iran
| | - Vahid Bagheri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
16
|
Enderle L, Shalaby KH, Gorelik M, Weiss A, Blazer LL, Paduch M, Cardarelli L, Kossiakoff A, Adams JJ, Sidhu SS. A T cell redirection platform for co-targeting dual antigens on solid tumors. MAbs 2021; 13:1933690. [PMID: 34190031 PMCID: PMC8253144 DOI: 10.1080/19420862.2021.1933690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
In order to direct T cells to specific features of solid cancer cells, we engineered a bispecific antibody format, named Dual Antigen T cell Engager (DATE), by fusing a single-chain variable fragment targeting CD3 to a tumor-targeting antigen-binding fragment. In this format, multiple novel paratopes against different tumor antigens were able to recruit T-cell cytotoxicity to tumor cells in vitro and in an in vivo pancreatic ductal adenocarcinoma xenograft model. Since unique surface antigens in solid tumors are limited, in order to enhance selectivity, we further engineered “double-DATEs” targeting two tumor antigens simultaneously. The double-DATE contains an additional autonomous variable heavy-chain domain, which binds a second tumor antigen without itself eliciting a cytotoxic response. This novel modality provides a strategy to enhance the selectivity of immune redirection through binary targeting of native tumor antigens. The modularity and use of a common, stable human framework for all components enables a pipeline approach to rapidly develop a broad repertoire of tailored DATEs and double-DATEs with favorable biophysical properties and high potencies and selectivities.
Collapse
Affiliation(s)
- Leonie Enderle
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Karim H Shalaby
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Maryna Gorelik
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Alexander Weiss
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Levi L Blazer
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Marcin Paduch
- Institute for Biophysical Dynamics, Gordon Center for Integrative Science, Chicago, USA
| | - Lia Cardarelli
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Anthony Kossiakoff
- Institute for Biophysical Dynamics, Gordon Center for Integrative Science, Chicago, USA.,Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, USA
| | - Jarrett J Adams
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Sachdev S Sidhu
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
17
|
Dai X, Guo Y, Hu Y, Bao X, Zhu X, Fu Q, Zhang H, Tong Z, Liu L, Zheng Y, Zhao P, Fang W. Immunotherapy for targeting cancer stem cells in hepatocellular carcinoma. Theranostics 2021; 11:3489-3501. [PMID: 33537099 PMCID: PMC7847682 DOI: 10.7150/thno.54648] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
The rapid development and remarkable success of checkpoint inhibitors have provided significant breakthroughs in cancer treatment, including hepatocellular carcinoma (HCC). However, only 15-20% of HCC patients can benefit from checkpoint inhibitors. Cancer stem cells (CSCs) are responsible for recurrence, metastasis, and local and systemic therapy resistance in HCC. Accumulating evidence has suggested that HCC CSCs can create an immunosuppressive microenvironment through certain intrinsic and extrinsic mechanisms, resulting in immune evasion. Intrinsic evasion mechanisms mainly include activation of immune-related CSC signaling pathways, low-level expression of antigen presenting molecules, and high-level expression of immunosuppressive molecules. External evasion mechanisms are mainly related to HBV/HCV infection, alcoholic/nonalcoholic steatohepatitis, hypoxia stimulation, abnormal angiogenesis, and crosstalk between CSCs and immune cells. A better understanding of the complex mechanisms of CSCs involved in immune evasion will contribute to therapies for HCC. Here we will outline the detailed mechanisms of immune evasion for CSCs, and provide an overview of the current immunotherapies targeting CSCs in HCC.
Collapse
|
18
|
Cancer Stem Cells: Acquisition, Characteristics, Therapeutic Implications, Targeting Strategies and Future Prospects. Stem Cell Rev Rep 2020; 15:331-355. [PMID: 30993589 DOI: 10.1007/s12015-019-09887-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since last two decades, the major cancer research has focused on understanding the characteristic properties and mechanism of formation of Cancer stem cells (CSCs), due to their ability to initiate tumor growth, self-renewal property and multi-drug resistance. The discovery of the mechanism of acquisition of stem-like properties by carcinoma cells via epithelial-mesenchymal transition (EMT) has paved a way towards a deeper understanding of CSCs and presented a possible avenue for the development of therapeutic strategies. In spite of years of research, various challenges, such as identification of CSC subpopulation, lack of appropriate experimental models, targeting cancer cells and CSCs specifically without harming normal cells, are being faced while dealing with CSCs. Here, we discuss the biology and characteristics of CSCs, mode of acquisition of stemness (via EMT) and development of multi-drug resistance, the role of tumor niche, the process of dissemination and metastasis, therapeutic implications of CSCs and necessity of targeting them. We emphasise various strategies being developed to specifically target CSCs, including those targeting biomarkers, key pathways and microenvironment. Finally, we focus on the challenges that need to be subdued and propose the aspects that need to be addressed in future studies in order to broaden the understanding of CSCs and develop novel strategies to eradicate them in clinical applications. Graphical Abstract Cancer Stem Cells(CSCs) have gained much attention in the last few decades due to their ability to initiate tumor growth and, self-renewal property and multi-drug resistance. Here, we represent the CSC model of cancer, Characteristics of CSCs, acquisition of stemness and metastatic dissemination of cancer, Therapeutic implications of CSCs and Various strategies being employed to target and eradicate CSCs.
Collapse
|
19
|
Immunotherapy: Newer Therapeutic Armamentarium against Cancer Stem Cells. JOURNAL OF ONCOLOGY 2020; 2020:3963561. [PMID: 32211043 PMCID: PMC7085385 DOI: 10.1155/2020/3963561] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 02/04/2020] [Indexed: 12/15/2022]
Abstract
Mounting evidence from the literature suggests the existence of a subpopulation of cancer stem cells (CSCs) in almost all types of human cancers. These CSCs possessing a self-renewal capacity inhabit primary tumors and are more defiant to standard antimitotic and molecularly targeted therapies which are used for eliminating actively proliferating and differentiated cancer cells. Clinical relevance of CSCs emerges from the fact that they are the root cause of therapy resistance, relapse, and metastasis. Earlier, surgery, chemotherapy, and radiotherapy were established as cancer treatment modalities, but recently, immunotherapy is also gaining importance in the management of various cancer patients, mostly those of the advanced stage. This review abridges potential off-target effects of inhibiting CSC self-renewal pathways on immune cells and some recent immunological studies specifically targeting CSCs on the basis of their antigen expression profile, even though molecular markers or antigens that have been described till date as expressed by cancer stem cells are not specifically expressed by these cells which is a major limitation to target CSCs. We propose that owing to CSC stemness property to mediate immunotherapy response, we can apply a combination therapy approach by targeting CSCs and tumor microenvironment (TME) along with conventional treatment strategies as an effective means to eradicate cancer cells.
Collapse
|
20
|
Gastric Cancer Stem Cells: Current Insights into the Immune Microenvironment and Therapeutic Targets. Biomedicines 2020; 8:biomedicines8010007. [PMID: 31935894 PMCID: PMC7168269 DOI: 10.3390/biomedicines8010007] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/27/2019] [Accepted: 01/04/2020] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is a leading cause of cancer-related death worldwide. Cancer stem cells (CSCs) are known to be involved in chemotherapy resistance and the development of metastases. Although CSCs harbor self-renewal and tumorigenic abilities, the immune microenvironment surrounding CSCs provides various factors and supports the maintenance of CSC properties. The current review summarizes the accumulating findings regarding the relationship between the immune microenvironment and gastric CSCs (GCSCs), which will support the possibility of developing novel therapeutic strategies for targeting GCSCs.
Collapse
|
21
|
Gupta PK, Dharanivasan G, Misra R, Gupta S, Verma RS. Nanomedicine in Cancer Stem Cell Therapy. Nanobiomedicine (Rij) 2020. [DOI: 10.1007/978-981-32-9898-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
22
|
Alvarado-Ortiz E, Sarabia-Sánchez MÁ, García-Carrancá A. Molecular Mechanisms Underlying the Functions of Cellular Markers Associated with the Phenotype of Cancer Stem Cells. Curr Stem Cell Res Ther 2019; 14:405-420. [PMID: 30147013 DOI: 10.2174/1574888x13666180821154752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/18/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
Cancer Stem Cells (CSC) generally constitute a minor cellular population within tumors that exhibits some capacities of normal Stem Cells (SC). The existence of CSC, able to self-renew and differentiate, influences central aspects of tumor biology, in part because they can continue tumor growth, give rise to metastasis, and acquire drug and radioresistance, which open new avenues for therapeutics. It is well known that SC constantly interacts with their niche, which includes mesenchymal cells, extracellular ligands, and the Extra Cellular Matrix (ECM). These interactions regularly lead to homeostasis and maintenance of SC characteristics. However, the exact participation of each of these components for CSC maintenance is not clear, as they appear to be context- or cell-specific. In the recent past, surface cellular markers have been fundamental molecular tools for identifying CSC and distinguishing them from other tumor cells. Importantly, some of these cellular markers have been shown to possess functional roles that affect central aspects of CSC. Likewise, some of these markers can participate in regulating the interaction of CSC with their niche, particularly the ECM. We focused this review on the molecular mechanisms of surface cellular markers commonly employed to identify CSC, highlighting the signaling pathways and mechanisms involved in CSC-ECM interactions, through each of the cellular markers commonly used in the study of CSC, such as CD44, CD133, CD49f, CD24, CXCR4, and LGR5. Their presence does not necessarily implicate them in CSC biology.
Collapse
Affiliation(s)
- Eduardo Alvarado-Ortiz
- Programa de Maestría y Doctorado en Ciencias Biológicas, Facultad de Ciencias, Universidad Nacional Autónoma de México, México City, México.,Laboratory of Virus and Cancer, Unidad de Investigacion Biomedica en Cáncer, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico & Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Ciudad de Mexico, Mexico
| | - Miguel Á Sarabia-Sánchez
- Laboratory of Virus and Cancer, Unidad de Investigacion Biomedica en Cáncer, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico & Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Ciudad de Mexico, Mexico.,Programa de Maestría y Doctorado en Ciencias Bioquímicas, Facultad de Química, Universidad Nacional Autónoma de México, , México City, México
| | - Alejandro García-Carrancá
- Laboratory of Virus and Cancer, Unidad de Investigacion Biomedica en Cáncer, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico & Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Ciudad de Mexico, Mexico
| |
Collapse
|
23
|
Ruan S, Lin M, Zhu Y, Lum L, Thakur A, Jin R, Shao W, Zhang Y, Hu Y, Huang S, Hurt EM, Chang AE, Wicha MS, Li Q. Integrin β4-Targeted Cancer Immunotherapies Inhibit Tumor Growth and Decrease Metastasis. Cancer Res 2019; 80:771-783. [PMID: 31843981 DOI: 10.1158/0008-5472.can-19-1145] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/30/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022]
Abstract
Integrin β4 (ITGB4) has been shown to play an important role in the regulation of cancer stem cells (CSC). Immune targeting of ITGB4 represents a novel approach to target this cell population, with potential clinical benefit. We developed two immunologic strategies to target ITGB4: ITGB4 protein-pulsed dendritic cells (ITGB4-DC) for vaccination and adoptive transfer of anti-CD3/anti-ITGB4 bispecific antibody (ITGB4 BiAb)-armed tumor-draining lymph node T cells. Two immunocompetent mouse models were utilized to assess the efficacy of these immunotherapies in targeting both CSCs and bulk tumor populations: 4T1 mammary tumors and SCC7 head and neck squamous carcinoma cell line. Immunologic targeting of ITGB4 utilizing either ITGB4-DC or ITGB4 BiAb-T cells significantly inhibited local tumor growth and metastases in both the 4T1 and SCC7 tumor models. Furthermore, the efficacy of both of these ITGB4-targeted immunotherapies was significantly enhanced by the addition of anti-PD-L1. Both ITGB4-targeted immunotherapies induced endogenous T-cell cytotoxicity directed at CSCs as well as non-CSCs, which expressed ITGB4, and immune plasma-mediated killing of CSCs. As a result, ITGB4-targeted immunotherapy reduced not only the number of ITGB4high CSCs in residual 4T1 and SCC7 tumors but also their tumor-initiating capacity in secondary mouse implants. In addition, treated mice demonstrated no apparent toxicity. The specificity of these treatments was demonstrated by the lack of effects observed using ITGB4 knockout 4T1 or ITGB4-negative CT26 colon carcinoma cells. Because ITGB4 is expressed by CSCs across a variety of tumor types, these results support immunologic targeting of ITGB4 as a promising therapeutic strategy.Significance: This study identifies a novel mechanism of resistance to anti-PD-1/PD-L1 immunotherapy mediated by HPV E5, which can be exploited using the HPV E5 inhibitor rimantadine to improve outcomes for head and neck cancer patients.
Collapse
Affiliation(s)
- Shasha Ruan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.,Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ming Lin
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.,Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yong Zhu
- Guangzhou Improve Medical Instruments Co., Ltd. Guangzhou, Guangdong, China
| | - Lawrence Lum
- Division of Hematology/Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, Virginia
| | - Archana Thakur
- Division of Hematology/Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, Virginia
| | - Runming Jin
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenlong Shao
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Yalei Zhang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Yangyang Hu
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shiang Huang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | - Alfred E Chang
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Max S Wicha
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Qiao Li
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
24
|
Razi E, Radak M, Mahjoubin-Tehran M, Talebi S, Shafiee A, Hajighadimi S, Moradizarmehri S, Sharifi H, Mousavi N, Sarvizadeh M, Nejati M, Taghizadeh M, Ghasemi F. Cancer stem cells as therapeutic targets of pancreatic cancer. Fundam Clin Pharmacol 2019; 34:202-212. [PMID: 31709581 DOI: 10.1111/fcp.12521] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 11/02/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022]
Abstract
The discovery of stem cells and their potential abilities in self-renewal and differentiation has opened a new horizon in medicine. Scientists have found a small population of stem cells in some types of cancers with the same functions as normal stem cells. There are two models for tumor progression: clonal (stochastic) and cancer stem cell (CSCs) models. According to the first model, all transformed cells in the tumor have carcinogenic potential and are able to proliferate and produce the same cells. The latter model, which has received more attention recently, considers the role of CSCs in drug resistance and tumor metastasis. Following the model, researchers have found that targeting CSCs may be a promising way in cancer therapy. This review describes CSC characteristics in general, while also focusing on CSC properties in the context of pancreatic cancer.
Collapse
Affiliation(s)
- Ebrahim Razi
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Mehran Radak
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Talebi
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alimohammad Shafiee
- Division of General Internal Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Sarah Hajighadimi
- Division of General Internal Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Sanaz Moradizarmehri
- Division of General Internal Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Hossein Sharifi
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Nousin Mousavi
- Department of Surgery, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mostafa Sarvizadeh
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Majid Nejati
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
25
|
Clara JA, Monge C, Yang Y, Takebe N. Targeting signalling pathways and the immune microenvironment of cancer stem cells - a clinical update. Nat Rev Clin Oncol 2019; 17:204-232. [PMID: 31792354 DOI: 10.1038/s41571-019-0293-2] [Citation(s) in RCA: 440] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs) have important roles in tumour development, relapse and metastasis; the intrinsic self-renewal characteristics and tumorigenic properties of these cells provide them with unique capabilities to resist diverse forms of anticancer therapy, seed recurrent tumours, and disseminate to and colonize distant tissues. The findings of several studies indicate that CSCs originate from non-malignant stem or progenitor cells. Accordingly, inhibition of developmental signalling pathways that are crucial for stem and progenitor cell homeostasis and function, such as the Notch, WNT, Hedgehog and Hippo signalling cascades, continues to be pursued across multiple cancer types as a strategy for targeting the CSCs hypothesized to drive cancer progression - with some success in certain malignancies. In addition, with the renaissance of anticancer immunotherapy, a better understanding of the interplay between CSCs and the tumour immune microenvironment might be the key to unlocking a new era of oncological treatments associated with a reduced propensity for the development of resistance and with enhanced antimetastatic activity, thus ultimately resulting in improved patient outcomes. Herein, we provide an update on the progress to date in the clinical development of therapeutics targeting the Notch, WNT, Hedgehog and Hippo pathways. We also discuss the interactions between CSCs and the immune system, including the potential immunological effects of agents targeting CSC-associated developmental signalling pathways, and provide an overview of the emerging approaches to CSC-targeted immunotherapy.
Collapse
Affiliation(s)
- Joseph A Clara
- National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Cecilia Monge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Naoko Takebe
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
26
|
Abstract
Cancer stem cells (CSCs) are crucial for tumor recurrence and distant metastasis. Immunologically targeting CSCs represents a promising strategy to improve efficacy of multimodal cancer therapy. Modulating the innate immune response involving Toll-like receptors, macrophages, natural killer cells, and γδT cells has therapeutic effects on CSCs. Antigens expressed by CSCs provide specific targets for immunotherapy. CSC-primed dendritic cell-based vaccines have induced significant antitumor immunity as an adjuvant therapy in experimental models of established tumors. Targeting the tumor microenvironment CSC niche with cytokines or checkpoint blockade provides additional strategies to eliminate CSCs.
Collapse
Affiliation(s)
- Jing Zhang
- Division of Surgical Oncology, University of Michigan Rogel Cancer Center, Room 3410, 1150 East Medical Center Drive, Ann Arbor, MI 48109, USA; Department of the 2nd Thoracic Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Road, Hongshan District, Wuhan, Hubei Province 430070, China
| | - Qiao Li
- Division of Surgical Oncology, University of Michigan Rogel Cancer Center, 3520B MSRB-1, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Alfred E Chang
- Division of Surgical Oncology, University of Michigan Rogel Cancer Center, Room 3304, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
27
|
Shirjang S, Alizadeh N, Mansoori B, Mahmoodpoor A, Kafil HS, Hojjat-Farsangi M, Yousefi M. Promising immunotherapy: Highlighting cytokine-induced killer cells. J Cell Biochem 2018; 120:8863-8883. [PMID: 30556298 DOI: 10.1002/jcb.28250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
For many years, cancer therapy has appeared to be a challenging issue for researchers and physicians. By the introduction of novel methods in immunotherapy, the prospect of cancer therapy even more explained than before. Cytokine-induced killer (CIK) cell-based immunotherapy demonstrated to have potentiality in improving clinical outcomes and relieving major side effects of standard treatment options. In addition, given the distinctive features such as high safety, low toxicity effects on healthy cells, numerous clinical trials conducted on CIK cells. Due to the shortcomings that observed in CIK cell immunotherapy alone, arising a tendency to make modifications (combined modality therapy or combination therapy) including the addition of various types of cytokines, genetic engineering, combination with immune checkpoints, and so on. In this review, we have tried to bring forth the latest immunotherapy methods and their overview. We have discussed the combination therapies with CIK cells and the conducted clinical trials. This helps the future studies to use integrated therapies with CIK cells as a promising treatment of many types of cancers.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Nunes T, Hamdan D, Leboeuf C, El Bouchtaoui M, Gapihan G, Nguyen TT, Meles S, Angeli E, Ratajczak P, Lu H, Di Benedetto M, Bousquet G, Janin A. Targeting Cancer Stem Cells to Overcome Chemoresistance. Int J Mol Sci 2018; 19:E4036. [PMID: 30551640 PMCID: PMC6321478 DOI: 10.3390/ijms19124036] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/08/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Cancers are heterogeneous at the cell level, and the mechanisms leading to cancer heterogeneity could be clonal evolution or cancer stem cells. Cancer stem cells are resistant to most anti-cancer treatments and could be preferential targets to reverse this resistance, either targeting stemness pathways or cancer stem cell surface markers. Gold nanoparticles have emerged as innovative tools, particularly for photo-thermal therapy since they can be excited by laser to induce hyperthermia. Gold nanoparticles can be functionalized with antibodies to specifically target cancer stem cells. Preclinical studies using photo-thermal therapy have demonstrated the feasibility of targeting chemo-resistant cancer cells to reverse clinical chemoresistance. Here, we review the data linking cancer stem cells and chemoresistance and discuss the way to target them to reverse resistance. We particularly focus on the use of functionalized gold nanoparticles in the treatment of chemo-resistant metastatic cancers.
Collapse
Affiliation(s)
- Toni Nunes
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Diaddin Hamdan
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Hôpital de La Porte Verte, F-78004 Versailles, France.
| | - Christophe Leboeuf
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Morad El Bouchtaoui
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Guillaume Gapihan
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Thi Thuy Nguyen
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
| | - Solveig Meles
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
| | - Eurydice Angeli
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
| | - Philippe Ratajczak
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - He Lu
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Mélanie Di Benedetto
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
- Université Paris 13, F-93430 Villetaneuse, France.
| | - Guilhem Bousquet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
- Université Paris 13, F-93430 Villetaneuse, France.
- Service d'Oncologie Médicale, AP-HP-Hôpital Avicenne, F-93008 Bobigny, France.
| | - Anne Janin
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
- Service de Pathologie, AP-HP-Hôpital Saint-Louis, F-75010 Paris, France.
| |
Collapse
|
29
|
Golay J, Martinelli S, Alzani R, Cribioli S, Albanese C, Gotti E, Pasini B, Mazzanti B, Saccardi R, Rambaldi A, Introna M. Cord blood-derived cytokine-induced killer cells combined with blinatumomab as a therapeutic strategy for CD19 + tumors. Cytotherapy 2018; 20:1077-1088. [PMID: 30093325 DOI: 10.1016/j.jcyt.2018.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 04/17/2018] [Accepted: 08/06/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND Cytokine-induced killer cells (CIKs) are an advanced therapeutic medicinal product (ATMP) that has shown therapeutic activity in clinical trials but needs optimization. We developed a novel strategy using CIKs from banked cryopreserved cord blood units (CBUs) combined with bispecific antibody (BsAb) blinatumomab to treat CD19+ malignancies. METHODS CB-CIKs were expanded in vitro and fully characterized in comparison with peripheral blood (PB)-derived CIKs. RESULTS CB-CIKs, like PB-CIKs, were mostly CD3+ T cells with mean 45% CD3+CD56+ and expressing mostly TCR(T cell receptor)αβ with a TH1 phenotype. CB-CIK cultures had, however, a larger proportion of CD4+ cells, mostly CD56-, as well as a greater proportion of naïve CCR7+CD45RA+ and a lower percentage of effector memory cells, compared with PB-CIKs. CB-CIKs were very similar to PB-CIKs in their expression of a large panel of co-stimulatory and inhibitory/exhaustion markers, except for higher CD28 expression among CD8+ cells. Like PB-CIKs, CB-CIKs were highly cytotoxic in vitro against natural killer (NK) cell targets and efficiently lysed CD19+ tumor cells in the presence of blinatumomab, with 30-60% lysis of target cells at very low effector:target ratios. Finally, both CB-CIKs and PB-CIKs, combined with blinatumomab, showed significant therapeutic activity in an aggressive PDX Ph+ CD19+ acute lymphoblastic leukemia model in NOD-SCID mice, without sign of toxicity or graft-versus-host disease. The improved expansion protocol was finally validated in good manufacturing practice conditions, showing reproducible expansion of CIKs from cryopreserved cord blood units with a median of 28.8 × 106 CIK/kg. DISCUSSION We conclude that CB-CIKs, combined with bispecific T-cell-engaging antibodies, offer a novel, effective treatment strategy for leukemia.
Collapse
Affiliation(s)
- Josée Golay
- Center of Cellular Therapy "G. Lanzani", Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy; Fondazione per la Ricerca Ospedale Maggiore, Bergamo, Italy
| | - Simona Martinelli
- Center of Cellular Therapy "G. Lanzani", Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | | | | | | | - Elisa Gotti
- Center of Cellular Therapy "G. Lanzani", Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Bruna Pasini
- Obstetrics and Gynecology Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Benedetta Mazzanti
- Cord Blood Bank, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Riccardo Saccardi
- Cord Blood Bank, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplantation Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy; Department of Oncology and Hemato-oncology, University of Milan, Italy
| | - Martino Introna
- Center of Cellular Therapy "G. Lanzani", Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy.
| |
Collapse
|
30
|
Kholodenko IV, Kalinovsky DV, Doronin II, Deyev SM, Kholodenko RV. Neuroblastoma Origin and Therapeutic Targets for Immunotherapy. J Immunol Res 2018; 2018:7394268. [PMID: 30116755 PMCID: PMC6079467 DOI: 10.1155/2018/7394268] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/27/2018] [Indexed: 01/30/2023] Open
Abstract
Neuroblastoma is a pediatric solid cancer of heterogeneous clinical behavior. The unique features of this type of cancer frequently hamper the process of determining clinical presentation and predicting therapy effectiveness. The tumor can spontaneously regress without treatment or actively develop and give rise to metastases despite aggressive multimodal therapy. In recent years, immunotherapy has become one of the most promising approaches to the treatment of neuroblastoma. Still, only one drug for targeted immunotherapy of neuroblastoma, chimeric monoclonal GD2-specific antibodies, is used in the clinic today, and its application has significant limitations. In this regard, the development of effective and safe GD2-targeted immunotherapies and analysis of other potential molecular targets for the treatment of neuroblastoma represents an important and topical task. The review summarizes biological characteristics of the origin and development of neuroblastoma and outlines molecular markers of neuroblastoma and modern immunotherapy approaches directed towards these markers.
Collapse
Affiliation(s)
- Irina V. Kholodenko
- Orekhovich Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow 119121, Russia
| | - Daniel V. Kalinovsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Igor I. Doronin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
- Real Target LLC, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Sergey M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
- Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University “MEPhI”, Moscow 115409, Russia
| | - Roman V. Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
- Real Target LLC, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| |
Collapse
|
31
|
Emerging functional markers for cancer stem cell-based therapies: Understanding signaling networks for targeting metastasis. Semin Cancer Biol 2018; 53:90-109. [PMID: 29966677 DOI: 10.1016/j.semcancer.2018.06.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 12/18/2022]
Abstract
Metastasis is one of the most challenging issues in cancer patient management, and effective therapies to specifically target disease progression are missing, emphasizing the urgent need for developing novel anti-metastatic therapeutics. Cancer stem cells (CSCs) gained fast attention as a minor population of highly malignant cells within liquid and solid tumors that are responsible for tumor onset, self-renewal, resistance to radio- and chemotherapies, and evasion of immune surveillance accelerating recurrence and metastasis. Recent progress in the identification of their phenotypic and molecular characteristics and interactions with the tumor microenvironment provides great potential for the development of CSC-based targeted therapies and radical improvement in metastasis prevention and cancer patient prognosis. Here, we report on newly uncovered signaling mechanisms controlling CSC's aggressiveness and treatment resistance, and CSC-specific agents and molecular therapeutics, some of which are currently under investigation in clinical trials, gearing towards decisive functional CSC intrinsic or surface markers. One special research focus rests upon subverted regulatory pathways such as insulin-like growth factor 1 receptor signaling and its interactors in metastasis-initiating cell populations directly related to the gain of stem cell- and EMT-associated properties, as well as key components of the E2F transcription factor network regulating metastatic progression, microenvironmental changes, and chemoresistance. In addition, the study provides insight into systems biology tools to establish complex molecular relationships behind the emergence of aggressive phenotypes from high-throughput data that rely on network-based analysis and their use to investigate immune escape mechanisms or predict clinical outcome-relevant CSC receptor signaling signatures. We further propose that customized vector technologies could drastically enhance systemic drug delivery to target sites, and summarize recent progress and remaining challenges. This review integrates available knowledge on CSC biology, computational modeling approaches, molecular targeting strategies, and delivery techniques to envision future clinical therapies designed to conquer metastasis-initiating cells.
Collapse
|
32
|
Zhang D, Tang DG, Rycaj K. Cancer stem cells: Regulation programs, immunological properties and immunotherapy. Semin Cancer Biol 2018; 52:94-106. [PMID: 29752993 DOI: 10.1016/j.semcancer.2018.05.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023]
Abstract
It is becoming increasingly clear that virtually all types of human cancers harbor a small population of stem-like cancer cells (i.e., cancer stem cells, CSCs). These CSCs preexist in primary tumors, can self-renew and are more tolerant of standard treatments, such as antimitotic and molecularly targeted agents, most of which preferentially eliminate differentiated and proliferating cancer cells. CSCs are therefore postulated as the root of therapy resistance, relapse and metastasis. Aside from surgery, radiation, and chemotherapy, immunotherapy is now established as the fourth pillar in the therapeutic armamentarium for patients with cancer, especially late-stage and advanced cancers. A better understanding of CSC immunological properties should lead to development of novel immunologic approaches targeting CSCs, which, in turn, may help prevent tumor recurrence and eliminate residual diseases. Here, with a focus on CSCs in solid tumors, we review CSC regulation programs and recent transcriptomics-based immunological profiling data specific to CSCs. By highlighting CSC antigens that could potentially be immunogenic, we further discuss how CSCs can be targeted immunologically.
Collapse
Affiliation(s)
- Dingxiao Zhang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA; Key Lab of Agricultural Animal Genetics, Breeding & Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA; Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Kiera Rycaj
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
33
|
Liu J, Chen C, Ling C, Hu H, Cao J, Gao Y. The effects of hemocyanin on T cells cultured in vitro. Oncol Lett 2018; 15:2655-2660. [PMID: 29434988 DOI: 10.3892/ol.2017.7587] [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: 10/19/2016] [Accepted: 11/02/2017] [Indexed: 11/06/2022] Open
Abstract
As a broad-spectrum antibiotic, gentamicin is used extensively in T cell culturing in vitro, but preliminary studies have identified that T cell activity is significantly affected by gentamicin. In the present study, the hemocyanin from Litopenaeus vannamei (L. vannamei) was selected as an additive for T cell cultures in vitro. Compared with those in the control group, the cell quantity exhibited no significant difference, and the formation rate of cell colony increased gradually with increases in the hemocyanin concentration. Additionally, flow cytometry assays identified that cluster of differentiation (CD)3+CD4+ and CD4+CD25+ T cells in the 0.2 µg/ml hemocyanin (Hem 3) group were all significantly increased. Furthermore, cell cycle analysis demonstrated that T cells in the G0/G1 phase were significantly decreased in the Hem 3 group compared with in the control, 0.05 µg/ml (Hem 1) and 0.1 µg/ml (Hem 2) groups, and cells in the S phase were significantly elevated in the Hem 3 group compared with in the control and Hem 1 groups. In addition, MTT analysis indicated that the cytotoxicity of T cells towards HepG2 cells was significantly increased in the Hem 3 group compared with in the control, Hem 1 and Hem 2 groups. Taken together, the present study identified that hemocyanin may improve the proliferation and cytotoxicity of T cells, and the results supported the use of hemocyanin in T cell adoptive immunotherapy.
Collapse
Affiliation(s)
- Jiani Liu
- Department of Immunology, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Cong Chen
- Department of Immunology, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Chen Ling
- Department of Immunology, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Haoyun Hu
- Department of Immunology, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jingsong Cao
- Department of Immunology, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yongqiang Gao
- Department of Immunology, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
34
|
Abstract
Harnessing the power of the human immune system to treat cancer is the essence of immunotherapy. Monoclonal antibodies engage the innate immune system to destroy targeted cells. For the last 30years, antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity have been the main mechanisms of anti-tumor action of unconjugated antibody drugs. Efforts to exploit the potentials of other immune cells, in particular T cells, culminated in the recent approval of two T cell engaging bispecific antibody (T-BsAb) drugs, thereby stimulating new efforts to accelerate similar platforms through preclinical and clinical trials. In this review, we have compiled the worldwide effort in exploring T cell engaging bispecific antibodies. Our special emphasis is on the lessons learned, with the hope to derive insights in this fast evolving field with tremendous clinical potential.
Collapse
Affiliation(s)
- Z Wu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - N V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
35
|
Sahin IH, Askan G, Hu ZI, O’Reilly EM. Immunotherapy in pancreatic ductal adenocarcinoma: an emerging entity? Ann Oncol 2017; 28:2950-2961. [PMID: 28945842 PMCID: PMC5834032 DOI: 10.1093/annonc/mdx503] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The genomic-plasticity of the immune system creates a broad immune repertoire engaged to tackle cancer cells. Promising clinical activity has been observed with several immune therapy strategies in solid tumors including melanoma, lung, kidney, and bladder cancers, albeit as yet immunotherapy-based treatment approaches in pancreatic ductal adenocarcinoma (PDAC) remain to have proven value. While translational and early clinical studies have demonstrated activation of antitumor immunity, most recent late-phase clinical trials have not confirmed the early promise in PDAC except in MSI-High PDAC patients. These results may in part be explained by multiple factors, including the poorly immunogenic nature of PDAC along with immune privilege, the complex tumor microenvironment, and the genetic plasticity of PDAC cells. These challenges have led to disappointments in the field, nonetheless they have also advanced our understanding that may tailor the future steps for immunotherapy for PDAC. Therefore, there is significant hope that progress is on the horizon.
Collapse
Affiliation(s)
- I H Sahin
- Department of Medicine, Emory University School of Medicine, Atlanta
| | - G Askan
- Department of Pathology, Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - Z I Hu
- Department of Medicine, Icahn School of Medicine, Mount Sinai Health System, New York
| | - E M O’Reilly
- Department of Pathology, Pathology, Memorial Sloan Kettering Cancer Center, New York
- Department of Medicine, Weill Cornell Medicine, New York, USA
| |
Collapse
|
36
|
Cao J, Chen C, Gao Y, Hu L, Liang Y, Xiao J. Identification of a protein associated with the activity of cytokine-induced killer cells. Oncol Lett 2017; 14:6937-6942. [PMID: 29163711 DOI: 10.3892/ol.2017.7042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/14/2017] [Indexed: 12/19/2022] Open
Abstract
Cytokine-induced killer cells (CIKs) adoptive immunotherapy for efficient antitumor ability is used clinically, but details regarding the proteins associated with CIK activity remain unclear. In the current study, the cytotoxicity of CIKs on hepatoma was identified to be significantly downregulated by 1.61-fold following gentamincin treatment. Further research revealed that a differentially expressed protein (P43) was significantly downregulated by 1.22-fold using one-dimensional gel electrophoresis analysis. Of these, the P43 was identified as human haptoglobin using liquid chromatography-mass spectrometry. Western blotting demonstrated that the haptoglobin specifically reacted with rabbit anti-human-haptoglobin. Furthermore, western blotting results verified that the haptoglobin was significantly downregulated by 1.17-fold compared with the control group. In addition, the expression of haptoglobin mRNA was significantly downregulated by 1.73-fold following gentamincin treatment. Taken together, the results of the present study demonstrated that the expression of haptoglobin protein was associated with the activity of CIKs, and the results will be beneficial to the further investigation of CIK activity-enhancement mechanism.
Collapse
Affiliation(s)
- Jingsong Cao
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Cong Chen
- Laboratory Department, The Second Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yongqiang Gao
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Li Hu
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yu Liang
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jianhua Xiao
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
37
|
Ramos EK, Hoffmann AD, Gerson SL, Liu H. New Opportunities and Challenges to Defeat Cancer Stem Cells. Trends Cancer 2017; 3:780-796. [PMID: 29120754 PMCID: PMC5958547 DOI: 10.1016/j.trecan.2017.08.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 12/13/2022]
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells that are capable of self-renewal, proliferation, differentiation, plastic adaptation, and immune regulation, thereby mediating tumorigenesis, metastasis, and therapy resistance. CSCs are associated with cancer progression and clinical outcome in cancer patients. Successful targeting of CSCs will therefore be necessary to eradicate and cure cancer. Functional regulators of stem cell (stemness) signaling pathways in human cancers have brought new opportunities to target CSCs and reframe cancer-targeting strategies in clinical settings. However, challenges remain due to a lack of complete understanding of CSC plasticity/heterogeneity and the limited efficacy of individual stemness inhibitors in cancer treatment. In this article we review CSC signaling pathways and the current state of CSC-targeting therapeutics in combinatory treatments in clinical trials.
Collapse
Affiliation(s)
- Erika K Ramos
- Driskill Graduate Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department Pharmacology, Northwestern University, Chicago, IL, USA; These authors equally contributed to the manuscript preparation
| | - Andrew D Hoffmann
- Department Pharmacology, Northwestern University, Chicago, IL, USA; These authors equally contributed to the manuscript preparation
| | - Stanton L Gerson
- The Case Comprehensive Cancer Center, Cleveland, OH, USA; The National Center for Regenerative Medicine, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| | - Huiping Liu
- Department Pharmacology, Northwestern University, Chicago, IL, USA; The Case Comprehensive Cancer Center, Cleveland, OH, USA; The National Center for Regenerative Medicine, Cleveland, OH, USA; Department of Medicine, Division of Hematology and Oncology, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
38
|
Schmohl JU, Gleason MK, Dougherty PR, Miller JS, Vallera DA. Heterodimeric Bispecific Single Chain Variable Fragments (scFv) Killer Engagers (BiKEs) Enhance NK-cell Activity Against CD133+ Colorectal Cancer Cells. Target Oncol 2017; 11:353-61. [PMID: 26566946 DOI: 10.1007/s11523-015-0391-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Natural killer (NK) cells are potent cytotoxic lymphocytes that play a critical role in tumor immunosurveillance and control. Cancer stem cells (CSC) initiate and sustain tumor cell growth, mediate drug refractory cancer relapse, and express the well-known surface marker CD133. METHODS DNA fragments from two fully humanized single chain fragment variable (scFv) antibodies recognizing CD16 on NK-cells and CD133 on CSC were genetically spliced forming a novel drug, 16 × 133 BiKE that simultaneously recognizes these antigens to facilitate an immunologic synapse. The anti-CD133 was created using a fusion protein prepared by fusing DNA fragments encoding the two extracellular domains of CD133. Immunization of mice with the resulting fusion protein generated a unique antibody that recognized the molecular framework and was species cross-reactive. RESULTS In vitro chromium-51 ((51)Cr) release cytotoxicity assays at both high and low effector:target ratios demonstrated the ability of the heterodimeric biological drug to greatly enhance NK-cell killing of human Caco-2 colorectal carcinoma cells known to overexpress CD133. The tumor associated antigen specificity of the drug for CD133 even enhanced NK-cell cytotoxicity against the NK-resistant human Burkitt's lymphoma Daudi cell line, which has less than 5 % CD133 surface expression. Flow cytometry analysis revealed increases in NK-cell degranulation and Interferon-γ production upon co-culture with Caco-2 targets in the presence of the drug. CONCLUSION These studies demonstrate that the innate immune system can be effectively recruited to kill CSC using bispecific antibodies targeting CD133 and that this anti-CD133 scFv may be useful in this bispecific platform or perhaps in the design of more complex trispecific molecules for carcinoma therapy.
Collapse
Affiliation(s)
- J U Schmohl
- University of Minnesota Masonic Cancer Center, Section of Molecular Cancer Therapeutics, Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN, USA.,Department for Hematology and Oncology, Medicine Department 2, University Hospital of Tuebingen, Tuebingen, 72076, Germany
| | - M K Gleason
- University of Minnesota Masonic Cancer Center, Adult Division of Hematology, Oncology and Transplantation Oncology, University of Minnesota, Minneapolis, MN, USA.,Medical Science Liaison, Immuno-Oncology, Novartis Oncology/Sandoz, Minneapolis, MN, USA
| | - P R Dougherty
- University of Minnesota Masonic Cancer Center, Section of Molecular Cancer Therapeutics, Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN, USA
| | - J S Miller
- University of Minnesota Masonic Cancer Center, Adult Division of Hematology, Oncology and Transplantation Oncology, University of Minnesota, Minneapolis, MN, USA
| | - D A Vallera
- University of Minnesota Masonic Cancer Center, Section of Molecular Cancer Therapeutics, Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
39
|
Chen Y, Sun W, He R, Zhang F, Wang H, Li P, Shao RG, Xu X. Lidamycin decreases CD133 expression in hepatocellular carcinoma via the Notch signaling pathway. Oncol Lett 2017; 14:7889-7895. [PMID: 29344233 DOI: 10.3892/ol.2017.7248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 09/13/2017] [Indexed: 12/15/2022] Open
Abstract
Cluster of differentiation (CD)133 is considered a molecular marker of cancer stem cells in hepatocellular carcinoma. In the present study, the effect of lidamycin (LDM) on CD133 expression in hepatocellular carcinoma (Huh7 cells) was evaluated and the potential molecular mechanism was investigated. Flow cytometry analysis, as well as sorting, sphere formation and western-blot assays, were performed in vitro to explore the effects of LDM on CD133 expression. A subcutaneous tumor model in nude mice was used to observe the effects of LDM on tumor volume and CD133 protein in vivo. To investigate the potential underlying molecular mechanism, Notch signaling pathway activity was detected by western blot analysis and reverse transcription-quantitative polymerase chain reaction. The proportion of CD133+ cells and the expression of CD133 protein were revealed to be downregulated by LDM. Sphere formation of sorted CD133+ cells was suppressed 7 days after LDM treatment. In addition, LDM inhibited tumor volume formed from sorted CD133+ cells and CD133 protein level in vivo. LDM decreased the mRNA level of NOTCH1, Hes1 (Hes family BHLH transcription factor 1) and Hey1 (Hes-related family BHLH transcription factor with YRPW motif 1) genes; consequently, the protein expression of NOTCH1, Notch intracellular domain, Hes1 and Hey1 was decreased by LDM. Downregulation of the Notch signaling pathway by LDM was enhanced through combination with N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester. In brief, these data suggest that LDM suppresses CD133 expression via the Notch signaling pathway, indicating the potential mechanism of LDM on CD133 and the benefits for further clinical application.
Collapse
Affiliation(s)
- Yi Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Wenwei Sun
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Ran He
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Feiyan Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Hongyu Wang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Panhong Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Rong-Guang Shao
- Laboratory of Oncology, Ministry of Health Key Laboratory of Antibiotic Bioengineering, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Xiaoyu Xu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| |
Collapse
|
40
|
Pützer BM, Solanki M, Herchenröder O. Advances in cancer stem cell targeting: How to strike the evil at its root. Adv Drug Deliv Rev 2017; 120:89-107. [PMID: 28736304 DOI: 10.1016/j.addr.2017.07.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/10/2017] [Accepted: 07/16/2017] [Indexed: 12/18/2022]
Abstract
Cancer progression to metastatic stages is still unmanageable and the promise of effective anti-metastatic therapy remains largely unmet, emphasizing the need to develop novel therapeutics. The special focus here is on cancer stem cells (CSC) as the seed of tumor initiation, epithelial-mesenchymal transition, chemoresistance and, as a consequence, drivers of metastatic dissemination. We report on targeted therapies gearing towards the CSC's internal and membrane-anchored markers using agents such as antibody derivatives, nucleic therapeutics, small molecules and genetic payloads. Another emphasis lies on novel proceedings envisaged to deliver current and prospective therapies to the target sites using newest viral and non-viral vector technologies. In this review, we summarize recent progress and remaining challenges in therapeutic strategies to combat CSC.
Collapse
Affiliation(s)
- Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany.
| | - Manish Solanki
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| | - Ottmar Herchenröder
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| |
Collapse
|
41
|
Sato-Dahlman M, Miura Y, Huang JL, Hajeri P, Jacobsen K, Davydova J, Yamamoto M. CD133-targeted oncolytic adenovirus demonstrates anti-tumor effect in colorectal cancer. Oncotarget 2017. [PMID: 29100290 DOI: 10.18632/oncotarget.18340.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Oncolytic Adenoviruses (OAds) are one of the most promising anti-cancer agents that can induce cancer specific cell death. Recently, we generated infectivity-selective OAd, and the resultant OAd tumor-specific binding shows strong efficacy and mitigates toxicity. In this study, we applied this strategy based on adenovirus library screening system for generation of CD133-targeted OAd, and examined their oncolytic activity against colorectal cancer (CRC) in vitro and in vivo. CD133 (Prominin-1) is an important cell surface marker of cancer stem (like) cells (CSCs) in various cancers, including CRC. Elimination of CSCs has a high likelihood to improve CRC treatment because CSCs population in the tumor contributes to recurrence, metastases, chemotherapy resistance, and poor survival. The OAd with CD133-targeting motif (AdML-TYML) selectively infected CD133+ cultured cells and lysed them efficiently. Treatment with AdML-TYML prior to tumor inoculation inhibited the establishment of tumor of CD133+ CRC cell lines in nude mice. AdML-TYML also showed strong antitumor effect after intratumoral injections in already established CD133+ CRC subcutaneous xenografts. Our results indicate that CD133-targeted OAd selectively infected CD133+ CRC, and exhibited anti-tumorigenicity and therapeutic effect in established tumors. This novel infectivity selective virus could be a potent tool for the prevention of metastases and relapses in CRC.
Collapse
Affiliation(s)
| | - Yoshiaki Miura
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jing Li Huang
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Kari Jacobsen
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Julia Davydova
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
42
|
Sato-Dahlman M, Miura Y, Huang JL, Hajeri P, Jacobsen K, Davydova J, Yamamoto M. CD133-targeted oncolytic adenovirus demonstrates anti-tumor effect in colorectal cancer. Oncotarget 2017; 8:76044-76056. [PMID: 29100290 PMCID: PMC5652684 DOI: 10.18632/oncotarget.18340] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 04/19/2017] [Indexed: 12/19/2022] Open
Abstract
Oncolytic Adenoviruses (OAds) are one of the most promising anti-cancer agents that can induce cancer specific cell death. Recently, we generated infectivity-selective OAd, and the resultant OAd tumor-specific binding shows strong efficacy and mitigates toxicity. In this study, we applied this strategy based on adenovirus library screening system for generation of CD133-targeted OAd, and examined their oncolytic activity against colorectal cancer (CRC) in vitro and in vivo. CD133 (Prominin-1) is an important cell surface marker of cancer stem (like) cells (CSCs) in various cancers, including CRC. Elimination of CSCs has a high likelihood to improve CRC treatment because CSCs population in the tumor contributes to recurrence, metastases, chemotherapy resistance, and poor survival. The OAd with CD133-targeting motif (AdML-TYML) selectively infected CD133+ cultured cells and lysed them efficiently. Treatment with AdML-TYML prior to tumor inoculation inhibited the establishment of tumor of CD133+ CRC cell lines in nude mice. AdML-TYML also showed strong antitumor effect after intratumoral injections in already established CD133+ CRC subcutaneous xenografts. Our results indicate that CD133-targeted OAd selectively infected CD133+ CRC, and exhibited anti-tumorigenicity and therapeutic effect in established tumors. This novel infectivity selective virus could be a potent tool for the prevention of metastases and relapses in CRC.
Collapse
Affiliation(s)
| | - Yoshiaki Miura
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jing Li Huang
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Kari Jacobsen
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Julia Davydova
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
43
|
Bozorg-Ghalati F, Hedayati M, Dianatpour M, Azizi F, Mosaffa N, Mehrabani D. Effects of a Phosphoinositide-3-Kinase Inhibitor on Anaplastic Thyroid Cancer Stem Cells. Asian Pac J Cancer Prev 2017; 18:2287-2291. [PMID: 28843268 PMCID: PMC5697493 DOI: 10.22034/apjcp.2017.18.8.2287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background: Thyroidectomy, radioactive iodine therapy, chemotherapy, or their combination are treatments of choice for thyroid cancers. However, cancer stem cells (CSCs) may become resistant to therapy, and mutations in somatic genes affect radioiodine uptake. This study determined the effect of a phosphoinositide-3-kinase (PI3K) inhibitor on anaplastic thyroid CSCs. Materials and Methods: The magnetic-activated cell sorting assay was used for segregating CD133-positive CSCs from three anaplastic thyroid carcinoma (ATC) cell lines (C643, SW1736, and 8305C). After confirming the cells’ purity by flow cytometry, they were treated with 5, 10, 20, or 25 μM LY294002, a PI3K inhibitor, and then evaluated at 24 and 48 h. The sodium-iodide symporter (NIS) mRNA level was determined using the quantitative real-time polymerase chain reaction. NIS protein expression was evaluated using western blotting. Results: The PI3K inhibitor, at different concentrations and times, increased the NIS mRNA level (1.30-6.17-fold, P < 0.0001). If the NIS mRNA level in LY294002-treated CD133-positive CSCs was increased more than 2-fold, the NIS protein content was detectable. Conclusions: CD133-positive CSCs isolated from ATC cell lines expressed NIS mRNA and protein after PI3K inhibition. Our findings suggest that molecularly targeted CSC therapy may improve the treatment efficacy of aggressive cancers like ATC.
Collapse
Affiliation(s)
- Farzaneh Bozorg-Ghalati
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. , dianatpour@sums.
ac.ir
| | | | | | | | | | | |
Collapse
|
44
|
Fang D, Kitamura H. Cancer stem cells and epithelial-mesenchymal transition in urothelial carcinoma: Possible pathways and potential therapeutic approaches. Int J Urol 2017; 25:7-17. [PMID: 28697535 DOI: 10.1111/iju.13404] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/22/2017] [Indexed: 12/12/2022]
Abstract
There is growing evidence of the presence of cancer stem cells in urothelial carcinoma. Cancer stem cells have the ability to self-renew and to differentiate into all cell types of the original heterogeneous tumor. A panel of diverse cancer stem cell markers might be suitable for simulation studies of urothelial cancer stem cells and for the development of optimized treatment protocols. The present review focuses on the advances in recognizing the markers of urothelial cancer stem cells and possible therapeutic targets. The commonly reported markers and pathways that were evaluated include CD44, CD133, ALDH1, SOX2 & SOX4, BMI1, EZH1, PD-L1, MAGE-A3, COX2/PGE2/STAT3, AR, and autophagy. Studies on the epithelial-mesenchymal transition-related pathways (Shh, Wnt/β-catenin, Notch, PI3K/Akt, TGF-β, miRNA) are also reviewed. Most of these markers were recognized through the expression patterns of cancer stem cell-rich side populations. Their regulative role in the development and differentiation of urothelial cancer stem cells was confirmed in vitro by functional analyses (e.g. cell migration, colony formation, sphere formation), and in vivo in xenograft experiments. Although a small number of these pathways are targeted by currently available drugs or drugs that are the currently being tested in clinical trials, a clear treatment approach has not been developed for most pathways. A greater understanding of the mechanisms that control the proliferation and differentiation of cancer stem cells is expected to lead to improvements in targeted therapy.
Collapse
Affiliation(s)
- Dong Fang
- Department of Urology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama, Japan.,Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| | - Hiroshi Kitamura
- Department of Urology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama, Japan
| |
Collapse
|
45
|
Gao X, Mi Y, Guo N, Xu H, Xu L, Gou X, Jin W. Cytokine-Induced Killer Cells As Pharmacological Tools for Cancer Immunotherapy. Front Immunol 2017; 8:774. [PMID: 28729866 PMCID: PMC5498561 DOI: 10.3389/fimmu.2017.00774] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/19/2017] [Indexed: 12/31/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are a heterogeneous population of effector CD3+CD56+ natural killer T cells, which can be easily expanded in vitro from peripheral blood mononuclear cells. CIK cells work as pharmacological tools for cancer immunotherapy as they exhibit MHC-unrestricted, safe, and effective antitumor activity. Much effort has been made to improve CIK cells cytotoxicity and treatments of CIK cells combined with other antitumor therapies are applied. This review summarizes some strategies, including the combination of CIK with additional cytokines, dendritic cells, check point inhibitors, antibodies, chemotherapeutic agents, nanomedicines, and engineering CIK cells with a chimeric antigen receptor. Furthermore, we briefly sum up the clinical trials on CIK cells and compare the effect of clinical CIK therapy with other immunotherapies. Finally, further research is needed to clarify the pharmacological mechanism of CIK and provide evidence to formulate uniform culturing criteria for CIK expansion.
Collapse
Affiliation(s)
- Xingchun Gao
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, Key Lab for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yajing Mi
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Na Guo
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Hao Xu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Lixian Xu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Weilin Jin
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China.,Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, Key Lab for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China.,National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
46
|
Geddert H, Braun A, Kayser C, Dimmler A, Faller G, Agaimy A, Haller F, Moskalev EA. Epigenetic Regulation of CD133 in Gastrointestinal Stromal Tumors. Am J Clin Pathol 2017; 147:515-524. [PMID: 28398518 DOI: 10.1093/ajcp/aqx028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES This study ascertained the regulation of the stem cell marker CD133 and its potential applicability for prognostication of gastrointestinal stromal tumors (GISTs). METHODS A total of 95 resected GISTs were included in the study. CD133 protein expression was assessed immunohistochemically on tissue microarrays. Methylation percentage was quantified by pyrosequencing. Gene expression in cell lines GIST48b and GIST882 upon treatment with DNA demethylation agent 5-aza-2'-deoxycytidine was analyzed by quantitative polymerase chain reaction. RESULTS The expression of hypermethylated CD133 could be reactivated in the GIST cell line upon hypomethylation with the drug. Similarly, in patient material, CD133 methylation percentage correlated inversely with the protein expression and reflected tumor size with hypermethylation in small (<2 cm) tumors and virtually no methylation in large (>10 cm) GISTs. The gene's methylation percentage and expression level were clearly specific to anatomic sites and distinct driver mutations. KIT -mutant gastric GISTs exhibited significantly lower methylation degrees and concomitant high CD133 protein abundance compared with KIT -mutant GISTs from the small intestine. CD133 hypermethylation was documented in PDGFRA -mutant gastric GISTs along with low CD133 expression compared with KIT -mutant gastric GISTs. High CD133 expression was a prognosticator of shorter disease-free survival in all patients. In a subgroup of KIT -mutant gastric GISTs, low CD133 methylation degree was correlated with a shorter disease-free survival. CONCLUSIONS Our results strongly suggest epigenetic regulation of CD133 expression by promoter methylation in GISTs. Pending further validation studies, high abundance of the protein can serve as a marker for malignant GISTs.
Collapse
Affiliation(s)
- Helene Geddert
- Institute for Pathology, St Vincentius Hospital, Karlsruhe, Germany
| | - Alexander Braun
- Institute for Pathology, Albert Ludwigs University, Freiburg, Germany
| | - Claudia Kayser
- Institute for Pathology, Albert Ludwigs University, Freiburg, Germany
| | - Arno Dimmler
- Institute for Pathology, St Vincentius Hospital, Karlsruhe, Germany
| | - Gerhard Faller
- Institute for Pathology, St Vincentius Hospital, Karlsruhe, Germany
| | - Abbas Agaimy
- Institute for Pathology, Friedrich Alexander University, Erlangen, Germany
| | - Florian Haller
- Institute for Pathology, Friedrich Alexander University, Erlangen, Germany
| | - Evgeny A Moskalev
- Institute for Pathology, Friedrich Alexander University, Erlangen, Germany
| |
Collapse
|
47
|
Song Y, Wang Y, Tong C, Xi H, Zhao X, Wang Y, Chen L. A unified model of the hierarchical and stochastic theories of gastric cancer. Br J Cancer 2017; 116:973-989. [PMID: 28301871 PMCID: PMC5396111 DOI: 10.1038/bjc.2017.54] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/16/2017] [Accepted: 01/26/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is a life-threatening disease worldwide. Despite remarkable advances in treatments for GC, it is still fatal to many patients due to cancer progression, recurrence and metastasis. Regarding the development of novel therapeutic techniques, many studies have focused on the biological mechanisms that initiate tumours and cause treatment resistance. Tumours have traditionally been considered to result from somatic mutations, either via clonal evolution or through a stochastic model. However, emerging evidence has characterised tumours using a hierarchical organisational structure, with cancer stem cells (CSCs) at the apex. Both stochastic and hierarchical models are reasonable systems that have been hypothesised to describe tumour heterogeneity. Although each model alone inadequately explains tumour diversity, the two models can be integrated to provide a more comprehensive explanation. In this review, we discuss existing evidence supporting a unified model of gastric CSCs, including the regulatory mechanisms of this unified model in addition to the current status of stemness-related targeted therapy in GC patients.
Collapse
Affiliation(s)
- Yanjing Song
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Yao Wang
- Department of Immunology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China
| | - Chuan Tong
- Department of Immunology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China
| | - Hongqing Xi
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Xudong Zhao
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Yi Wang
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Lin Chen
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
48
|
Ikemoto T, Shimada M, Yamada S. Pathophysiology of recurrent hepatocellular carcinoma after radiofrequency ablation. Hepatol Res 2017; 47:23-30. [PMID: 26990590 DOI: 10.1111/hepr.12705] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/10/2016] [Accepted: 03/10/2016] [Indexed: 12/13/2022]
Abstract
Radiofrequency ablation (RFA) is effective for the local control of hepatocellular carcinoma (HCC), particularly when a patient's liver functional reserve does not allow radical resection. There is controversy regarding the superiority of surgical resection compared with RFA for such patients, particularly those with three or fewer tumors with diameters ≤3 cm. Moreover, HCC often recurs after RFA, and the tumor cells show distinct phenotypic changes. Incomplete ablation accounts for tumor recurrence, and recent studies provide new insights into the biological mechanisms responsible for the pathological changes of HCC after RFA. This review focuses on the roles of epithelial-mesenchymal transition and cancer stemness that are driven by a mechanism that involves microRNA-mediated upregulation of hypoxia-inducible factor-1. The studies reviewed here provide compelling evidence that complete ablation of HCC is required to prevent recurrence and indicate that further research is urgently required to develop a new systematic strategy to prevent tumor recurrence by targeting hypoxia-inducible factor-1.
Collapse
Affiliation(s)
- Tetsuya Ikemoto
- Department of Surgery, Tokushima University, Tokushima City, Japan
| | - Mitsuo Shimada
- Department of Surgery, Tokushima University, Tokushima City, Japan
| | | |
Collapse
|
49
|
Liu Y, Liu H, Liu H, He P, Li J, Liu X, Chen L, Wang M, Xi J, Wang H, Zhang H, Zhu Y, Zhu W, Ning J, Guo C, Sun C, Zhang M. Dendritic cell-activated cytokine-induced killer cell-mediated immunotherapy is safe and effective for cancer patients >65 years old. Oncol Lett 2016; 12:5205-5210. [PMID: 28105230 DOI: 10.3892/ol.2016.5337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/25/2016] [Indexed: 01/17/2023] Open
Abstract
Individuals >65 years old account for a large proportion of cancer patients, and usually have poor prognoses due to relative weaker physiological function and lower drug tolerance. To characterize the efficacy and safety of dendritic cell (DC)-activated cytokine-induced killer cell (CIK)-mediated treatment, and develop an adoptive immunotherapy for cancer patients >65 years old, a retrospective study was performed in 58 cancer sufferers who received 1-4 cycles of DC-activated CIK (DC-CIK) treatment and evaluated the response (tumor remission rate) and toxicity (side effects to the treatment). The present results showed that DCs and CIKs could be expanded rapidly in vitro, and following co-culture with DCs, the population of cluster of differentiation (CD) 3+, CD3+CD4+, CD3+CD8+ and CD3+CD56+ CIKs was significantly increased compared to CIKs without DC activation (P=0.044). In addition, DC-CIK infusion produced marked clinical outcomes, resulting in an objective remission rate, overall clinical benefit rate and Karnofsky performance status of 44.83, 75.86 and 87.28±5.46%, respectively, which was significantly improved compared with prior to treatment (P<0.05). Additionally, subsequent to two cycles of this immunotherapy, several tumor marker expression levels declined, returning to the normal range. The proportion of CD3+CD4+ (P=0.017) and CD3+CD8+ (P=0.023) lymphocytes, and the population of CD4/CD8 cells (P=0.024) were also increased. In conclusion, the present study suggests that the immunotherapy mediated by DC-CIK is safe and effective for cancer patients aged >65 years.
Collapse
Affiliation(s)
- Yanfeng Liu
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Haibo Liu
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Hausheng Liu
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Pengcheng He
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Li
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xin Liu
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Limei Chen
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Mengchang Wang
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jiejing Xi
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Huaiyu Wang
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Haitao Zhang
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ying Zhu
- Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wei Zhu
- Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Ning
- Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Caili Guo
- Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Chunhong Sun
- Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Mei Zhang
- Department of Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Biological Immune Therapy Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
50
|
Wei F, Rong XX, Xie RY, Jia LT, Wang HY, Qin YJ, Chen L, Shen HF, Lin XL, Yang J, Yang S, Hao WC, Chen Y, Xiao SJ, Zhou HR, Lin TY, Chen YS, Sun Y, Yao KT, Xiao D. Cytokine-induced killer cells efficiently kill stem-like cancer cells of nasopharyngeal carcinoma via the NKG2D-ligands recognition. Oncotarget 2016; 6:35023-39. [PMID: 26418951 PMCID: PMC4741506 DOI: 10.18632/oncotarget.5280] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/04/2015] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are considered to be the root cause for cancer treatment failure. Thus, there remains an urgent need for more potent and safer therapies against CSCs for curing cancer. In this study, the antitumor activity of cytokine-induced killer (CIK) cells against putative CSCs of nasopharyngeal carcinoma (NPC) was fully evaluated in vitro and in vivo. To visualize putative CSCs in vitro by fluorescence imaging, and image and quantify putative CSCs in tumor xenograft-bearing mice by in vivo bioluminescence imaging, NPC cells were engineered with CSC detector vector encoding GFP and luciferase (Luc) under control of Nanog promoter. Our study reported in vitro intense tumor-killing activity of CIK cells against putative CSCs of NPC, as revealed by percentage analysis of side population cells, tumorsphere formation assay and Nanog-promoter-GFP-Luc reporter gene strategy plus time-lapse recording. Additionally, time-lapse imaging firstly illustrated that GFP-labeled or PKH26-labeled putative CSCs or tumorspheres were usually attacked simultaneously by many CIK cells and finally killed by CIK cells, suggesting the necessity of achieving sufficient effector-to-target ratios. We firstly confirmed that NKG2D blockade by anti-NKG2D antibody significantly but partially abrogated CIK cell-mediated cytolysis against putative CSCs. More importantly, intravenous infusion of CIK cells significantly delayed tumor growth in NOD/SCID mice, accompanied by a remarkable reduction in putative CSC number monitored by whole-body bioluminescence imaging. Taken together, our findings suggest that CIK cells demonstrate the intense tumor-killing activity against putative CSCs of NPC, at least in part, by NKG2D-ligands recognition. These results indicate that CIK cell-based therapeutic strategy against CSCs presents a promising and safe approach for cancer treatment.
Collapse
Affiliation(s)
- Fang Wei
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China
| | - Xiao-Xiang Rong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Rao-Ying Xie
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Li-Ting Jia
- Department of Pathology, Guilin Medical College, Guilin 541001, China
| | - Hui-Yan Wang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yu-Juan Qin
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Lin Chen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Hong-Fen Shen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Lin Lin
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jie Yang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Sheng Yang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Wei-Chao Hao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yan Chen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Sheng-Jun Xiao
- Department of Pathology, Guilin Medical College, Guilin 541001, China
| | - Hui-Rong Zhou
- Department of Pathology, Guilin Medical College, Guilin 541001, China
| | - Tao-Yan Lin
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yu-Shuang Chen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yan Sun
- Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kai-Tai Yao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Dong Xiao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|