1
|
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
|
2
|
Mamun MA, Mannoor K, Cao J, Qadri F, Song X. SOX2 in cancer stemness: tumor malignancy and therapeutic potentials. J Mol Cell Biol 2021; 12:85-98. [PMID: 30517668 PMCID: PMC7109607 DOI: 10.1093/jmcb/mjy080] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 11/18/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs), a minor subpopulation of tumor bulks with self-renewal and seeding capacity to generate new tumors, posit a significant challenge to develop effective and long-lasting anti-cancer therapies. The emergence of drug resistance appears upon failure of chemo-/radiation therapy to eradicate the CSCs, thereby leading to CSC-mediated clinical relapse. Accumulating evidence suggests that transcription factor SOX2, a master regulator of embryonic and induced pluripotent stem cells, drives cancer stemness, fuels tumor initiation, and contributes to tumor aggressiveness through major drug resistance mechanisms like epithelial-to-mesenchymal transition, ATP-binding cassette drug transporters, anti-apoptotic and/or pro-survival signaling, lineage plasticity, and evasion of immune surveillance. Gaining a better insight and comprehensive interrogation into the mechanistic basis of SOX2-mediated generation of CSCs and treatment failure might therefore lead to new therapeutic targets involving CSC-specific anti-cancer strategies.
Collapse
Affiliation(s)
- Mahfuz Al Mamun
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Kaiissar Mannoor
- Oncology Laboratory, Institute for Developing Science & Health Initiatives (ideSHi), Dhaka, Bangladesh
| | - Jun Cao
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Firdausi Qadri
- Oncology Laboratory, Institute for Developing Science & Health Initiatives (ideSHi), Dhaka, Bangladesh
| | - Xiaoyuan Song
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Brain Function and Disease, CAS Center for Excellence in Molecular Cell Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
| |
Collapse
|
3
|
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
|
4
|
Dhodapkar MV, Dhodapkar KM. Moving Immunoprevention Beyond Virally Mediated Malignancies: Do We Need to Link It to Early Detection? Front Immunol 2019; 10:2385. [PMID: 31649683 PMCID: PMC6795703 DOI: 10.3389/fimmu.2019.02385] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/23/2019] [Indexed: 12/23/2022] Open
Abstract
Vaccines can successfully prevent viral infections and have emerged as an effective strategy for preventing some virally mediated malignancies. They also represent our major hope for cost-effective reduction of the cancer burden. The concept that the immune system mediates surveillance and editing roles against tumors is now well-established in murine models. However, harnessing the immune system to prevent human cancers that do not have a known viral etiology has not yet been realized. Most human cancers originate in a premalignant phase that is more common than the cancer itself. Many of the genetic changes that underlie carcinogenesis originate at this stage when the malignant phenotype is not manifest. Studies evaluating host response in human premalignancy have documented that these lesions are immunogenic, setting the stage for immune-based approaches for targeted prevention of human cancer. However, recent studies suggest that the hierarchy of T cell exhaustion and immune-suppressive factors have already begun to emerge in many preneoplastic states. These considerations underscore the need to link immune prevention to earlier detection of such lesions and to personalize such approaches based on the status of the pre-existing immune response.
Collapse
Affiliation(s)
- Madhav V. Dhodapkar
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Kavita M. Dhodapkar
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, School of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
5
|
Morrison BJ, Steel JC, Morris JC. Reduction of MHC-I expression limits T-lymphocyte-mediated killing of Cancer-initiating cells. BMC Cancer 2018; 18:469. [PMID: 29699516 PMCID: PMC5918869 DOI: 10.1186/s12885-018-4389-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 04/17/2018] [Indexed: 12/15/2022] Open
Abstract
Background It has been proposed that cancer establishment, maintenance, and recurrence may be attributed to a unique population of tumor cells termed cancer-initiating cells (CICs) that may include characteristics of putative cancer stem cell-like cells. Studies in lung cancer have shown that such cells can be enriched and propagated in vitro by culturing tumor cells in serum-free suspension as tumorspheres. CICs have been characterized for their phenotype, stem cell-like qualities, and their role in establishing tumor and maintaining tumor growth. Less is known about the interaction of CICs with the immune system. Methods We established CIC-enriched tumorspheres from murine TC-1 lung cancer cells, expressing human papillomavirus 16 (HPV-16) E6/E7 antigens, and evaluated their susceptibility to antitumor immune responses both in vitro and in vivo. Results TC-1 CICs demonstrated reduced expression of surface major histocompatibility complex (MHC)-I molecules compared to non-CICs. We similarly determined decreased MHC-I expression in five of six human lung cancer cell lines cultured under conditions enriching for CICs. In vivo, TC-1 cells enriched for CICs were resistant to human papillomavirus 16 E6/E7 peptide vaccine-mediated killing. We found that vaccinated mice challenged with CIC enriched tumorspheres demonstrated shorter survivals and showed significantly fewer CD8+ tumor infiltrating lymphocytes compared to CIC non-enriched challenged mice. Furthermore, cultured cytotoxic T lymphocytes (CTLs) from vaccinated mice demonstrated reduced capacity to lyse TC-1 cells enriched for CICs compared to non-enriched TC-1 cells. Following treatment with IFN-γ, both CIC enriched and non-enriched TC-1 cells expressed similar levels of MHC-I, and the increased MHC-I expression on CICs resulted in greater CTL-mediated tumor lysis and improved tumor-free survival in mice. Conclusions These results suggest that the attenuated expression of MHC-I molecules by CICs represents a potential strategy of CICs to escape immune recognition, and that the development of successful immunotherapy strategies targeting CICs may decrease their resistance to T cell-mediated immune detection by enhancing CIC MHC-I expression. Electronic supplementary material The online version of this article (10.1186/s12885-018-4389-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Brian J Morrison
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, Maryland, 20910, USA.
| | - Jason C Steel
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, Queensland, Australia
| | - John C Morris
- Division of Hematology-Oncology, University of Cincinnati Cancer Institute, University of Cincinnati, Cincinnati, Ohio, 45267, USA
| |
Collapse
|
6
|
Sultan M, Coyle KM, Vidovic D, Thomas ML, Gujar S, Marcato P. Hide-and-seek: the interplay between cancer stem cells and the immune system. Carcinogenesis 2017; 38:107-118. [PMID: 27866156 DOI: 10.1093/carcin/bgw115] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 11/15/2016] [Indexed: 12/26/2022] Open
Abstract
The enhanced ability of cancer stem cells (CSCs) to give rise to new tumors suggests that these cells may also have an advantage in evading immune detection and elimination. This tumor-forming ability, combined with the known plasticity of the immune system, which can play both protumorigenic and antitumorigenic roles, has motivated investigations into the interaction between CSCs and the immune system. Herein, we review the interplay between host immunity and CSCs by examining the immune-related mechanisms that favor CSCs and the CSC-mediated expansion of protumorigenic immune cells. Furthermore, we discuss immune cells, such as natural killer cells, that preferentially target CSCs and the strategies used by CSCs to evade immune detection and destruction. An increased understanding of these interactions and the pathways that regulate them may allow us to harness immune system components to create new adjuvant therapies that eradicate CSCs and improve patient survival.
Collapse
Affiliation(s)
| | | | | | | | - Shashi Gujar
- Department of Pathology and.,Department of Microbiology and Immunology, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada
| | - Paola Marcato
- Department of Pathology and.,Department of Microbiology and Immunology, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada
| |
Collapse
|
7
|
Zheng Q, Zheng Y, Chen J, You J, Zhu Y, Liu Y, Jiang JJ. A hepatic stem cell vaccine is superior to an embryonic stem cell vaccine in the prophylaxis and treatment of murine hepatocarcinoma. Oncol Rep 2017; 37:1716-1724. [PMID: 28098898 DOI: 10.3892/or.2017.5381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 10/31/2016] [Indexed: 11/05/2022] Open
Abstract
Stem cells and cancer cells express a common subset of antigens called oncofetal antigens. Theoretically, vaccination with stem cells is effective at boosting the preexisting anticancer immune response. Herein we describe the efficacy of two stem cell-based vaccines in the prophylaxis and treatment of subcutaneous hepatic tumors transplanted into mice. C57BL/6j mice were vaccinated weekly with either hepatic stem cells (HSCs) or embryonic stem cells (ESCs) for three weeks, followed by a subcutaneous challenge with Hepa 1-6 cells at one week (group 1) or four weeks (group 2) after vaccination. No tumor formation was observed in HSC-vaccinated mice when challenged within one week after vaccination (group 1), but tumors formed in 10% of mice in the ESC-vaccinated group and in 60% of mice in the unvaccinated group. When the long-term memory response was examined (group 2), only 10% of HSC-vaccinated mice and 20% of ESC-vaccinated mice developed macroscopic hepatocarcinomas compared to 60% of the unvaccinated mice. Besides their function as prophylactic vaccines, administration of either HSC or ESC could be a potential treatment for cancer. In mice with subcutaneous hepatocarcinomas, complete clearance of tumor burden was observed in 80% of mice receiving HSC vaccination, but 40% of ESC-vaccinated mice presented with tumors that did not increase in size over time. These data support that HSC is a superior vaccine candidate for durable antitumor protection in this hepatocarcinoma model.
Collapse
Affiliation(s)
- Qi Zheng
- Center for Liver Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Yichao Zheng
- Center for Liver Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Jing Chen
- Center for Liver Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Jia You
- Center for Liver Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Yueyong Zhu
- Center for Liver Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Yurui Liu
- Center for Liver Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Jia Ji Jiang
- Center for Liver Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| |
Collapse
|
8
|
Harnessing shared antigens and T-cell receptors in cancer: Opportunities and challenges. Proc Natl Acad Sci U S A 2016; 113:7944-5. [PMID: 27410048 DOI: 10.1073/pnas.1608860113] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
9
|
Sehgal K, Ragheb R, Fahmy TM, Dhodapkar MV, Dhodapkar KM. Nanoparticle-mediated combinatorial targeting of multiple human dendritic cell (DC) subsets leads to enhanced T cell activation via IL-15-dependent DC crosstalk. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:2297-305. [PMID: 25080481 PMCID: PMC6195217 DOI: 10.4049/jimmunol.1400489] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Most vaccines depend on coadministration of Ags and adjuvants that activate APCs. Nanoparticles (NPs) have emerged as an attractive vehicle for synchronized delivery of Ags and adjuvants to APCs and can be targeted to specific cell types, such as dendritic cells (DCs), which are potent APCs. Which subset of human DCs should be targeted for optimal activation of T cell immunity, however, remains unknown. In this article, we describe a poly-lactic-coglycolic acid-based NP platform, wherein avidin-decorated NPs can be targeted to multiple human DC subsets via biotinylated Abs. Both BDCA3(+) and monocyte-derived DC-SIGN(+) NP-loaded DCs were equally effective at generating Ag-specific human T cells in culture, including against complex peptide mixtures from viral and tumor Ags across multiple MHC molecules. Ab-mediated targeting of NPs to distinct DC subsets led to enhanced T cell immunity. However, combination targeting to both DC-SIGN and BDCA3(+) DCs led to significantly greater activation of T cells compared with targeting either DC subset alone. Enhanced T cell activation following combination targeting depended on DC-mediated cytokine release and was IL-15 dependent. These data demonstrate that simultaneous targeting of multiple DC subsets may improve NP vaccines by engaging DC crosstalk and provides a novel approach to improving vaccines against pathogens and tumors.
Collapse
Affiliation(s)
- Kartik Sehgal
- Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT 06510; Department of Medicine, Yale School of Medicine, Yale University, New Haven, CT 06510
| | - Ragy Ragheb
- Department of Biomedical Engineering, Yale School of Engineering, Yale University, New Haven, CT 06510
| | - Tarek M Fahmy
- Department of Biomedical Engineering, Yale School of Engineering, Yale University, New Haven, CT 06510; Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT 06510; and
| | - Madhav V Dhodapkar
- Department of Medicine, Yale School of Medicine, Yale University, New Haven, CT 06510; Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT 06510; and Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT 06510
| | - Kavita M Dhodapkar
- Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT 06510; Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT 06510
| |
Collapse
|
10
|
Pascutti F, Cunha LL, Rizzatti EG, Colleoni GWB. Understanding myeloma cancer stem cells. Immunotherapy 2014; 5:1291-4. [PMID: 24283839 DOI: 10.2217/imt.13.132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
11
|
Dhodapkar KM, Gettinger SN, Das R, Zebroski H, Dhodapkar MV. SOX2-specific adaptive immunity and response to immunotherapy in non-small cell lung cancer. Oncoimmunology 2013; 2:e25205. [PMID: 24073380 PMCID: PMC3782159 DOI: 10.4161/onci.25205] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 05/23/2013] [Accepted: 05/29/2013] [Indexed: 01/08/2023] Open
Abstract
Immunotherapeutic strategies including the blockade of programmed death 1 (PD-1) receptors hold promise for the treatment of various cancers including non-small cell lung carcinoma (NSCLC). Preclinical data suggest that pre-existing tumor immunity is important for disease regression upon checkpoint blockade-based therapies. However, the nature of antigen-specific T-cell responses that correlate with the clinical response to immunotherapy in NSCLC patients is not known. The embryonic stem cell gene SRY (sex determining region Y)-box 2 (SOX2) has recently emerged as a major oncogenic driver in NSCLC. Here, we show that nearly 50% of a cohort of NSCLC patients mounted both CD4+ and CD8+ T-cell responses against SOX2, which could be readily detected among peripheral blood mononuclear cells. T-cell responses against SOX2 were associated with NSCLC regression upon immunotherapy with anti-PD-1 monoclonal antibodies, whereas none of the patients lacking SOX2-specific T cells experienced disease regression following immune checkpoint blockade. Conversely, cellular and humoral responses against viral antigens or another tumor-associated antigen (NY-ESO-1) failed to correlate with the clinical response of NSCLC patients to immunotherapy. Of note, the administration of PD-1-blocking antibodies was associated with intramolecular epitope spread as well as with the amplification of SOX2-specific immune responses in vivo. These findings identify SOX2 as an important tumor-associated antigen in NSCLC and link the presence of SOX2-specific T cells with the clinical response of lung cancer patients to immunotherapy.
Collapse
Affiliation(s)
- Kavita M Dhodapkar
- Department of Pediatrics; Yale University; New Haven, CT USA ; Yale Cancer Center; Yale University; New Haven, CT USA
| | | | | | | | | |
Collapse
|