1
|
Zanotta S, Galati D, De Filippi R, Pinto A. Enhancing Dendritic Cell Cancer Vaccination: The Synergy of Immune Checkpoint Inhibitors in Combined Therapies. Int J Mol Sci 2024; 25:7509. [PMID: 39062753 PMCID: PMC11277144 DOI: 10.3390/ijms25147509] [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: 06/11/2024] [Revised: 06/27/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Dendritic cell (DC) cancer vaccines are a promising therapeutic approach, leveraging the immune system to fight tumors. These vaccines utilize DCs' ability to present tumor-associated antigens to T cells, triggering a robust immune response. DC vaccine development has progressed through three generations. The first generation involved priming DCs with tumor-associated antigens or messenger RNA outside the body, showing limited clinical success. The second generation improved efficacy by using cytokine mixtures and specialized DC subsets to enhance immunogenicity. The third generation used blood-derived DCs to elicit a stronger immune response. Clinical trials indicate that cancer vaccines have lower toxicity than traditional cytotoxic treatments. However, achieving significant clinical responses with DC immunotherapy remains challenging. Combining DC vaccines with immune checkpoint inhibitors (ICIs), such as anticytotoxic T-lymphocyte Antigen 4 and antiprogrammed death-1 antibodies, has shown promise by enhancing T-cell responses and improving clinical outcomes. These combinations can transform non-inflamed tumors into inflamed ones, boosting ICIs' efficacy. Current research is exploring new checkpoint targets like LAG-3, TIM-3, and TIGIT, considering their potential with DC vaccines. Additionally, engineering T cells with chimeric antigen receptors or T-cell receptors could further augment the antitumor response. This comprehensive strategy aims to enhance cancer immunotherapy, focusing on increased efficacy and improved patient survival rates.
Collapse
Affiliation(s)
- Serena Zanotta
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (A.P.)
| | - Domenico Galati
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (A.P.)
| | - Rosaria De Filippi
- Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy;
| | - Antonio Pinto
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (A.P.)
| |
Collapse
|
2
|
Galati D, Zanotta S. Dendritic Cell and Cancer Therapy. Int J Mol Sci 2023; 24:ijms24044253. [PMID: 36835665 PMCID: PMC9968100 DOI: 10.3390/ijms24044253] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Dendritic cells (DCs) are acknowledged as the most potent professional antigen-presenting cells (APCs), able to induce adaptive immunity and support the innate immune response [...].
Collapse
|
3
|
Huang F, Liu J, Liu Y. Engineering living cells with cucurbit[7]uril-based supramolecular polymer chemistry: from cell surface engineering to manipulation of subcellular organelles. Chem Sci 2022; 13:8885-8894. [PMID: 35975152 PMCID: PMC9350592 DOI: 10.1039/d2sc02797f] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/04/2022] [Indexed: 12/20/2022] Open
Abstract
Supramolecular polymer chemistry, which closely integrates noncovalent interactions with polymeric structures, is a promising toolbox for living cell engineering. Here, we report our recent progress in exploring the applications of cucurbit[7]uril (CB[7])-based supramolecular polymer chemistry for engineering living cells. First, a modular polymer-analogous approach was established to prepare multifunctional polymers that contain CB[7]-based supramolecular recognition motifs. The supramolecular polymeric systems were successfully applied to cell surface engineering and subcellular organelle manipulation. By anchoring polymers on the cell membranes, cell–cell interactions were established by CB[7]-based host–guest recognition, which further facilitated heterogeneous cell fusion. In addition to cell surface engineering, placing the multifunctional polymers on specific subcellular organelles, including the mitochondria and endoplasmic reticulum, has led to enhanced physical contact between subcellular organelles. It is highly anticipated that the CB[7]-based supramolecular polymer chemistry will provide a new strategy for living cell engineering to advance the development of cell-based therapeutic materials. Cucurbit[7]uril-based supramolecular polymer chemistry, which closely integrates host–guest recognition with multifunctional polymeric structures, is a promising toolbox for living cell engineering.![]()
Collapse
Affiliation(s)
- Fang Huang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Jiaxiong Liu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Yiliu Liu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
4
|
Galati D, Zanotta S, Bocchino M, De Filippi R, Pinto A. The subtle interplay between gamma delta T lymphocytes and dendritic cells: is there a role for a therapeutic cancer vaccine in the era of combinatorial strategies? Cancer Immunol Immunother 2021; 70:1797-1809. [PMID: 33386466 PMCID: PMC10991494 DOI: 10.1007/s00262-020-02805-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/21/2020] [Indexed: 12/12/2022]
Abstract
Human gamma delta (γδ) T cells represent heterogeneous subsets of unconventional lymphocytes with an HLA-unrestricted target cell recognition. γδ T cells display adaptive clonally restricted specificities coupled to a powerful cytotoxic function against transformed/injured cells. Dendritic cells (DCs) are documented to be the most potent professional antigen-presenting cells (APCs) able to induce adaptive immunity and support the innate immune response independently from T cells. Several data show that the cross-talk of γδ T lymphocytes with DCs can play a crucial role in the orchestration of immune response by bridging innate to adaptive immunity. In the last decade, DCs, as well as γδ T cells, have been of increasing clinical interest, especially as monotherapy for cancer immunotherapy, even though with unpredictable results mainly due to immune suppression and/or tumor-immune escape. For these reasons, new vaccine strategies have to be explored to reach cancer immunotherapy's full potential. The effect of DC-based vaccines on γδ T cell is less extensively investigated, and a combinatorial approach using DC-based vaccines with γδ T cells might promote a strong synergy for long-term tumor control and protection against escaping tumor clones. Here, we discuss the therapeutic potential of the interaction between DCs and γδ T cells to improve cancer vaccination. In particular, we describe the most relevant and updated evidence of such combinatorial approaches, including the use of Zoledronate, Interleukin-15, and protamine RNA, also looking towards future strategies such as CAR therapies.
Collapse
Affiliation(s)
- Domenico Galati
- Hematology-Oncology and Stem Cell Transplantation Unit, Department of Hematology and Developmental Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy.
| | - Serena Zanotta
- Hematology-Oncology and Stem Cell Transplantation Unit, Department of Hematology and Developmental Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Marialuisa Bocchino
- Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Rosaria De Filippi
- Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Antonio Pinto
- Hematology-Oncology and Stem Cell Transplantation Unit, Department of Hematology and Developmental Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
5
|
Zhang XW, Huck K, Jähne K, Cichon F, Sonner JK, Ufer F, Bauer S, Woo MS, Green E, Lu K, Kilian M, Friese MA, Platten M, Sahm K. Activity-regulated cytoskeleton-associated protein/activity-regulated gene 3.1 (Arc/Arg3.1) enhances dendritic cell vaccination in experimental melanoma. Oncoimmunology 2021; 10:1920739. [PMID: 34026332 PMCID: PMC8128181 DOI: 10.1080/2162402x.2021.1920739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Dendritic cell (DC) vaccination has proven to be an effective and safe adjuvant for cancer immunotherapies. As the presence of DCs within the tumor microenvironment promotes adaptive antitumor immunity, enhancement of DC migration toward the tumor microenvironment following DC vaccination might represent one possible approach to increase its therapeutic efficacy. While recent findings suggest the activity-regulated cytoskeleton-associated protein/activity-regulated gene 3.1 (Arc/Arg3.1) as critical regulator of DC migration in the context of autoimmune diseases, we aimed to investigate the impact of Arc/Arg3.1 expression for DC-based cancer vaccines. To this end, DC migration capacity as well as the induction of T cell-mediated antitumor immunity was assessed in an experimental B16 melanoma model with Arc/Arg3.1−/- and Arc/Arg3.1-expressing BMDCs applied as a subcutaneous vaccine. While antigen presentation on DCs was critical for unleashing effective T cell mediated antitumor immune responses, Arc/Arg3.1 expression enhanced DC migration toward the tumor and secondary lymphoid organs. Moreover, Arc/Arg3.1-expressing BMDCs shape the tumor immune microenvironment by facilitating tumor recruitment of antigen-specific effector T cells. Thus, Arc/Arg3.1 may represent a novel therapeutic target in DCs in order to increase the therapeutic efficacy of DC vaccination.
Collapse
Affiliation(s)
- Xin-Wen Zhang
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Katrin Huck
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Kristine Jähne
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Frederik Cichon
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Jana K Sonner
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Friederike Ufer
- Institute of Neuroimmunology Und Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Bauer
- Institute of Neuroimmunology Und Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcel Seungsu Woo
- Institute of Neuroimmunology Und Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ed Green
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Kevin Lu
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Michael Kilian
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology Und Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Platten
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Katharina Sahm
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
6
|
Bocchino M, Zanotta S, Capitelli L, Galati D. Dendritic Cells Are the Intriguing Players in the Puzzle of Idiopathic Pulmonary Fibrosis Pathogenesis. Front Immunol 2021; 12:664109. [PMID: 33995394 PMCID: PMC8121252 DOI: 10.3389/fimmu.2021.664109] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most devastating progressive interstitial lung disease that remains refractory to treatment. Pathogenesis of IPF relies on the aberrant cross-talk between injured alveolar cells and myofibroblasts, which ultimately leads to an aberrant fibrous reaction. The contribution of the immune system to IPF remains not fully explored. Recent evidence suggests that both innate and adaptive immune responses may participate in the fibrotic process. Dendritic cells (DCs) are the most potent professional antigen-presenting cells that bridge innate and adaptive immunity. Also, they exert a crucial role in the immune surveillance of the lung, where they are strategically placed in the airway epithelium and interstitium. Immature DCs accumulate in the IPF lung close to areas of epithelial hyperplasia and fibrosis. Conversely, mature DCs are concentrated in well-organized lymphoid follicles along with T and B cells and bronchoalveolar lavage of IPF patients. We have recently shown that all sub-types of peripheral blood DCs (including conventional and plasmacytoid DCs) are severely depleted in therapy naïve IPF patients. Also, the low frequency of conventional CD1c+ DCs is predictive of a worse prognosis. The purpose of this mini-review is to focus on the main evidence on DC involvement in IPF pathogenesis. Unanswered questions and opportunities for future research ranging from a better understanding of their contribution to diagnosis and prognosis to personalized DC-based therapies will be explored.
Collapse
Affiliation(s)
- Marialuisa Bocchino
- Respiratory Medicine Division, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Serena Zanotta
- Hematology-Oncology and Stem Cell Transplantation Unit, Department of Hematology and Developmental Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Ludovica Capitelli
- Respiratory Medicine Division, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Domenico Galati
- Hematology-Oncology and Stem Cell Transplantation Unit, Department of Hematology and Developmental Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
7
|
Wang W, Bai L, Xu D, Li W, Cui J. Immunotherapy: A Potential Approach to Targeting Cancer Stem Cells. Curr Cancer Drug Targets 2021; 21:117-131. [PMID: 32364076 DOI: 10.2174/1568009620666200504111914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/14/2020] [Accepted: 04/04/2020] [Indexed: 12/24/2022]
Abstract
Tumor recurrence and drug resistance are two of the key factors affecting the prognosis of cancer patients. Cancer stem cells (CSCs) are a group of cells with infinite proliferation potential which are not sensitive to traditional therapies, including radio- and chemotherapy. These CSCs are considered to be central to tumor recurrence and the development of drug resistance. In addition, CSCs are important targets in cancer immunotherapy because of their expression of novel tumorassociated antigens, which result from mutations in cancer cells over the course of treatment. Emerging immunotherapies, including cancer vaccines, checkpoint blockade therapies, and transferred immune cell therapies, have all been shown to be more effective when they selectively target CSCs. Such therapies may also provide novel additions to the current therapeutic milieu and may offer new therapeutic combinations for treatment. This review summarizes the relationships between various immunotherapies and CSCs and provides novel insights into potential therapeutic applications for these approaches in the future.
Collapse
Affiliation(s)
- Wenjun Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Ling Bai
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Dongsheng Xu
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| |
Collapse
|
8
|
Relitti N, Saraswati AP, Federico S, Khan T, Brindisi M, Zisterer D, Brogi S, Gemma S, Butini S, Campiani G. Telomerase-based Cancer Therapeutics: A Review on their Clinical Trials. Curr Top Med Chem 2020; 20:433-457. [PMID: 31894749 DOI: 10.2174/1568026620666200102104930] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/11/2022]
Abstract
Telomeres are protective chromosomal ends that shield the chromosomes from DNA damage, exonucleolytic degradation, recombination, and end-to-end fusion. Telomerase is a ribonucleoprotein that adds TTAGGG tandem repeats to the telomeric ends. It has been observed that 85 to 90% of human tumors express high levels of telomerase, playing a crucial role in the development of cancers. Interestingly, the telomerase activity is generally absent in normal somatic cells. This selective telomerase expression has driven scientists to develop novel anti-cancer therapeutics with high specificity and potency. Several advancements have been made in this area, which is reflected by the enormous success of the anticancer agent Imetelstat. Since the discovery of Imetelstat, several research groups have contributed to enrich the therapeutic arsenal against cancer. Such contributions include the application of new classes of small molecules, peptides, and hTERT-based immunotherapeutic agents (p540, GV1001, GRNVAC1 or combinations of these such as Vx-001). Many of these therapeutic tools are under different stages of clinical trials and have shown promising outcomes. In this review, we highlight the current status of telomerase-based cancer therapeutics and the outcome of these investigations.
Collapse
Affiliation(s)
- Nicola Relitti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Akella P Saraswati
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Stefano Federico
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Tuhina Khan
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Margherita Brindisi
- Department of Pharmacy, Department of Excellence 2018-2022, University of Napoli Federico II, via D. Montesano 49, I-80131 Napoli, Italy
| | - Daniela Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160, Pearse Street, Dublin 2, Ireland
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, I-56126 Pisa, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| |
Collapse
|
9
|
Zhai Y, Lu Q, Lou T, Cao G, Wang S, Zhang Z. MUC16 affects the biological functions of ovarian cancer cells and induces an antitumor immune response by activating dendritic cells. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1494. [PMID: 33313239 PMCID: PMC7729312 DOI: 10.21037/atm-20-6388] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Ovarian cancer is the 5th most common lethal gynecological malignancy with a 5-year survival rate of about 47% and a localized stage diagnosis of 15%, leading to about 125,000 global deaths each year. Therefore, it is urgent to explore novel and effective strategies for radical cure. Methods Short hairpin RNA targeting the Mucin16 (MUC16) gene was used to establish MUC16 knockdown in ovarian cancer cells. RT-PCR was performed to quantify the expression of MUC16 mRNA, and western blotting was performed to detect the expression of MUC16 and epithelial-mesenchymal transition-related proteins. Cell counting kit 8 (CCK8) wound healing and transwell assays were performed to assess cell proliferation and cell invasion. Flow cytometry was used to detect CD80-, CD83-, and CD86-expressing dendritic cells (DCs) and cytotoxic T lymphocytes (CTLs) activated by MUC16-pulsed DCs. Results In this study, we identified MUC16 as a novel target antigen for immunotherapy against ovarian cancer, which was significantly up regulated in ovarian cancer cells and high-grade ovarian serous adenocarcinoma tissues. MUC16 knockdown in Ovcar3 cells using short hairpin RNA targeting the MUC16 gene suppressed the proliferation of migration, invasion, epithelial-mesenchymal transition (EMT), and PI3K/Akt signaling pathway in Ovcar3 cells markedly. MUC16 significantly up-regulated CD80, CD83, and CD86 (mature makers) expression in DCs and T-cell transformation into CD8+ T-cells detected by Flow cytometry. Conclusions For malignant ovarian cancer, MUC16 overexpression promoted cell proliferation, migration, and invasion via the PI3K/AKT signaling pathway. MUC16 pulsing mediated DC maturation and activated CTL response in vitro. Our study offers promising DC-based immunotherapy of considerable clinical value for patients with ovarian cancer.
Collapse
Affiliation(s)
- Yan Zhai
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qi Lu
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Tong Lou
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guangming Cao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shuzhen Wang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Bastien JP, Fekete N, Beland AV, Lachambre MP, Laforte V, Juncker D, Dave V, Roy DC, Hoesli CA. Closing the system: production of viral antigen-presenting dendritic cells eliciting specific CD8 + T cell activation in fluorinated ethylene propylene cell culture bags. J Transl Med 2020; 18:383. [PMID: 33036618 PMCID: PMC7547414 DOI: 10.1186/s12967-020-02543-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Background A major obstacle to anti-viral and -tumor cell vaccination and T cell immunotherapy is the ability to produce dendritic cells (DCs) in a suitable clinical setting. It is imperative to develop closed cell culture systems to accelerate the translation of promising DC-based cell therapy products to the clinic. The objective of this study was to investigate whether viral antigen-loaded monocyte-derived DCs (Mo-DCs) capable of eliciting specific T cell activation can be manufactured in fluorinated ethylene propylene (FEP) bags. Methods Mo-DCs were generated through a protocol applying cytokine cocktails combined with lipopolysaccharide or with a CMV viral peptide antigen in conventional tissue culture polystyrene (TCPS) or FEP culture vessels. Research-scale (< 10 mL) FEP bags were implemented to increase R&D throughput. DC surface marker profiles, cytokine production, and ability to activate antigen-specific cytotoxic T cells were characterized. Results Monocyte differentiation into Mo-DCs led to the loss of CD14 expression with concomitant upregulation of CD80, CD83 and CD86. Significantly increased levels of IL-10 and IL-12 were observed after maturation on day 9. Antigen-pulsed Mo-DCs activated antigen-responsive CD8+ cytotoxic T cells. No significant differences in surface marker expression or tetramer-specific T cell activating potency of Mo-DCs were observed between TCPS and FEP culture vessels. Conclusions Our findings demonstrate that viral antigen-loaded Mo-DCs produced in downscaled FEP bags can elicit specific T cell responses. In view of the dire clinical need for closed system DC manufacturing, FEP bags represent an attractive option to accelerate the translation of promising emerging DC-based immunotherapies.
Collapse
Affiliation(s)
- Jean-Philippe Bastien
- Hematology-Oncology and Cell Therapy Institute, Hopital Maisonneuve-Rosemont Research Center, Montreal, Québec, Canada
| | - Natalie Fekete
- Department of Chemical Engineering, McGill University, Montreal, Québec, Canada
| | - Ariane V Beland
- Department of Chemical Engineering, McGill University, Montreal, Québec, Canada
| | - Marie-Paule Lachambre
- Hematology-Oncology and Cell Therapy Institute, Hopital Maisonneuve-Rosemont Research Center, Montreal, Québec, Canada
| | - Veronique Laforte
- Department of Biomedical Engineering, McGill University, Montreal, Québec, Canada.,McGill Genome Centre, McGill University, Montreal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada
| | - David Juncker
- Department of Biomedical Engineering, McGill University, Montreal, Québec, Canada.,McGill Genome Centre, McGill University, Montreal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada
| | - Vibhuti Dave
- Hematology-Oncology and Cell Therapy Institute, Hopital Maisonneuve-Rosemont Research Center, Montreal, Québec, Canada.,Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Québec, Canada
| | - Denis-Claude Roy
- Hematology-Oncology and Cell Therapy Institute, Hopital Maisonneuve-Rosemont Research Center, Montreal, Québec, Canada.,Department of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Corinne A Hoesli
- Department of Chemical Engineering, McGill University, Montreal, Québec, Canada. .,Department of Biomedical Engineering, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
11
|
Qi FL, Wang MF, Li BZ, Lu ZF, Nie GJ, Li SP. Reversal of the immunosuppressive tumor microenvironment by nanoparticle-based activation of immune-associated cells. Acta Pharmacol Sin 2020; 41:895-901. [PMID: 32467568 PMCID: PMC7470798 DOI: 10.1038/s41401-020-0423-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy that activates the host immune system to reverse immunosuppression has emerged as a new generation of cancer treatment in both preclinical studies and clinical trials. Although immunotherapy has shown significant achievements in the treatment of various cancers, it faces challenges that limit its further evolution such as poor permeation and modest responsiveness. The development of nanoparticle drug delivery system has provided an opportunity to overcome these drawbacks and to achieve optimized immunotherapy. Based on the research of our group, we here introduce the new strategies being employed using nanoscale intelligent drug delivery systems to enhance the effects of cancer immunotherapy. We also provide a perspective on the further possible application of nanoparticles in more effective antitumor immunotherapy.
Collapse
Affiliation(s)
- Fei-Long Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mei-Fang Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Bo-Zhao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Ze-Fang Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guang-Jun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Su-Ping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
12
|
Trybek T, Kowalik A, Góźdź S, Kowalska A. Telomeres and telomerase in oncogenesis. Oncol Lett 2020; 20:1015-1027. [PMID: 32724340 PMCID: PMC7377093 DOI: 10.3892/ol.2020.11659] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/25/2020] [Indexed: 12/24/2022] Open
Abstract
Telomeres are located at the ends of chromosomes and protect them from degradation. Suppressing the activity of telomerase, a telomere-synthesizing enzyme, and maintaining short telomeres is a protective mechanism against cancer in humans. In most human somatic cells, the expression of telomerase reverse transcriptase (TERT) is repressed and telomerase activity is inhibited. This leads to the progressive shortening of telomeres and inhibition of cell growth in a process called replicative senescence. Most types of primary cancer exhibit telomerase activation, which allows uncontrolled cell proliferation. Previous research indicates that TERT activation also affects cancer development through activities other than the canonical function of mediating telomere elongation. Recent studies have improved the understanding of the structure and function of telomeres and telomerase as well as key mechanisms underlying the activation of TERT and its role in oncogenesis. These advances led to a search for drugs that inhibit telomerase as a target for cancer therapy. The present review article summarizes the organization and function of telomeres, their role in carcinogenesis, and advances in telomerase-targeted therapy.
Collapse
Affiliation(s)
- Tomasz Trybek
- Endocrinology Clinic, Holycross Cancer Center, 25-734 Kielce, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holycross Cancer Center, 25-734 Kielce, Poland
| | - Stanisław Góźdź
- The Faculty of Health Sciences, Jan Kochanowski University, 25-319 Kielce, Poland.,Oncology Clinic, Holycross Cancer Center, 25-734 Kielce, Poland
| | - Aldona Kowalska
- Endocrinology Clinic, Holycross Cancer Center, 25-734 Kielce, Poland.,The Faculty of Health Sciences, Jan Kochanowski University, 25-319 Kielce, Poland
| |
Collapse
|
13
|
Blood Eosinophilia is an on-Treatment Biomarker in Patients with Solid Tumors Undergoing Dendritic Cell Vaccination with Autologous Tumor-RNA. Pharmaceutics 2020; 12:pharmaceutics12030210. [PMID: 32121531 PMCID: PMC7150785 DOI: 10.3390/pharmaceutics12030210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 12/20/2022] Open
Abstract
Background: The approvals of immune checkpoint inhibitors for several cancer types and the rapidly growing recognition that T cell-based immunotherapy significantly improves outcomes for cancer patients led to a re-emergence of cancer vaccines, including dendritic cell (DC)-based immunotherapy. Blood and tissue biomarkers to identify responders and long-term survivors and to optimize cost and cost-effectiveness of treatment are greatly needed. We wanted to investigate whether blood eosinophilia is a predictive biomarker for patients with solid tumors receiving vaccinations with DCs loaded with autologous tumor-RNA. Methods: In total, 67 patients with metastatic solid tumors, who we treated with autologous monocyte-derived DCs transfected with total tumor mRNA, were serially analyzed for eosinophil counts and survival over the course of up to 14 years. Eosinophilic counts were performed on peripheral blood smears. Results: Up to 87% of the patients treated with DC-based immunotherapy experienced at least once an eosinophilia of ≥ 5% after initiation of therapy; 61 % reached levels of ≥ 10% eosinophils, and 13% of patients showed eosinophil counts of 20% or above. While prevaccination eosinophil levels were not associated with survival, patients with blood eosinophilia at any point after initiation of DC-based immunotherapy showed a trend towards longer survival. There was a statistically significant difference for the patients with eosinophil counts of 20% or more (p = 0.03). In those patients, survival was prolonged to a median of 58 months (range 2–111 months), compared to a median of 20 months (range 0–119 months) in patients with lower eosinophil counts. In 12% of the patients, an immediate increase in eosinophil count of at least 10 percentage points could be detected after the first vaccine, which also appeared to correlate with survival (65 vs. 24 months; p = 0.06). Conclusion: Blood eosinophilia appears to be an early, on-therapy biomarker in patients with solid tumors undergoing vaccination with RNA-transfected DC, specifically autologous tumor mRNA-transfected DC vaccines, and it correlates with long-term patient outcome. Eosinophilia should be systematically investigated in future trials.
Collapse
|
14
|
Telomerase-Targeted Cancer Immunotherapy. Int J Mol Sci 2019; 20:ijms20081823. [PMID: 31013796 PMCID: PMC6515163 DOI: 10.3390/ijms20081823] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/05/2019] [Accepted: 04/10/2019] [Indexed: 01/03/2023] Open
Abstract
Telomerase, an enzyme responsible for the synthesis of telomeres, is activated in many cancer cells and is involved in the maintenance of telomeres. The activity of telomerase allows cancer cells to replicate and proliferate in an uncontrolled manner, to infiltrate tissue, and to metastasize to distant organs. Studies to date have examined the mechanisms involved in the survival of cancer cells as targets for cancer therapeutics. These efforts led to the development of telomerase inhibitors as anticancer drugs, drugs targeting telomere DNA, viral vectors carrying a promoter for human telomerase reverse transcriptase (hTERT) genome, and immunotherapy targeting hTERT. Among these novel therapeutics, this review focuses on immunotherapy targeting hTERT and discusses the current evidence and future perspectives.
Collapse
|
15
|
Radiotherapy for Melanoma: More than DNA Damage. Dermatol Res Pract 2019; 2019:9435389. [PMID: 31073304 PMCID: PMC6470446 DOI: 10.1155/2019/9435389] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/26/2018] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
Despite its reputation as a radioresistant tumour, there is evidence to support a role for radiotherapy in patients with melanoma and we summarise current clinical practice. Melanoma is a highly immunogenic tumour and in this era of immunotherapy, there is renewed interest in the potential of irradiation, not only as an adjuvant and palliative treatment, but also as an immune stimulant. It has long been known that radiation causes not only DNA strand breaks, apoptosis, and necrosis, but also immunogenic modulation and cell death through the induction of dendritic cells, cell adhesion molecules, death receptors, and tumour-associated antigens, effectively transforming the tumour into an individualised vaccine. This immune response can be enhanced by the application of clinical hyperthermia as evidenced by randomised trial data in patients with melanoma. The large fraction sizes used in cranial radiosurgery and stereotactic body radiotherapy are more immunogenic than conventional fractionation, which provides additional radiobiological justification for these techniques in this disease entity. Given the immune priming effect of radiotherapy, there is a strong but complex biological rationale and an increasing body of evidence for synergy in combination with immune checkpoint inhibitors, which are now first-line therapy in patients with recurrent or metastatic melanoma. There is great potential to increase local control and abscopal effects by combining radiotherapy with both immunotherapy and hyperthermia, and a combination of all three modalities is suggested as the next important trial in this refractory disease.
Collapse
|
16
|
Maletzki C, Wiegele L, Nassar I, Stenzel J, Junghanss C. Chemo-immunotherapy improves long-term survival in a preclinical model of MMR-D-related cancer. J Immunother Cancer 2019; 7:8. [PMID: 30630527 PMCID: PMC6329128 DOI: 10.1186/s40425-018-0476-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/06/2018] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Mismatch Repair Deficiency (MMR-D)-related tumors are highly immunogenic and constitute ideal vaccination targets. In a proof-of-concept study delayed tumorigenesis and prolonged survival has been shown in a clinically-relevant mouse model for MMR-D-related diseases (=MLH1 knock out mice). To refine this approach, vaccination was combined with immune modulatory low-dose chemotherapy to polarize immune regulatory subtypes. METHODS Mice (prophylactic: 8-10 weeks; therapeutic: > 36 weeks) received a single injection of cyclophosphamide (CPX, 120 mg/kg bw, i.p.) or gemcitabine (GEM, 100 mg/kg bw, i.p.) prior to vaccination (lysate of a gastrointestinal tumor allograft, 10 mg/kg bw, n = 9 mice/group). The vaccine was given repetitively (10 mg/kg bw, s.c., 4 x / once a week, followed by monthly boosts) until tumor formation or progression. Tumor growth ([18F] FDG PET/CT imaging) and immune responses were monitored (flow cytometry, IFNγ ELISpot). The microenvironment was analyzed by immunofluorescence. RESULTS Prophylactic application of GEM + lysate delayed tumorigenesis compared to lysate monotherapy and CPX-pre-treatment (median time of onset: 53 vs. 47 vs. 48 weeks). 33% of mice even remained tumor-free until the experimental endpoint (= 65 weeks). This was accompanied by long-term effect on cytokine plasma levels; splenic myeloid derived suppressor cells (MDSC) as well as regulatory T cell numbers. Assessment of tumor microenvironment from GEM + lysate treated mice revealed low numbers of MDSCs, but enhanced T cell infiltration, in some cases co-expressing PD-L1. Therapeutic chemo-immunotherapy (GEM + lysate) had minor impact on overall survival (median time: 12 (GEM + lysate) vs. 11.5 (lysate) vs. 3 weeks (control)), but induced complete remission in one case. Dendritic and T cell infiltrates increased in both treatment groups. Reactive T cells specifically recognized MLH1-/- tumor cells in IFNγ ELISpot, but lacked response towards NK cell targets YAC-1. CONCLUSIONS Combined chemo-immunotherapy impairs tumor onset and growth likely attributable to modulation of immune responses. Depleting or 're-educating' immunosuppressive cell types, such as MDSC, may help moving a step closer to combat cancer.
Collapse
Affiliation(s)
- Claudia Maletzki
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.
| | - Leonie Wiegele
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Ingy Nassar
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Jan Stenzel
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Schillingallee 69a, 18057, Rostock, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| |
Collapse
|