1
|
Fan R, Satilmis H, Vandewalle N, Verheye E, Vlummens P, Maes A, Muylaert C, De Bruyne E, Menu E, Evans H, Chantry A, De Beule N, Hose D, Törngren M, Eriksson H, Vanderkerken K, Maes K, Breckpot K, De Veirman K. Tasquinimod suppresses tumor cell growth and bone resorption by targeting immunosuppressive myeloid cells and inhibiting c-MYC expression in multiple myeloma. J Immunother Cancer 2023; 11:jitc-2022-005319. [PMID: 36650020 PMCID: PMC9853259 DOI: 10.1136/jitc-2022-005319] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Immunotherapy emerged as a promising treatment option for multiple myeloma (MM) patients. However, therapeutic efficacy can be hampered by the presence of an immunosuppressive bone marrow microenvironment including myeloid cells. S100A9 was previously identified as a key regulator of myeloid cell accumulation and suppressive activity. Tasquinimod, a small molecule inhibitor of S100A9, is currently in a phase Ib/IIa clinical trial in MM patients (NCT04405167). We aimed to gain more insights into its mechanisms of action both on the myeloma cells and the immune microenvironment. METHODS We analyzed the effects of tasquinimod on MM cell viability, cell proliferation and downstream signaling pathways in vitro using RNA sequencing, real-time PCR, western blot analysis and multiparameter flow cytometry. Myeloid cells and T cells were cocultured at different ratios to assess tasquinimod-mediated immunomodulatory effects. The in vivo impact on immune cells (myeloid cell subsets, macrophages, dendritic cells), tumor load, survival and bone disease were elucidated using immunocompetent 5TMM models. RESULTS Tasquinimod treatment significantly decreased myeloma cell proliferation and colony formation in vitro, associated with an inhibition of c-MYC and increased p27 expression. Tasquinimod-mediated targeting of the myeloid cell population resulted in increased T cell proliferation and functionality in vitro. Notably, short-term tasquinimod therapy of 5TMM mice significantly increased the total CD11b+ cells and shifted this population toward a more immunostimulatory state, which resulted in less myeloid-mediated immunosuppression and increased T cell activation ex vivo. Tasquinimod significantly reduced the tumor load and increased the trabecular bone volume, which resulted in prolonged overall survival of MM-bearing mice in vivo. CONCLUSION Our study provides novel insights in the dual therapeutic effects of the immunomodulator tasquinimod and fosters its evaluation in combination therapy trials for MM patients.
Collapse
Affiliation(s)
- Rong Fan
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hatice Satilmis
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Niels Vandewalle
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Emma Verheye
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium,Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium,Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
| | - Philip Vlummens
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium,Department of Clinical Hematology, Universitair Ziekenhuis Gent, Ghent, Belgium
| | - Anke Maes
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Catharina Muylaert
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Holly Evans
- Department of Oncology and Metabolism, Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
| | - Andrew Chantry
- Department of Oncology and Metabolism, Sheffield Myeloma Research Team, University of Sheffield, Sheffield, UK
| | - Nathan De Beule
- Department of Clinical Hematology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Dirk Hose
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | - Karin Vanderkerken
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ken Maes
- Center for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
2
|
Gong P, Liu H, Liu X, Zhou G, Liu M, Yang X, Xiong W, Wang Q, Ma J, Ren Z, He M, Zhang X. Efficacy of tasquinimod in men with metastatic castration-resistant prostate cancer: A meta-analysis of randomized controlled trials. Medicine (Baltimore) 2018; 97:e13204. [PMID: 30431595 PMCID: PMC6257339 DOI: 10.1097/md.0000000000013204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/15/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Tasquinimod is an oral quinoline-3-carboxamide derivative for the treatment of metastatic castration-resistant prostate cancer (mCRPC). Tasquinimod has antiangiogenic, immunomodulatory, and antimetastatic properties, but it is also associated with toxicities, including skeletal pain, digestive disorders, fatigue, insomnia, and mental disorders. We aimed to perform a meta-analysis to evaluate the efficacy, safety, and long-term survival for tasquinimod in patients with mCRPC. METHODS Searches were carried out in PubMed, Embase, and the Cochrane Library. Eligible articles included randomized clinical trials (RCTs) comparing systemic or combination therapy (excluding primary or secondary androgen deprivation therapy, bone protective agents, or radionuclides) with placebo in men with mCRPC. RESULTS Three RCTs were selected for final evaluation. The pooled results from the 3 studies indicated that tasquinimod was associated with good radiologic progression-free survival (rPFS) in mCRPC. For adverse effects (AEs), the results of meta-analysis indicated that patients with mCRPC who received tasquinimod had obvious anemia (risk ratio (RR) 1.35, 95% confidence interval (CI) 1.06-1.73, P = .02), back pain (RR: 1.57, 95% CI: 1.01-2.47, P = .05), pain in the extremities (RR: 1.90, 95% CI: 1.14-3.17, P = .01), insomnia (RR: 1.50, 95% CI: 1.03-2.17, P = .03), vomiting (RR: 1.52, 95% CI: 1.04-2.21, P = .03), and peripheral edema (RR: 1.52, 95% CI: 1.03-2.23, P = .03). CONCLUSIONS Tasquinimod is associated with better rPFS in mCRPC. The toxicity of tasquinimod requires further investigation, it is not recommended for routine clinical use.
Collapse
Affiliation(s)
- Ping Gong
- Department of Epidemiology and Biostatistics
| | | | - Xinyu Liu
- Department of Epidemiology and Biostatistics
| | - Ge Zhou
- Department of Epidemiology and Biostatistics
| | - Meitian Liu
- Department of Epidemiology and Biostatistics
| | - Xiaodi Yang
- Department of Epidemiology and Biostatistics
| | | | - Qi Wang
- Department of Epidemiology and Biostatistics
| | - Juan Ma
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Zheng Ren
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Minfu He
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Xiumin Zhang
- Department of Social Medicine and Health Management, School of Public Health, Jilin University, Changchun, Jilin, China
| |
Collapse
|
3
|
Ubiquitous Nature of Fluoroquinolones: The Oscillation between Antibacterial and Anticancer Activities. Antibiotics (Basel) 2017; 6:antibiotics6040026. [PMID: 29112154 PMCID: PMC5745469 DOI: 10.3390/antibiotics6040026] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 10/26/2017] [Accepted: 11/03/2017] [Indexed: 12/15/2022] Open
Abstract
Fluoroquinolones are synthetic antibacterial agents that stabilize the ternary complex of prokaryotic topoisomerase II enzymes (gyrase and Topo IV), leading to extensive DNA fragmentation and bacteria death. Despite the similar structural folds within the critical regions of prokaryotic and eukaryotic topoisomerases, clinically relevant fluoroquinolones display a remarkable selectivity for prokaryotic topoisomerase II, with excellent safety records in humans. Typical agents that target human topoisomerases (such as etoposide, doxorubicin and mitoxantrone) are associated with significant toxicities and secondary malignancies, whereas clinically relevant fluoroquinolones are not known to exhibit such propensities. Although many fluoroquinolones have been shown to display topoisomerase-independent antiproliferative effects against various human cancer cells, those that are significantly active against eukaryotic topoisomerase show the same DNA damaging properties as other topoisomerase poisons. Empirical models also show that fluoroquinolones mediate some unique immunomodulatory activities of suppressing pro-inflammatory cytokines and super-inducing interleukin-2. This article reviews the extended roles of fluoroquinolones and their prospects as lead for the unmet needs of "small and safe" multimodal-targeting drug scaffolds.
Collapse
|
4
|
|
5
|
Wen YC, Lee WJ, Tan P, Yang SF, Hsiao M, Lee LM, Chien MH. By inhibiting snail signaling and miR-23a-3p, osthole suppresses the EMT-mediated metastatic ability in prostate cancer. Oncotarget 2016; 6:21120-36. [PMID: 26110567 PMCID: PMC4673254 DOI: 10.18632/oncotarget.4229] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/13/2015] [Indexed: 02/07/2023] Open
Abstract
Here we showed that Osthole, 7-methoxy-8-(3-methyl-2-butenyl) coumarin, a bioactive coumarin derivative extracted from medicinal plants, inhibited migration, invasion, epithelial to mesenchymal transition (EMT) in androgen-independent prostate cancer (AIPC) cells in vitro and metastasis of AIPC in vivo. In patients, high Snail levels were correlated with a higher histological Gleason sum and poor survival rates. Osthole inhibited the TGF-β/Akt/MAPK pathways, reduced Snail-DNA-binding activity and induced E-cadherin. We found that osthole decreased miR-23a-3p. Ectopic miR-23a-3p suppressed E-cadherin 3′ untranslated region reporter activity and E-cadherin expression, and relieved the motility suppression caused by osthole treatment.
Collapse
Affiliation(s)
- Yu-Ching Wen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wei-Jiunn Lee
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Peng Tan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Michael Hsiao
- The Genomics Research Center, Academia Sinica; Taipei, Taiwan
| | - Liang-Ming Lee
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
6
|
Wang G, Lu X, Dey P, Deng P, Wu CC, Jiang S, Fang Z, Zhao K, Konaparthi R, Hua S, Zhang J, Li-Ning-Tapia EM, Kapoor A, Wu CJ, Patel NB, Guo Z, Ramamoorthy V, Tieu TN, Heffernan T, Zhao D, Shang X, Khadka S, Hou P, Hu B, Jin EJ, Yao W, Pan X, Ding Z, Shi Y, Li L, Chang Q, Troncoso P, Logothetis CJ, McArthur MJ, Chin L, Wang YA, DePinho RA. Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression. Cancer Discov 2015; 6:80-95. [PMID: 26701088 DOI: 10.1158/2159-8290.cd-15-0224] [Citation(s) in RCA: 386] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 10/16/2015] [Indexed: 12/20/2022]
Abstract
UNLABELLED The signaling mechanisms between prostate cancer cells and infiltrating immune cells may illuminate novel therapeutic approaches. Here, utilizing a prostate adenocarcinoma model driven by loss of Pten and Smad4, we identify polymorphonuclear myeloid-derived suppressor cells (MDSC) as the major infiltrating immune cell type, and depletion of MDSCs blocks progression. Employing a novel dual reporter prostate cancer model, epithelial and stromal transcriptomic profiling identified CXCL5 as a cancer-secreted chemokine to attract CXCR2-expressing MDSCs, and, correspondingly, pharmacologic inhibition of CXCR2 impeded tumor progression. Integrated analyses identified hyperactivated Hippo-YAP signaling in driving CXCL5 upregulation in cancer cells through the YAP-TEAD complex and promoting MDSC recruitment. Clinicopathologic studies reveal upregulation and activation of YAP1 in a subset of human prostate tumors, and the YAP1 signature is enriched in primary prostate tumor samples with stronger expression of MDSC-relevant genes. Together, YAP-driven MDSC recruitment via heterotypic CXCL5-CXCR2 signaling reveals an effective therapeutic strategy for advanced prostate cancer. SIGNIFICANCE We demonstrate a critical role of MDSCs in prostate tumor progression and discover a cancer cell nonautonomous function of the Hippo-YAP pathway in regulation of CXCL5, a ligand for CXCR2-expressing MDSCs. Pharmacologic elimination of MDSCs or blocking the heterotypic CXCL5-CXCR2 signaling circuit elicits robust antitumor responses and prolongs survival.
Collapse
Affiliation(s)
- Guocan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xin Lu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Prasenjit Dey
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pingna Deng
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chia Chin Wu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shan Jiang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhuangna Fang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Kun Zhao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ramakrishna Konaparthi
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sujun Hua
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianhua Zhang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elsa M Li-Ning-Tapia
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Avnish Kapoor
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chang-Jiun Wu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neelay Bhaskar Patel
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhenglin Guo
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vandhana Ramamoorthy
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Trang N Tieu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tim Heffernan
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Di Zhao
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaoying Shang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunada Khadka
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pingping Hou
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Baoli Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eun-Jung Jin
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Biological Science, College of Natural Sciences, Wonkwang University, Cheonbuk, Iksan, South Korea
| | - Wantong Yao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaolu Pan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhihu Ding
- Sanofi Oncology, Cambridge, Massachusetts
| | - Yanxia Shi
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Liren Li
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Qing Chang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark J McArthur
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lynda Chin
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Y Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
7
|
Frieling JS, Basanta D, Lynch CC. Current and emerging therapies for bone metastatic castration-resistant prostate cancer. Cancer Control 2015; 22:109-20. [PMID: 25504285 PMCID: PMC4673894 DOI: 10.1177/107327481502200114] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A paucity of therapeutic options is available to treat men with metastatic castration-resistant prostate cancer (mCRPC). However, recent developments in our understanding of the disease have resulted in several new therapies that show promise in improving overall survival rates in this patient population. METHODS Agents approved for use in the United States and those undergoing clinical trials for the treatment of mCRPC are reviewed. Recent contributions to the understanding of prostate biology and bone metastasis are discussed as well as how the underlying mechanisms may represent opportunities for therapeutic intervention. New challenges to delivering effective mCRPC treatment will also be examined. RESULTS New and emerging treatments that target androgen synthesis and utilization or the microenvironment may improve overall survival rates for men diagnosed with mCRPC. Determining how factors derived from the primary tumor can promote the development of premetastatic niches and how prostate cancer cells parasitize niches in the bone microenvironment, thus remaining dormant and protected from systemic therapy, could yield new therapies to treat mCRPC. Challenges such as intratumoral heterogeneity and patient selection can potentially be circumvented via computational biology approaches. CONCLUSIONS The emergence of novel treatments for mCRPC, combined with improved patient stratification and optimized therapy sequencing, suggests that significant gains may be made in terms of overall survival rates for men diagnosed with this form of cancer.
Collapse
Affiliation(s)
- Jeremy S Frieling
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
8
|
Bloy N, Pol J, Aranda F, Eggermont A, Cremer I, Fridman WH, Fučíková J, Galon J, Tartour E, Spisek R, Dhodapkar MV, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based anticancer therapy. Oncoimmunology 2014; 3:e963424. [PMID: 25941593 DOI: 10.4161/21624011.2014.963424] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 09/05/2014] [Indexed: 02/06/2023] Open
Abstract
The use of patient-derived dendritic cells (DCs) as a means to elicit therapeutically relevant immune responses in cancer patients has been extensively investigated throughout the past decade. In this context, DCs are generally expanded, exposed to autologous tumor cell lysates or loaded with specific tumor-associated antigens (TAAs), and then reintroduced into patients, often in combination with one or more immunostimulatory agents. As an alternative, TAAs are targeted to DCs in vivo by means of monoclonal antibodies, carbohydrate moieties or viral vectors specific for DC receptors. All these approaches have been shown to (re)activate tumor-specific immune responses in mice, often mediating robust therapeutic effects. In 2010, the first DC-based preparation (sipuleucel-T, also known as Provenge®) has been approved by the US Food and Drug Administration (FDA) for use in humans. Reflecting the central position occupied by DCs in the regulation of immunological tolerance and adaptive immunity, the interest in harnessing them for the development of novel immunotherapeutic anticancer regimens remains high. Here, we summarize recent advances in the preclinical and clinical development of DC-based anticancer therapeutics.
Collapse
Key Words
- DC, dendritic cell
- DC-based vaccination
- FDA, Food and Drug Administration
- IFN, interferon
- MRC1, mannose receptor, C type 1
- MUC1, mucin 1
- TAA, tumor-associated antigen
- TLR, Toll-like receptor
- Toll-like receptor agonists
- Treg, regulatory T cell
- WT1, Wilms tumor 1
- antigen cross-presentation
- autophagy
- iDC, immature DC
- immunogenic cell death
- mDC, mature DC
- pDC, plasmacytoid DC
- regulatory T cells
Collapse
Affiliation(s)
- Norma Bloy
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France ; Université Paris-Sud/Paris XI ; Orsay, France
| | - Jonathan Pol
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France
| | - Fernando Aranda
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France
| | | | - Isabelle Cremer
- INSERM , U1138; Paris France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris France ; Université Pierre et Marie Curie/Paris VI ; Paris France
| | - Wolf Hervé Fridman
- INSERM , U1138; Paris France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris France ; Université Pierre et Marie Curie/Paris VI ; Paris France
| | - Jitka Fučíková
- Department of Immunology; 2nd Medical School Charles University and University Hospital Motol ; Prague, Czech Republic ; Sotio a.s. ; Prague, Czech Republic
| | - Jérôme Galon
- INSERM , U1138; Paris France ; Université Pierre et Marie Curie/Paris VI ; Paris France ; Laboratory of Integrative Cancer Immunology; Centre de Recherche des Cordeliers ; Paris France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France ; INSERM , U970; Paris France ; Pôle de Biologie; Hôpital Européen Georges Pompidou, AP-HP ; Paris France
| | - Radek Spisek
- Department of Immunology; 2nd Medical School Charles University and University Hospital Motol ; Prague, Czech Republic ; Sotio a.s. ; Prague, Czech Republic
| | - Madhav V Dhodapkar
- Department of Medicine; Immunobiology and Yale Cancer Center; Yale University ; New Haven, CT USA
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1015, CICBT507 ; Villejuif, France
| | - Guido Kroemer
- INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France ; Pôle de Biologie; Hôpital Européen Georges Pompidou, AP-HP ; Paris France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France
| |
Collapse
|
9
|
Manganese(II) complexes of quinoline derivatives: characterization, catalase activity, interaction with mitochondria and anticancer activity. TRANSIT METAL CHEM 2014. [DOI: 10.1007/s11243-014-9876-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
10
|
S100 to receptor for advanced glycation end-products binding assay: Looking for inhibitors. Biochem Biophys Res Commun 2014; 446:404-9. [PMID: 24613850 DOI: 10.1016/j.bbrc.2014.02.143] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 02/28/2014] [Indexed: 01/14/2023]
|