1
|
Slezak AJ, Chang K, Beckman TN, Refvik KC, Alpar AT, Lauterbach AL, Solanki A, Kwon JW, Gomes S, Mansurov A, Hubbell JA. Cysteine-binding adjuvant enhances survival and promotes immune function in a murine model of acute myeloid leukemia. Blood Adv 2024; 8:1747-1759. [PMID: 38324726 PMCID: PMC10985806 DOI: 10.1182/bloodadvances.2023012529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024] Open
Abstract
ABSTRACT Therapeutic vaccination has long been a promising avenue for cancer immunotherapy but is often limited by tumor heterogeneity. The genetic and molecular diversity between patients often results in variation in the antigens present on cancer cell surfaces. As a result, recent research has focused on personalized cancer vaccines. Although promising, this strategy suffers from time-consuming production, high cost, inaccessibility, and targeting of a limited number of tumor antigens. Instead, we explore an antigen-agnostic polymeric in situ cancer vaccination platform for treating blood malignancies, in our model here with acute myeloid leukemia (AML). Rather than immunizing against specific antigens or targeting adjuvant to specific cell-surface markers, this platform leverages a characteristic metabolic and enzymatic dysregulation in cancer cells that produces an excess of free cysteine thiols on their surfaces. These thiols increase in abundance after treatment with cytotoxic agents such as cytarabine, the current standard of care in AML. The resulting free thiols can undergo efficient disulfide exchange with pyridyl disulfide (PDS) moieties on our construct and allow for in situ covalent attachment to cancer cell surfaces and debris. PDS-functionalized monomers are incorporated into a statistical copolymer with pendant mannose groups and TLR7 agonists to target covalently linked antigen and adjuvant to antigen-presenting cells in the liver and spleen after IV administration. There, the compound initiates an anticancer immune response, including T-cell activation and antibody generation, ultimately prolonging survival in cancer-bearing mice.
Collapse
Affiliation(s)
- Anna J. Slezak
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Kevin Chang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Taryn N. Beckman
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL
| | - Kirsten C. Refvik
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Aaron T. Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | | | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL
| | - Jung Woo Kwon
- Department of Pathology, University of Chicago, Chicago, IL
| | - Suzana Gomes
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Aslan Mansurov
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
- Committee on Immunology, University of Chicago, Chicago, IL
- Committee on Cancer Biology, University of Chicago, Chicago, IL
| |
Collapse
|
2
|
Yu F, Chen Y, Zhou M, Liu L, Liu B, Liu J, Pan T, Luo Y, Zhang X, Ou H, Huang W, Lv X, Xi Z, Xiao R, Li W, Cao L, Ma X, Zhang J, Lu L, Zhang H. Generation of a new therapeutic D-peptide that induces the differentiation of acute myeloid leukemia cells through A TLR-2 signaling pathway. Cell Death Discov 2024; 10:51. [PMID: 38272890 PMCID: PMC10810823 DOI: 10.1038/s41420-024-01822-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 12/28/2023] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Acute myeloid leukemia (AML) is caused by clonal disorders of hematopoietic stem cells. Differentiation therapy is emerging as an important treatment modality for leukemia, given its less toxicity and wider applicable population, but the arsenal of differentiation-inducing agents is still very limited. In this study, we adapted a competitive peptide phage display platform to search for candidate peptides that could functionally induce human leukemia cell differentiation. A monoclonal phage (P6) and the corresponding peptide (pep-P6) were identified. Both L- and D-chirality of pep-P6 showed potent efficiency in inducing AML cell line differentiation, driving their morphologic maturation and upregulating the expression of macrophage markers and cytokines, including CD11b, CD14, IL-6, IL-1β, and TNF-α. In the THP-1 xenograft animal model, administration of D-pep-P6 was effective in inhibiting disease progression. Importantly, exposure to D-pep-P6 induced the differentiation of primary human leukemia cells isolated AML patients in a similar manner to the AML cell lines. Further mechanism study suggested that D-pep-P6 induced human leukemia cell differentiation by directly activating a TLR-2 signaling pathway. These findings identify a novel D-peptide that may promote leukemia differentiation therapy.
Collapse
Affiliation(s)
- Fei Yu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Yingshi Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Mo Zhou
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lingling Liu
- Department of Hematology, The Third Affiliated Hospital, Sun-yat Sen University, Guangzhou, Guangdong, China
| | - Bingfeng Liu
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Liu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Pan
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuewen Luo
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xu Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hailan Ou
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Wenjing Huang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xi Lv
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Zhihui Xi
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ruozhi Xiao
- Department of Hematology, The Third Affiliated Hospital, Sun-yat Sen University, Guangzhou, Guangdong, China
| | - Wenyu Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Lixue Cao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| | - Xiancai Ma
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, China.
| | - Jingwen Zhang
- Department of Hematology, The Third Affiliated Hospital, Sun-yat Sen University, Guangzhou, Guangdong, China.
| | - Lijuan Lu
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Hui Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Veneziani I, Alicata C, Moretta L, Maggi E. Human toll-like receptor 8 (TLR8) in NK cells: Implication for cancer immunotherapy. Immunol Lett 2023; 261:13-16. [PMID: 37451320 DOI: 10.1016/j.imlet.2023.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 07/18/2023]
Abstract
Toll-like receptors (TLR)s are homo- or heterodimeric proteins, whose structure and function were widely described in the antigen presenting cells (APC), such as Dendritic cells (DC). Recently, the expression and the role of TLRs in fighting against pathogens, was described also in NK cells. Their activation and functional properties can be directly and indirectly modulated by agonists for TLRs. In particular CD56bright NK cells subset, that is the most abundant NK cell subset in tissues and tumor microenvironment (TME), was mostly activated in terms of pro-inflammatory cytokine production, proliferation and cytotoxicity, by agonists specific for endosomal TLR8. The interplay between DC and NK, that depends on both cell-to-cell contact and soluble factors such as cytokines, promote both DC maturation and NK cell activation. Based on this concept, a TLR based immunotherapy aimed to activate NK-DC axis, may modulate TME by inducing a pro-inflammatory phenotype, thus improving DC ability to present tumor-associated antigens to T cells, and NK cell cytotoxicity against tumor cells. In this mini-review, we report data of recent literature about TLRs on human NK cells and their application as adjuvant in cancer vaccines or in combined tumor immunotherapy.
Collapse
Affiliation(s)
- Irene Veneziani
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Claudia Alicata
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
4
|
Veneziani I, Alicata C, Moretta L, Maggi E. The Latest Approach of Immunotherapy with Endosomal TLR Agonists Improving NK Cell Function: An Overview. Biomedicines 2022; 11:biomedicines11010064. [PMID: 36672572 PMCID: PMC9855813 DOI: 10.3390/biomedicines11010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptors (TLRs) are the most well-defined pattern recognition receptors (PRR) of several cell types recognizing pathogens and triggering innate immunity. TLRs are also expressed on tumor cells and tumor microenvironment (TME) cells, including natural killer (NK) cells. Cell surface TLRs primarily recognize extracellular ligands from bacteria and fungi, while endosomal TLRs recognize microbial DNA or RNA. TLR engagement activates intracellular pathways leading to the activation of transcription factors regulating gene expression of several inflammatory molecules. Endosomal TLR agonists may be considered as new immunotherapeutic adjuvants for dendritic cell (DC) vaccines able to improve anti-tumor immunity and cancer patient outcomes. The literature suggests that endosomal TLR agonists modify TME on murine models and human cancer (clinical trials), providing evidence that locally infused endosomal TLR agonists may delay tumor growth and induce tumor regression. Recently, our group demonstrated that CD56bright NK cell subset is selectively responsive to TLR8 engagement. Thus, TLR8 agonists (loaded or not to nanoparticles or other carriers) can be considered a novel strategy able to promote anti-tumor immunity. TLR8 agonists can be used to activate and expand in vitro circulating or intra-tumoral NK cells to be adoptively transferred into patients.
Collapse
Affiliation(s)
- Irene Veneziani
- Translational Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Claudia Alicata
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Enrico Maggi
- Translational Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
- Correspondence:
| |
Collapse
|
5
|
Liu X, Sato N, Yabushita T, Li J, Jia Y, Tamura M, Asada S, Fujino T, Fukushima T, Yonezawa T, Tanaka Y, Fukuyama T, Tsuchiya A, Shikata S, Iwamura H, Kinouchi C, Komatsu K, Yamasaki S, Shibata T, Sasaki AT, Schibler J, Wunderlich M, O'Brien E, Mizukawa B, Mulloy JC, Sugiura Y, Takizawa H, Shibata T, Miyake K, Kitamura T, Goyama S. IMPDH inhibition activates TLR-VCAM1 pathway and suppresses the development of MLL-fusion leukemia. EMBO Mol Med 2022; 15:e15631. [PMID: 36453131 PMCID: PMC9832838 DOI: 10.15252/emmm.202115631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
Inosine monophosphate dehydrogenase (IMPDH) is a rate-limiting enzyme in de novo guanine nucleotide synthesis pathway. Although IMPDH inhibitors are widely used as effective immunosuppressants, their antitumor effects have not been proven in the clinical setting. Here, we found that acute myeloid leukemias (AMLs) with MLL-fusions are susceptible to IMPDH inhibitors in vitro. We also showed that alternate-day administration of IMPDH inhibitors suppressed the development of MLL-AF9-driven AML in vivo without having a devastating effect on immune function. Mechanistically, IMPDH inhibition induced overactivation of Toll-like receptor (TLR)-TRAF6-NF-κB signaling and upregulation of an adhesion molecule VCAM1, which contribute to the antileukemia effect of IMPDH inhibitors. Consequently, combined treatment with IMPDH inhibitors and the TLR1/2 agonist effectively inhibited the development of MLL-fusion AML. These findings provide a rational basis for clinical testing of IMPDH inhibitors against MLL-fusion AMLs and potentially other aggressive tumors with active TLR signaling.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoTokyoJapan
| | - Naru Sato
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tomohiro Yabushita
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Jingmei Li
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoTokyoJapan
| | - Yuhan Jia
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoTokyoJapan
| | - Moe Tamura
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoTokyoJapan
| | - Shuhei Asada
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan,The Institute of Laboratory Animals, Tokyo Women's Medical UniversityTokyoJapan
| | - Takeshi Fujino
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tsuyoshi Fukushima
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Taishi Yonezawa
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoTokyoJapan
| | - Yosuke Tanaka
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tomofusa Fukuyama
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Akiho Tsuchiya
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Shiori Shikata
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Hiroyuki Iwamura
- FUJIFILM Corporation: Pharmaceutical Products DivisionTokyoJapan
| | - Chieko Kinouchi
- FUJIFILM Corporation: Bio Science & Engineering LaboratoriesKanagawaJapan
| | - Kensuke Komatsu
- FUJIFILM Corporation: Bio Science & Engineering LaboratoriesKanagawaJapan
| | - Satoshi Yamasaki
- Laboratory of Molecular Medicine, Human Genome Center, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tatsuhiro Shibata
- Laboratory of Molecular Medicine, Human Genome Center, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Atsuo T Sasaki
- Division of Hematology and Oncology, Department of Internal MedicineUniversity of CincinnatiCincinnatiOHUSA
| | - Janet Schibler
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
| | - Eric O'Brien
- Division of Oncology, Department of Pediatrics, University of CincinnatiCincinnatiOHUSA
| | - Benjamin Mizukawa
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
| | - James C Mulloy
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
| | - Yuki Sugiura
- Department of BiochemistryKeio University School of MedicineTokyoJapan
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, International Research Center for Medical SciencesKumamoto UniversityKumamotoJapan
| | - Takuma Shibata
- Division of Innate Immunity, Department of Microbiology and ImmunologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and ImmunologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Toshio Kitamura
- Division of Cellular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Susumu Goyama
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoTokyoJapan
| |
Collapse
|
6
|
Gamlen HA, Romer-Seibert JS, Lawler ME, Versace AM, Goetz ML, Feng Y, Guryanova OA, Palmisiano N, Meyer SE. miR-196b-TLR7/8 Signaling Axis Regulates Innate Immune Signaling and Myeloid Maturation in DNMT3A-Mutant AML. Clin Cancer Res 2022; 28:4574-4586. [PMID: 35943291 PMCID: PMC9588567 DOI: 10.1158/1078-0432.ccr-22-1598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/13/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE DNMT3A mutations confer a poor prognosis in acute myeloid leukemia (AML), but the molecular mechanisms downstream of DNMT3A mutations in disease pathogenesis are not completely understood, limiting targeted therapeutic options. The role of miRNA in DNMT3A-mutant AML pathogenesis is understudied. EXPERIMENTAL DESIGN DNA methylation and miRNA expression was evaluated in human AML patient samples and in Dnmt3a/Flt3-mutant AML mice. The treatment efficacy and molecular mechanisms of TLR7/8-directed therapies on DNMT3A-mutant AML were evaluated in vitro on human AML patient samples and in Dnmt3a/Flt3-mutant AML mice. RESULTS miR-196b is hypomethylated and overexpressed in DNMT3A-mutant AML and is associated with poor patient outcome. miR-196b overexpression in DNMT3A-mutant AML is important to maintain an immature state and leukemic cell survival through repression of TLR signaling. The TLR7/8 agonist resiquimod induces dendritic cell-like differentiation with costimulatory molecule expression in DNMT3A-mutant AML cells and provides a survival benefit to Dnmt3a/Flt3-mutant AML mice. The small molecule bryostatin-1 augments resiquimod-mediated AML growth inhibition and differentiation. CONCLUSIONS DNMT3A loss-of-function mutations cause miRNA locus-specific hypomethylation and overexpression important for mutant DNMT3A-mediated pathogenesis and clinical outcomes. Specifically, the overexpression of miR-196b in DNMT3A-mutant AML creates a novel therapeutic vulnerability by controlling sensitivity to TLR7/8-directed therapies.
Collapse
Affiliation(s)
- Holly A. Gamlen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, USA
| | | | - Michael E. Lawler
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, USA
| | - Amanda M. Versace
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, USA
| | - Melanie L. Goetz
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, USA
| | - Yang Feng
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, USA
| | - Olga A. Guryanova
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, USA,University of Florida Health Cancer Center, USA
| | - Neil Palmisiano
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, USA
| | - Sara E. Meyer
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, USA,Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, USA, Address correspondence to: Sara E. Meyer, Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 233 S. 10 St., Philadelphia, PA 19107,
| |
Collapse
|
7
|
A single-beam of light priming the immune responses and boosting cancer photoimmunotherapy. J Control Release 2022; 350:734-747. [PMID: 36063959 DOI: 10.1016/j.jconrel.2022.08.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 08/22/2022] [Accepted: 08/28/2022] [Indexed: 11/23/2022]
Abstract
Mirroring the rapid clinical performance, immune checkpoint blockade (ICB) leads a remarkable clinical advance in combating cancer, but suffers poor response in most cancers. The low presence of tumor-infiltration lymphocytes and the poor immunogenicity in tumor microenvironment (TME) are the main factors hindering the effectiveness of ICB in the treatment of immunological "cold" tumors. Aiming at boosting immune response via TME modulation, we report a near-infrared laser-guided photoimmuno-strategy in which synergistic phototherapy, immune adjuvant, and ICB are integrated into one versatile nanoporphyrin platform. The prepared nanoporphyrins are self-assembled from purpurin18-lipids and have photodynamic/photothermal and immunomodulatory effects that can be tuned under a single laser irradiation, concomitant with fluorescence or MSOT imaging. In this work, the contributions of each component in the nanoporphyrin platform were specified. In particular, phototherapy-driven in situ tumor cell death provided abundant tumor-associated antigens to initiate immune responses. With the assist of spatiotemporally delivered immune adjuvant, phototherapy potentiated tumor immunogenicity, reprogrammed "cold" tumors into "hot" ones, and sensitized tumors to ICB therapy. Further combined with PD-L1 blockade, the photoimmune-strategy substantially stimulated tumor-specific immune-responses and long-term immunological memory against primary tumor, abscopal tumor as well as metastatic foci. Such single light-primed photoimmunotherapy offers a promising solution to overcome common hurdles in ICB treatment and can potentially be integrated into existing clinical practice.
Collapse
|
8
|
Therapeutic applications of toll-like receptors (TLRs) agonists in AML. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 24:2319-2329. [PMID: 35962918 DOI: 10.1007/s12094-022-02917-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/27/2022] [Indexed: 10/15/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive type of blood cancer affecting bone marrow (BM). In AML, hematopoietic precursors are arrested in the early stages of development and are defined as the presence of ≥ 20% blasts (leukemia cells) in the BM. Toll-like receptors (TLR) are major groups of pattern recognition receptors expressed by almost all innate immune cells that enable them to detect a wide range of pathogen-associated molecular patterns and damage-associated molecular patterns to prime immune responses toward adaptive immunity. Because TLRs are commonly expressed on transformed immune system cells (ranging from blasts to memory cells), they can be a potential option for developing efficient clinical alternatives in hematologic tumors. This is because several in vitro and in vivo investigations have demonstrated that TLR signaling increased the immunogenicity of AML cells, making them more vulnerable to T cell-mediated invasion. This study aimed to review the current knowledge in this field and provide some insight into the therapeutic potentials of TLRs in AML.
Collapse
|
9
|
Veneziani I, Alicata C, Pelosi A, Landolina N, Ricci B, D'Oria V, Fagotti A, Scambia G, Moretta L, Maggi E. Toll-like receptor 8 agonists improve NK-cell function primarily targeting CD56brightCD16− subset. J Immunother Cancer 2022; 10:jitc-2021-003385. [PMID: 35091452 PMCID: PMC8804697 DOI: 10.1136/jitc-2021-003385] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 11/05/2022] Open
Abstract
Background Toll-like receptors (TLRs) are pattern-recognition sensors mainly expressed in innate immune cells that directly recognize conserved pathogen structures (pathogen-associated molecular patterns-PAMPs). Natural killer (NK) cells have been described to express different endosomal TLRs triggered by RNA and DNA sequences derived from both viruses and bacteria. This study was addressed to establish which endosomal TLR could directly mediate NK activation and function after proper stimuli. It was also important to establish the most suitable TLR agonist to be used as adjuvant in tumor vaccines or in combined cancer immunotherapies. Methods We assessed endosomal TLR expression in total NK cells by using RT-qPCR and western blotting technique. In some experiments, we purified CD56brightCD16− and CD56dimCD16+ cells subsets by using NK Cell Isolation Kit Activation marker, cytokine production, CD107a expression and cytotoxicity assay were evaluated by flow cytometry. Cytokine release was quantified by ELISA. NK cells obtained from ovarian ascites underwent the same analyses. Results Although the four endosomal TLRs (TLR3, TLR7/8, and TLR9) were uniformly expressed on CD56brightCD16− and CD56dimCD16+ cell subsets, the TLR7/8 (R848), TLR3 (polyinosinic-polycytidylic acid, Poly I:C) and TLR9 (ODN2395) ligands promoted NK-cell function only in the presence of suboptimal doses of cytokines, including interleukin (IL)-2, IL-12, IL-15, and IL-18, produced in vivo by other environmental cells. We showed that R848 rather than TLR3 and TLR9 agonists primarily activated CD56brightCD16− NK cells by increasing their proliferation, cytokine production and cytotoxic activity. Moreover, we demonstrated that R848, which usually triggers TLR7 and TLR8 on dendritic cells, macrophages and neutrophils cells, activated CD56brightCD16− NK-cell subset only via TLR8. Indeed, specific TLR8 but not TLR7 agonists increased cytokine production and cytotoxic activity of CD56brightCD16− NK cells. Importantly, these activities were also observed in peritoneal NK cells from patients with metastatic ovarian carcinoma, prevalently belonging to the CD56brightCD16− subset. Conclusion These data highlight the potential value of TLR8 in NK cells as a new target for immunotherapy in patients with cancer.
Collapse
Affiliation(s)
- Irene Veneziani
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Claudia Alicata
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Andrea Pelosi
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Nadine Landolina
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Biancamaria Ricci
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Valentina D'Oria
- Confocal Microscopy Core Facility, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Anna Fagotti
- Department of Woman's Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Campus di Roma, Roma, Lazio, Italy
| | - Giovanni Scambia
- Department of Woman's Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Campus di Roma, Roma, Lazio, Italy
| | - Lorenzo Moretta
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Enrico Maggi
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| |
Collapse
|
10
|
Li S, Yao JC, Li JT, Schmidt AP, Link DC. TLR7/8 agonist treatment induces an increase in bone marrow resident dendritic cells and hematopoietic progenitor expansion and mobilization. Exp Hematol 2021; 96:35-43.e7. [PMID: 33556431 PMCID: PMC9900459 DOI: 10.1016/j.exphem.2021.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 02/08/2023]
Abstract
There is accumulating evidence suggesting that toll-like receptor (TLR) signals play an important role in the regulation of hematopoietic stem/progenitor cells (HSPCs). TLR7/8 stimulation induces the myeloid differentiation of normal HSPCs and acute myeloid leukemia cells. However, the in vivo effect of TLR7/8 agonists on hematopoiesis is largely unknown. Here, we show that, similar to TLR4 and TLR2, treatment with the TLR7/8 agonist R848 induces an expansion of phenotypic hematopoietic stem cells (HSCs) with reduced repopulating potential and HSPC mobilization. In contrast to chronic TLR4 stimulation, treatment with R848 for 5 days did not induce a significant increase in myeloid-biased HSCs. Treatment with R848 results in a significant increase in classic dendritic cells (DCs) in the bone marrow, but a decrease in common dendritic cell progenitors and pre-DCs. Phenotypic analysis of DCs revealed that R848 treatment is associated with altered expression of certain chemokines, activation markers, and migratory receptors. Together, these data indicate that systemic administration of a TLR7/8 agonist has unique effects on hematopoiesis, including the expansion of DCs in the bone marrow, that might have clinical relevance to augment responses to certain immunotherapies, such as cancer vaccines and immune checkpoint blockade.
Collapse
Affiliation(s)
- Sidan Li
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.,Hematology Oncology Center, Beijing Children’s Hospital, National Center for Children’s Health, Capital Medial University, Beijing, China
| | - Juo-Chin Yao
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Justin T. Li
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Amy P. Schmidt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Daniel C. Link
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
11
|
Mer AS, Heath EM, Madani Tonekaboni SA, Dogan-Artun N, Nair SK, Murison A, Garcia-Prat L, Shlush L, Hurren R, Voisin V, Bader GD, Nislow C, Rantalainen M, Lehmann S, Gower M, Guidos CJ, Lupien M, Dick JE, Minden MD, Schimmer AD, Haibe-Kains B. Biological and therapeutic implications of a unique subtype of NPM1 mutated AML. Nat Commun 2021; 12:1054. [PMID: 33594052 PMCID: PMC7886883 DOI: 10.1038/s41467-021-21233-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/15/2021] [Indexed: 01/29/2023] Open
Abstract
In acute myeloid leukemia (AML), molecular heterogeneity across patients constitutes a major challenge for prognosis and therapy. AML with NPM1 mutation is a distinct genetic entity in the revised World Health Organization classification. However, differing patterns of co-mutation and response to therapy within this group necessitate further stratification. Here we report two distinct subtypes within NPM1 mutated AML patients, which we label as primitive and committed based on the respective presence or absence of a stem cell signature. Using gene expression (RNA-seq), epigenomic (ATAC-seq) and immunophenotyping (CyToF) analysis, we associate each subtype with specific molecular characteristics, disease differentiation state and patient survival. Using ex vivo drug sensitivity profiling, we show a differential drug response of the subtypes to specific kinase inhibitors, irrespective of the FLT3-ITD status. Differential drug responses of the primitive and committed subtype are validated in an independent AML cohort. Our results highlight heterogeneity among NPM1 mutated AML patient samples based on stemness and suggest that the addition of kinase inhibitors to the treatment of cases with the primitive signature, lacking FLT3-ITD, could have therapeutic benefit.
Collapse
Affiliation(s)
- Arvind Singh Mer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Karolinska Institute, Stockholm, Sweden
| | - Emily M Heath
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Seyed Ali Madani Tonekaboni
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Nergiz Dogan-Artun
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Alex Murison
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Laura Garcia-Prat
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Liran Shlush
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Corey Nislow
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
| | | | | | - Mark Gower
- The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.
- Ontario Institute for Cancer Research, Toronto, ON, Canada.
- Vector Institute, Toronto, ON, Canada.
| |
Collapse
|
12
|
Van Leeuwen-Kerkhoff N, Westers TM, Poddighe PJ, Povoleri GAM, Timms JA, Kordasti S, De Gruijl TD, Van de Loosdrecht AA. Reduced frequencies and functional impairment of dendritic cell subsets and non-classical monocytes in myelodysplastic syndromes. Haematologica 2021; 107:655-667. [PMID: 33567812 PMCID: PMC8883570 DOI: 10.3324/haematol.2020.268136] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Indexed: 11/09/2022] Open
Abstract
In myelodysplastic syndromes (MDS) the immune system is involved in pathogenesis as well as in disease progression. Dendritic cells (DC) are key players of the immune system by serving as regulators of immune responses. Their function has been scarcely studied in MDS and most of the reported studies didn't investigate naturally occurring DC subsets. Therefore, we here examined the frequency and function of DC subsets and slan+ non-classical monocytes in various MDS risk groups. Frequencies of DC as well as of slan+ monocytes were decreased in MDS bone marrow (BM) compared to normal bone marrow (NBM) samples. Transcriptional profiling revealed down-regulation of transcripts related to pro-inflammatory pathways in MDS-derived cells as compared to NBM. Additionally, their capacity to induce T cell proliferation was impaired. Multidimensional mass cytometry showed that whereas healthy donor-derived slan+ monocytes supported Th1/Th17/Treg differentiation/expansion their MDS-derived counterparts also mediated substantial Th2 expansion. Our findings point to a role for an impaired ability of DC subsets to adequately respond to cellular stress and DNA damage in the immune escape and progression of MDS. As such, it paves the way toward potential novel immunotherapeutic interventions.
Collapse
Affiliation(s)
- Nathalie Van Leeuwen-Kerkhoff
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam
| | - Theresia M Westers
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam
| | - Pino J Poddighe
- Department of Clinical Genetics, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam
| | - Giovanni A M Povoleri
- Department Inflammation Biology, King's College London, Centre for Inflammation Biology and Cancer Immunology, London
| | - Jessica A Timms
- Systems Cancer Immunology Lab, Comprehensive Cancer Center, King's College London, London
| | - Shahram Kordasti
- Systems Cancer Immunology Lab, Comprehensive Cancer Center, King's College London, London, United Kingdom; Dipartimento Scienze Cliniche e Molecolari, UNIVPM, Ancona
| | - Tanja D De Gruijl
- Department of Medical Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam
| | - Arjan A Van de Loosdrecht
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam.
| |
Collapse
|
13
|
Gadanec LK, McSweeney KR, Qaradakhi T, Ali B, Zulli A, Apostolopoulos V. Can SARS-CoV-2 Virus Use Multiple Receptors to Enter Host Cells? Int J Mol Sci 2021; 22:992. [PMID: 33498183 PMCID: PMC7863934 DOI: 10.3390/ijms22030992] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
The occurrence of the novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), responsible for coronavirus disease 2019 (COVD-19), represents a catastrophic threat to global health. Protruding from the viral surface is a densely glycosylated spike (S) protein, which engages angiotensin-converting enzyme 2 (ACE2) to mediate host cell entry. However, studies have reported viral susceptibility in intra- and extrapulmonary immune and non-immune cells lacking ACE2, suggesting that the S protein may exploit additional receptors for infection. Studies have demonstrated interactions between S protein and innate immune system, including C-lectin type receptors (CLR), toll-like receptors (TLR) and neuropilin-1 (NRP1), and the non-immune receptor glucose regulated protein 78 (GRP78). Recognition of carbohydrate moieties clustered on the surface of the S protein may drive receptor-dependent internalization, accentuate severe immunopathological inflammation, and allow for systemic spread of infection, independent of ACE2. Furthermore, targeting TLRs, CLRs, and other receptors (Ezrin and dipeptidyl peptidase-4) that do not directly engage SARS-CoV-2 S protein, but may contribute to augmented anti-viral immunity and viral clearance, may represent therapeutic targets against COVID-19.
Collapse
|
14
|
Silencing TLR4/MyD88/NF-κB Signaling Pathway Alleviated Inflammation of Corneal Epithelial Cells Infected by ISE. Inflammation 2020; 44:633-644. [PMID: 33174138 DOI: 10.1007/s10753-020-01363-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
The regulatory role of toll-like receptor 4 (TLR4) in the inactivate staphylococcus epidermidis (ISE)-induced cornea inflammation is not well investigated. Here, TLR4 silence could decrease inflammatory cytokines in corneal epithelial cells treated with ISE. The mouse corneal epithelial cells were exposed to ISE for 24 h, either alone or with the NF-κB inhibitor, TLR4 lentivirus to bilaterally (knock-down or and overexpression). The expression of TLR4 in mouse corneal epithelial cells was investigated using western blot and qRT-PCR assay. The inflammatory cytokine levels were evaluated by qRT-PCR and ELISA, respectively. The relative impact factors of TLR4-mediated NF-κB signaling detected using western blot assay. Results show the expression levels of TLR4 and some inflammatory cytokines were significantly increased in corneal epithelial cells treated with ISE. TLR4 Silence markedly decreased ISE-induced production of IL12, TNF-α, CCL5, and CCL9 in corneal epithelial cells. Furthermore, the nuclear translocation of NF-κB p65 and myeloid differentiation protein 88 (MyD88) in the cells treated with ISE were further reduced by silencing TLR4. Inhibition of TLR4-mediated NF-κB signaling by using BAY11-7082 also alleviated ISE-induced inflammation. In the rescue experiment, transfected the stable TLR4 silenced corneal epithelial cells with TLR4 overexpression lentivirus, we found that TLR4 overexpression can restore the down-regulation of TLR4 and inflammatory cytokines (IL12, TNF-α, CCL9) caused by TLR4 knocked down. Therefore, ISE-induced cornea inflammation was due to the activation of the TLR4/MyD88/NF-κB signaling pathway, and dramatically stimulated IL12, TNF-α, CCL9 secretion. TLR4 silence presented mitigates damage in corneal epithelial cells treated with ISE.
Collapse
|
15
|
Wang Y, Yang H, Li H, Zhao S, Zeng Y, Zhang P, Lin X, Sun X, Wang L, Fu G, Gao Y, Wang P, Gao D. Development of a novel TLR8 agonist for cancer immunotherapy. MOLECULAR BIOMEDICINE 2020; 1:6. [PMID: 35006413 PMCID: PMC8607422 DOI: 10.1186/s43556-020-00007-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/14/2020] [Indexed: 01/12/2023] Open
Abstract
Toll-like receptors (TLRs) are a family of proteins that recognize pathogen associated molecular patterns (PAMPs). Their primary function is to activate innate immune responses while also involved in facilitating adaptive immune responses. Different TLRs exert distinct functions by activating varied immune cascades. Several TLRs are being pursued as cancer drug targets. We discovered a novel, highly potent and selective small molecule TLR8 agonist DN052. DN052 exhibited strong in vitro cellular activity with EC50 at 6.7 nM and was highly selective for TLR8 over other TLRs including TLR4, 7 and 9. DN052 displayed excellent in vitro ADMET and in vivo PK profiles. DN052 potently inhibited tumor growth as a single agent. Moreover, combination of DN052 with the immune checkpoint inhibitor, selected targeted therapeutics or chemotherapeutic drugs further enhanced efficacy of single agents. Mechanistically, treatment with DN052 resulted in strong induction of pro-inflammatory cytokines in ex vivo human PBMC assay and in vivo monkey study. GLP toxicity studies in rats and monkeys demonstrated favorable safety profile. This led to the advancement of DN052 into phase 1 clinical trials.
Collapse
Affiliation(s)
- Yuxun Wang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China.
| | - Heping Yang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Huanping Li
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Shuda Zhao
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Yikun Zeng
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Panpan Zhang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Xiaoqin Lin
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Xiaoxiang Sun
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Longsheng Wang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Guangliang Fu
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Yaqiao Gao
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Pei Wang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Daxin Gao
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| |
Collapse
|
16
|
Li J, Ran Q, Xu B, Luo X, Song S, Xu D, Zhang X. Role of CD25 expression on prognosis of acute myeloid leukemia: A literature review and meta-analysis. PLoS One 2020; 15:e0236124. [PMID: 32687530 PMCID: PMC7371194 DOI: 10.1371/journal.pone.0236124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
The gene expression for interleukin-2 receptor subunit alpha (CD25/IL2RA) is frequently altered in adults with acute myeloid leukemia (AML). Increasing evidence indicates that the elevated expression of CD25 may be correlated with poor survival for AML patients. Thus, we performed this meta-analysis to further evaluate the prognostic value of elevated CD25 in AML. Eligible studies were gathered by searching on PubMed, Web of Science, and Embase. Using the R language 3.6.0 software, Pooled hazard ratios (HRs) with their corresponding 95% confidence intervals (CIs) of overall survival (OS) and disease-free survival (DFS)/relapse-free survival (RFS)/event-free survival (EFS) for total and subgroup analyses were calculated to investigate the association of elevated CD25 and outcomes of AML patients. Ten studies with a total of 1640 participants were enrolled in this meta-analysis. Pooled HRs suggested that overexpression of CD25 predicted poor outcomes on both OS (HR = 2.27, 95%CI 1.95–2.64) and DFS/RFS/EFS (HR = 1.77, 95%CI 1.44–2.17) in overall population. Subgroup analyses stratified by ethnicity, AML subtype, cut-off value, statistical methodologies and detection method draw similar results. Our meta-analysis indicates that elevated CD25 expression is a poor prognostic factor for AML patients. Considering limited number of samples, further relevant studies are warranted.
Collapse
Affiliation(s)
- Jingyuan Li
- Department of Hematology, The General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Qijie Ran
- Department of Hematology, The General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Biao Xu
- Department of Hematology, The General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Xiaojing Luo
- Department of Hematology, The General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Senhua Song
- Department of Hematology, The General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Dehong Xu
- Department of Hematology, The General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Xinhua Zhang
- Department of Hematology, The General Hospital of Central Theater Command, Wuhan, Hubei Province, China
- * E-mail:
| |
Collapse
|
17
|
Teodorescu P, Pasca S, Dima D, Tomuleasa C, Ghiaur G. Targeting the Microenvironment in MDS: The Final Frontier. Front Pharmacol 2020; 11:1044. [PMID: 32742264 PMCID: PMC7364152 DOI: 10.3389/fphar.2020.01044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of malignant disorders of hematopoietic stem and progenitor cells (HSPC), mainly characterized by ineffective hematopoiesis leading to peripheral cytopenias and progressive bone marrow failure. While clonal dominance is nearly universal at diagnosis, most genetic mutations identified in patients with MDS do not provide a conspicuous advantage to the malignant cells. In this context, malignant cells alter their adjacent bone marrow microenvironment (BME) and rely on cell extrinsic factors to maintain clonal dominance. The profoundly disturbed BME favors the myelodysplastic cells and, most importantly is detrimental to normal hematopoietic cells. Thus, the MDS microenvironment not only contributes to the observed cytopenias seen in these patients but could also negatively impact the engraftment of normal, allogeneic HSPCs in patients with MDS undergoing bone marrow transplant. Therefore, successful therapies in MDS should not only target the malignant cells but also reprogram their bone marrow microenvironment. Here, we will provide a synopsis of how drugs currently used or on the verge of being approved for the treatment of MDS may achieve this goal.
Collapse
Affiliation(s)
- Patric Teodorescu
- Department of Hematology, Iuliu Hategan University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Oncology, The Johns Hopkins Hospital, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hategan University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Delia Dima
- Department of Hematology, Iuliu Hategan University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hategan University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriel Ghiaur
- Department of Oncology, The Johns Hopkins Hospital, Johns Hopkins Medicine, Baltimore, MD, United States
| |
Collapse
|
18
|
Paracatu LC, Schuettpelz LG. Contribution of Aberrant Toll Like Receptor Signaling to the Pathogenesis of Myelodysplastic Syndromes. Front Immunol 2020; 11:1236. [PMID: 32625214 PMCID: PMC7313547 DOI: 10.3389/fimmu.2020.01236] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Toll like receptors (TLRs) are a family of pattern recognition receptors that play a central role in the innate immune response. These receptors are expressed on a wide variety of immune and non-immune cells, and they help shape the immune response to infection and injury through the recognition of pathogen-associated molecular patterns (PAMPs) as well as endogenous damage-associated molecular patterns (DAMPs). Accumulating evidence suggests that, in addition to regulating mature effector immune cells, TLRs can influence the immune response from the level of the hematopoietic stem cell (HSC). HSCs express TLRs, and exposure to TLR ligands influences the cycling, differentiation, and function of HSCs, with chronic TLR stimulation leading to impairment of normal HSC repopulating activity. Moreover, enhanced TLR expression and signaling is associated with myelodysplastic syndromes (MDS), a heterogenous group of HSC disorders characterized by ineffective hematopoiesis and a high risk of transformation to acute leukemias. In this review, we will discuss the role of TLR signaling in the pathogenesis of MDS, focusing on the known direct and indirect effects of this type of signaling on HSCs, the mechanisms of TLR signaling upregulation in MDS, the changes in TLR expression with disease progression, and the therapeutic implications for modulating TLR signaling in the treatment of MDS.
Collapse
Affiliation(s)
- Luana Chiquetto Paracatu
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Laura G Schuettpelz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
19
|
Schönherz AA, Bødker JS, Schmitz A, Brøndum RF, Jakobsen LH, Roug AS, Severinsen MT, El-Galaly TC, Jensen P, Johnsen HE, Bøgsted M, Dybkær K. Normal myeloid progenitor cell subset-associated gene signatures for acute myeloid leukaemia subtyping with prognostic impact. PLoS One 2020; 15:e0229593. [PMID: 32324791 PMCID: PMC7179860 DOI: 10.1371/journal.pone.0229593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/10/2020] [Indexed: 12/30/2022] Open
Abstract
Acute myeloid leukaemia (AML) is characterised by phenotypic heterogeneity, which we hypothesise is a consequence of deregulated differentiation with transcriptional reminiscence of the normal compartment or cell-of-origin. Here, we propose a classification system based on normal myeloid progenitor cell subset-associated gene signatures (MAGS) for individual assignments of AML subtypes. We generated a MAGS classifier including the progenitor compartments CD34+/CD38- for haematopoietic stem cells (HSCs), CD34+/CD38+/CD45RA- for megakaryocyte-erythroid progenitors (MEPs), and CD34+/CD38+/CD45RA+ for granulocytic-monocytic progenitors (GMPs) using regularised multinomial regression with three discrete outcomes and an elastic net penalty. The regularisation parameters were chosen by cross-validation, and MAGS assignment accuracy was validated in an independent data set (N = 38; accuracy = 0.79) of sorted normal myeloid subpopulations. The prognostic value of MAGS assignment was studied in two clinical cohorts (TCGA: N = 171; GSE6891: N = 520) and had a significant prognostic impact. Furthermore, multivariate Cox regression analysis using the MAGS subtype, FAB subtype, cytogenetics, molecular genetics, and age as explanatory variables showed independent prognostic value. Molecular characterisation of subtypes by differential gene expression analysis, gene set enrichment analysis, and mutation patterns indicated reduced proliferation and overrepresentation of RUNX1 and IDH2 mutations in the HSC subtype; increased proliferation and overrepresentation of CEBPA mutations in the MEP subtype; and innate immune activation and overrepresentation of WT1 mutations in the GMP subtype. We present a differentiation-dependent classification system for AML subtypes with distinct pathogenetic and prognostic importance that can help identify candidates poorly responding to combination chemotherapy and potentially guide alternative treatments.
Collapse
Affiliation(s)
- Anna A. Schönherz
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Julie Støve Bødker
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Alexander Schmitz
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Rasmus Froberg Brøndum
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Lasse Hjort Jakobsen
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Anne Stidsholt Roug
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Marianne T. Severinsen
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Tarec C. El-Galaly
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Paw Jensen
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Hans Erik Johnsen
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Martin Bøgsted
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Karen Dybkær
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- * E-mail:
| |
Collapse
|
20
|
Patinote C, Karroum NB, Moarbess G, Cirnat N, Kassab I, Bonnet PA, Deleuze-Masquéfa C. Agonist and antagonist ligands of toll-like receptors 7 and 8: Ingenious tools for therapeutic purposes. Eur J Med Chem 2020; 193:112238. [PMID: 32203790 PMCID: PMC7173040 DOI: 10.1016/j.ejmech.2020.112238] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
The discovery of the TLRs family and more precisely its functions opened a variety of gates to modulate immunological host responses. TLRs 7/8 are located in the endosomal compartment and activate a specific signaling pathway in a MyD88-dependant manner. According to their involvement into various autoimmune, inflammatory and malignant diseases, researchers have designed diverse TLRs 7/8 ligands able to boost or block the inherent signal transduction. These modulators are often small synthetic compounds and most act as agonists and to a much lesser extent as antagonists. Some of them have reached preclinical and clinical trials, and only one has been approved by the FDA and EMA, imiquimod. The key to the success of these modulators probably lies in their combination with other therapies as recently demonstrated. We gather in this review more than 360 scientific publications, reviews and patents, relating the extensive work carried out by researchers on the design of TLRs 7/8 modulators, which are classified firstly by their biological activities (agonist or antagonist) and then by their chemical structures, which total syntheses are not discussed here. This review also reports about 90 clinical cases, thereby showing the biological interest of these modulators in multiple pathologies.
Collapse
Affiliation(s)
- Cindy Patinote
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Nour Bou Karroum
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France; Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Georges Moarbess
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Natalina Cirnat
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | - Issam Kassab
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | | | | |
Collapse
|
21
|
Furusho K, Shibata T, Sato R, Fukui R, Motoi Y, Zhang Y, Saitoh SI, Ichinohe T, Moriyama M, Nakamura S, Miyake K. Cytidine deaminase enables Toll-like receptor 8 activation by cytidine or its analogs. Int Immunol 2020; 31:167-173. [PMID: 30535046 DOI: 10.1093/intimm/dxy075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 11/16/2018] [Indexed: 12/25/2022] Open
Abstract
Toll-like receptor 8 (TLR8), a sensor for pathogen-derived single-stranded RNA (ssRNA), binds to uridine (Uri) and ssRNA to induce defense responses. We here show that cytidine (Cyd) with ssRNA also activated TLR8 in peripheral blood leukocytes (PBLs) and a myeloid cell line U937, but not in an embryonic kidney cell line 293T. Cyd deaminase (CDA), an enzyme highly expressed in leukocytes, deaminates Cyd to Uri. CDA expression enabled TLR8 response to Cyd and ssRNA in 293T cells. CDA deficiency and a CDA inhibitor both reduced TLR8 responses to Cyd and ssRNA in U937. The CDA inhibitor also reduced PBL response to Cyd and ssRNA. A Cyd analogue, azacytidine, is used for the therapy of myelodysplastic syndrome and acute myeloid leukemia. Azacytidine with ssRNA induced tumor necrosis factor-α expression in U937 and PBLs in a manner dependent on CDA and TLR8. These results suggest that CDA enables TLR8 activation by Cyd or its analogues with ssRNA through deaminating activity. Nucleoside metabolism might impact TLR8 responses in a variety of situations such as the treatment with nucleoside analogues.
Collapse
Affiliation(s)
- Katsuhiro Furusho
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan.,Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takuma Shibata
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Saitama, Japan
| | - Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | - Yuji Motoi
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | - Yun Zhang
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | - Shin-Ichiroh Saitoh
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | - Takeshi Ichinohe
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | - Masafumi Moriyama
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Seiji Nakamura
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| |
Collapse
|
22
|
Yu YH, Xin F, Dong L, Ge L, Zhai CY, Shen XL. Weighted gene coexpression network analysis identifies critical genes in different subtypes of acute myeloid leukaemia. BIOTECHNOL BIOTEC EQ 2020. [DOI: 10.1080/13102818.2020.1811767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Yan-Hui Yu
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, PR China
| | - Fei Xin
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, PR China
| | - Lu Dong
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, PR China
| | - Li Ge
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, PR China
| | - Chun-Yan Zhai
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, PR China
| | - Xu-Liang Shen
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, PR China
| |
Collapse
|
23
|
Identification of prognostic genes in the acute myeloid leukemia immune microenvironment based on TCGA data analysis. Cancer Immunol Immunother 2019; 68:1971-1978. [PMID: 31650199 DOI: 10.1007/s00262-019-02408-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 10/01/2019] [Indexed: 12/30/2022]
Abstract
Acute myeloid leukemia (AML) is a common and lethal hematopoietic malignancy that is highly dependent on the bone marrow (BM) microenvironment. Infiltrating immune and stromal cells are important components of the BM microenvironment and significantly influence the progression of AML. This study aimed to elucidate the value of immune/stromal cell-associated genes for AML prognosis by integrated bioinformatics analysis. We obtained expression profiles from The Cancer Genome Atlas (TCGA) database and used the ESTIMATE algorithm to calculate immune scores and stromal scores; we then identified differentially expressed genes (DEGs) based on these scores. Overall survival analysis was applied to reveal common DEGs of prognostic value. Subsequently, we conducted a functional enrichment analysis, generated a protein-protein interaction (PPI) network and performed an interrelation analysis of immune system processes, showing that these genes are mainly associated with the immune/inflammatory response. Finally, eight genes (CD163, CYP27A1, KCNA5, PPM1J, FOLR1, IL1R2, MYOF, VSIG2) were verified to be significantly associated with AML prognosis in the Gene Expression Omnibus (GEO) database. In summary, we identified key microenvironment-related genes that affect the outcomes of AML patients and might serve as therapeutic targets.
Collapse
|
24
|
Brattås MK, Reikvam H, Tvedt THA, Bruserud Ø. Precision medicine for TP53-mutated acute myeloid leukemia. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1644164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
| | - Håkon Reikvam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Øystein Bruserud
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
25
|
Functional Toll-Like Receptors (TLRs) Are Expressed by a Majority of Primary Human Acute Myeloid Leukemia Cells and Inducibility of the TLR Signaling Pathway Is Associated with a More Favorable Phenotype. Cancers (Basel) 2019; 11:cancers11070973. [PMID: 31336716 PMCID: PMC6678780 DOI: 10.3390/cancers11070973] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/26/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous disease with regard to biological characteristics and receptor expression. Toll-like receptors (TLRs) are upstream to the transcription factor NFκB and part of the innate immune system. They are differentially expressed on AML blasts, and during normal hematopoiesis they initiate myeloid differentiation. In this study, we investigated the response upon TLR stimulation in an AML cohort (n = 83) by measuring the increase of NFκB-mediated cytokine secretion. We observed that TLR4 is readily induced in most patients, while TLR1/2 response was more restricted. General response to TLR stimulation correlated with presence of nucleophosmin gene mutations, increased mRNA expression of proteins, which are part of the TLR signaling pathway and reduced expression of transcription-related proteins. Furthermore, signaling via TLR1/2 appeared to be linked with prolonged patient survival. In conclusion, response upon TLR stimulation, and especially TLR1/2 induction, seems to be part of a more favorable phenotype, which also is characterized by higher basal cytokine secretion and a more mature blast population.
Collapse
|
26
|
Megías J, Martínez A, San-Miguel T, Gil-Benso R, Muñoz-Hidalgo L, Albert-Bellver D, Carratalá A, Gozalbo D, López-Ginés C, Gil ML, Cerdá-Nicolás M. Pam3CSK4, a TLR2 ligand, induces differentiation of glioblastoma stem cells and confers susceptibility to temozolomide. Invest New Drugs 2019; 38:299-310. [DOI: 10.1007/s10637-019-00788-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022]
|
27
|
Sokolova TM, Poloskov VV, Shuvalov AN, Burova OS, Sokolova ZA. Signaling TLR/RLR-mechanisms of immunomodulating action of ingavirin and thymogen preparations. ACTA ACUST UNITED AC 2019. [DOI: 10.17650/1726-9784-2019-18-1-60-66] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objective: to study drugs ingavirin and thymogen as activators of signal TLR and RLR reactions in a sensitive cell model of THP-1 monocytes and blood cells of donors.Materials and methods . Investigated drugs ingavirin (imidazolylethanamide pentanedioic acid – 6-[2-(1H-imidazol-4-yl)ethylami- no]-5-oxohexanoic acid; Valenta Pharmaceutics, Russia) and thymogen (alpha-glutamyl-tryptophan; Cytomed, Russia), registered in Russia as medicines. The expression of TLR/RLR receptor genes was determined under the action of ingavirin 50–300 μg/ml and thymogen 0.1–5 μg/ml (24 h, 37 °C) using quantitative RT-PCR. The level of fluid cytokines was determined using ELISA kits (Vec- tor-Best, Russia) in the culture fluid. Transfection of small inhibitory RNA (siRNA) MAVS was performed using the reagent Lipofect- amine 2000 (Invitrogen). The immunophenotype of the THP-1 cell line was determined by flow cytometry with labeled monoclonal antibodies FITC CD14 and PE CD34 (BD Biosciences) on a FACSCanto II instrument (Becton Dickinson).Results . For the first time, it has been shown that ingavirin (imidazolylethanamide) and thymogen (dipeptide Glu-Trp) preparations are activators of the immune TLR/RLR receptors and their signaling factors genes in the cultures of monocytic leukemia THP-1 and blood of healthy donors. In these cellular systems, ingavirin and thymogen preparations elicited similar immune responses and stimulated the expression of genes: endosomal TLR3/7/8/9 receptors, RIG1/MDA5 cytoplasmic sensors and NFκB1 and MAVS signaling factors. Induced cells secrete inflammatory cytokines of TNF-α and IL1-β. Ingavirin in THP-1 cell culture monocytes caused a decrease in CD34+ blast cells. Activation the genes of MAVS and co-receptor B2M of the main histocompatibility complex (MHCII) by ingavirin were interrelated. Transfection of siRNA MAVS reduced the level of homologous mRNA MAVS and heterologous mRNA B2M. Conclusion . The results obtained suggest that the antiviral and immunomodulating properties of the drugs ingavirin and thymogen are associated with the activation of a group of TLR/RLR signaling pathways of the innate and adaptive immunity and the differentiation of hematopoietic cell precursors.
Collapse
Affiliation(s)
- T. M. Sokolova
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of the Russian Federation
| | - V. V. Poloskov
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of the Russian Federation
| | - A. N. Shuvalov
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of the Russian Federation
| | - O. S. Burova
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation
| | - Z. A. Sokolova
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation
| |
Collapse
|
28
|
Seneviratne AK, Xu M, Henao JJA, Fajardo VA, Hao Z, Voisin V, Xu GW, Hurren R, Kim S, MacLean N, Wang X, Gronda M, Jeyaraju D, Jitkova Y, Ketela T, Mullokandov M, Sharon D, Thomas G, Chouinard-Watkins R, Hawley JR, Schafer C, Yau HL, Khuchua Z, Aman A, Al-Awar R, Gross A, Claypool SM, Bazinet RP, Lupien M, Chan S, De Carvalho DD, Minden MD, Bader GD, Stark KD, LeBlanc P, Schimmer AD. The Mitochondrial Transacylase, Tafazzin, Regulates for AML Stemness by Modulating Intracellular Levels of Phospholipids. Cell Stem Cell 2019; 24:621-636.e16. [PMID: 30930145 DOI: 10.1016/j.stem.2019.02.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/19/2018] [Accepted: 02/27/2019] [Indexed: 12/17/2022]
Abstract
Tafazzin (TAZ) is a mitochondrial transacylase that remodels the mitochondrial cardiolipin into its mature form. Through a CRISPR screen, we identified TAZ as necessary for the growth and viability of acute myeloid leukemia (AML) cells. Genetic inhibition of TAZ reduced stemness and increased differentiation of AML cells both in vitro and in vivo. In contrast, knockdown of TAZ did not impair normal hematopoiesis under basal conditions. Mechanistically, inhibition of TAZ decreased levels of cardiolipin but also altered global levels of intracellular phospholipids, including phosphatidylserine, which controlled AML stemness and differentiation by modulating toll-like receptor (TLR) signaling.
Collapse
Affiliation(s)
- Ayesh K Seneviratne
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mingjing Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Juan J Aristizabal Henao
- Laboratory of Nutritional Lipidomics, Department of Kinesiology, University of Waterloo, Waterloo, ON, Canada
| | - Val A Fajardo
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Zhenyue Hao
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Veronique Voisin
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - G Wei Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - S Kim
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Neil MacLean
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xiaoming Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Danny Jeyaraju
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yulia Jitkova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Troy Ketela
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - David Sharon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Geethu Thomas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - James R Hawley
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Caitlin Schafer
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Helen Loo Yau
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Zaza Khuchua
- Department of Biochemistry, Sechenov Medical University, Moscow, Russian Federation; Institute of Medical Research Ilia State University, Tbilisi, Georgia
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, ON, Canada
| | - Rima Al-Awar
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, ON, Canada
| | - Atan Gross
- Department of Biological Regulation, Weizmann Institute, Rehovot, Israel
| | - Steven M Claypool
- Department of Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Steven Chan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ken D Stark
- Laboratory of Nutritional Lipidomics, Department of Kinesiology, University of Waterloo, Waterloo, ON, Canada
| | - Paul LeBlanc
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
29
|
Yang J, Lan J, Du H, Zhang X, Li A, Zhang X, Liu Y, Zhang J, Zhang C, Ding Y, Zhang T. Icariside II induces cell cycle arrest and differentiation via TLR8/MyD88/p38 pathway in acute myeloid leukemia cells. Eur J Pharmacol 2019; 846:12-22. [DOI: 10.1016/j.ejphar.2018.12.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/23/2022]
|
30
|
Abo Elwafa R, Gamaleldin M, Ghallab O. The clinical and prognostic significance of FIS1, SPI1, PDCD7 and Ang2 expression levels in acute myeloid leukemia. Cancer Genet 2018; 233-234:84-95. [PMID: 30555023 DOI: 10.1016/j.cancergen.2018.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/12/2018] [Accepted: 12/03/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVES The marked heterogeneity of acute myeloid leukemia (AML) renders precisely predicting patient prognosis extremely difficult. Genetic alterations, fusions and mutations, may result in misexpression of key genes in AML. We aimed to investigate the expression patterns of 4 novel genes; FIS1, SPI1, PDCD7 and Ang2 to determine their potential prognostic role in AML patients. METHODS Bone marrow mononuclear cells were analyzed for of FIS1, SPI1, PDCD7 and Ang2 expression levels by real-time quantitative PCR as well as of FLT3/ITD and NPM1 mutations in 100 newly diagnosed cytogenetically normal (CN-AML) patients, and 100 non-malignant controls. RESULTS FIS1, SPI1, PDCD7 and Ang2 were significantly overexpressed in CN-AML patients (p < 0.001). Their high expression levels were significantly associated with lower complete remission (CR) rate, shorter relapse-free survival (RFS) and overall survival (OS). On multivariate analysis, high FIS1 expression showed a significant impact on CR response after induction therapy (OR = 88.777, 95% C.I: 2.85-2765.78, p = 0.011) while high PDCD7 appeared to be an independent risk factor for RFS (HR = 5.107, 95% C.I: 1.731-15.066, p = 0.003) and OS (HR = 7.353, 95% C.I: 1.859-29.079, p = 0.004) in CN-AML patients. CONCLUSIONS FIS1 and PDCD7 expression are considered independent risk factors and should be integrated into the current AML stratification system.
Collapse
Affiliation(s)
- Reham Abo Elwafa
- Clinical Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Marwa Gamaleldin
- Clinical Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Omar Ghallab
- Internal Medicine Department (Hematology Unit), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
31
|
Ramzi M, Khalafi-Nezhad A, Iravani Saadi M, Jowkar Z. Association between TLR2 and TLR4 Expression and Response to Induction Therapy in Acute Myeloid Leukemia Patients. Int J Hematol Oncol Stem Cell Res 2018; 12:303-312. [PMID: 30774831 PMCID: PMC6375370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background: Toll-like receptors (TLRs) are a family of transmembrane pattern-recognition receptors that play a crucial role in the realization of innate and adaptive immune response. TLRs may play a role in tumor development and growth because of expression or up-regulation of functional TLRs in some tumors and tumor cell lines. The participation of TLRs in the pathogenesis of acute myeloid leukemia (AML) remains unspecified. This study aimed to investigate the effect of TLR2 and TLR4 expression in peripheral blood mononuclear cells of AML patients in response to induction chemotherapy. Materials and Methods: Eighty- five patients with newly diagnosed AML were evaluated. Using quantitative reverse transcriptase PCR, the mRNA expression of genes TLR2 and TLR4 was measured before starting and after induction chemotherapy. The differences in the mean expression levels of TLR2 and TLR4 before and after chemotherapy were compared using a paired t-test. The mean expression levels of TLR2 and TLR4 regarding laboratory data were analyzed by one-way ANOVA and Chi-square test. Results: We found that the mRNA expression of TLR2 after induction chemotherapy was significantly lower as compared to before treatment (p=0.001). Also, we found a lower TLR4 gene expression level after chemotherapy as compared to before chemotherapy, albeit it was not statistically significant (p=0.21). Moreover, we observed significantly higher expression of TLR2 and TLR4 in AML-M3 cases compared to non-M3 AML patients. Conclusion: The decreased expression of TLR4 in leukemic samples after induction chemotherapy might indicate a novel potential prognostic role for this receptor, particularly in AML-M3 cases.
Collapse
Affiliation(s)
- Mani Ramzi
- Hematology Research Center, Department of Hematology, Medical Oncology and Stem Cell Transplantation, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abolfazl Khalafi-Nezhad
- Hematology Research Center, Department of Hematology, Medical Oncology and Stem Cell Transplantation, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Zahra Jowkar
- Department of Operative Dentistry, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
32
|
JMJD3 facilitates C/EBPβ-centered transcriptional program to exert oncorepressor activity in AML. Nat Commun 2018; 9:3369. [PMID: 30135572 PMCID: PMC6105679 DOI: 10.1038/s41467-018-05548-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 06/29/2018] [Indexed: 12/21/2022] Open
Abstract
JMJD3, a stress-inducible H3K27 demethylase, plays a critical regulatory role in the initiation and progression of malignant hematopoiesis. However, how this histone modifier affects in a cell type-dependent manner remains unclear. Here, we show that in contrast to its oncogenic effect in preleukemia state and lymphoid malignancies, JMJD3 relieves the differentiation-arrest of certain subtypes (such as M2 and M3) of acute myeloid leukemia (AML) cells. RNA sequencing and ChIP−PCR analyses revealed that JMJD3 exerts anti-AML effect by directly modulating H3K4 and H3K27 methylation levels to activate the expression of a number of key myelopoietic regulatory genes. Mechanistic exploration identified a physical and functional association of JMJD3 with C/EBPβ that presides the regulatory network of JMJD3. Thus, the leukemia regulatory role of JMJD3 varies in a disease phase- and lineage-dependent manner, and acts as a potential oncorepressor in certain subsets of AML largely by coupling to C/EBPβ-centered myelopoietic program. Histone demethylase JMJD3 is known to be oncogenic in preleukemic states and T-cell acute lymphocytic leukemia. Here, the authors show that in some acute myeloid leukemia subsets, JMJD3 can actually act as a potential oncorepressor via mediation of C/EBPβ-centered transcriptional programming.
Collapse
|
33
|
Roulston GDR, Burt CL, Kettyle LMJ, Matchett KB, Keenan HL, Mulgrew NM, Ramsey JM, Dougan C, McKiernan J, Grishagin IV, Mills KI, Thompson A. Low-dose salinomycin induces anti-leukemic responses in AML and MLL. Oncotarget 2018; 7:73448-73461. [PMID: 27612428 PMCID: PMC5341990 DOI: 10.18632/oncotarget.11866] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/15/2016] [Indexed: 11/25/2022] Open
Abstract
Development of anti-cancer drugs towards clinical application is costly and inefficient. Large screens of drugs, efficacious for non-cancer disease, are currently being used to identify candidates for repurposing based on their anti-cancer properties. Here, we show that low-dose salinomycin, a coccidiostat ionophore previously identified in a breast cancer screen, has anti-leukemic efficacy. AML and MLLr cell lines, primary cells and patient samples were sensitive to submicromolar salinomycin. Most strikingly, colony formation of normal hematopoietic cells was unaffected by salinomycin, demonstrating a lack of hemotoxicity at the effective concentrations. Furthermore, salinomycin treatment of primary cells resulted in loss of leukemia repopulation ability following transplantation, as demonstrated by extended recipient survival compared to controls. Bioinformatic analysis of a 17-gene signature identified and validated in primary MLLr cells, uncovered immunomodulatory pathways, hubs and protein interactions as potential transducers of low dose salinomycin treatment. Additionally, increased protein expression of p62/Sqstm1, encoded for by one of the 17 signature genes, demonstrates a role for salinomycin in aggresome/vesicle formation indicative of an autophagic response. Together, the data support the efficacy of salinomycin as an anti-leukemic at non-hemotoxic concentrations. Further investigation alone or in combination with other therapies is warranted for future clinical trial.
Collapse
Affiliation(s)
- Gary D R Roulston
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Charlotte L Burt
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Laura M J Kettyle
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Kyle B Matchett
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Heather L Keenan
- Cambridge University School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, CB2 0SP, United Kingdom
| | - Nuala M Mulgrew
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Joanne M Ramsey
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Caoifa Dougan
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - John McKiernan
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Ivan V Grishagin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Ken I Mills
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Alexander Thompson
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland, United Kingdom.,Division of Cancer and Stem Cells, School of Medicine, Wolfson Centre for Stem Cells, Tissue Engineering & Modelling (STEM), University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| |
Collapse
|
34
|
Villamón E, González-Fernández J, Such E, Cervera JV, Gozalbo D, Luisa Gil M. Imiquimod inhibits growth and induces differentiation of myeloid leukemia cell lines. Cancer Cell Int 2018; 18:15. [PMID: 29422777 PMCID: PMC5791367 DOI: 10.1186/s12935-018-0515-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/24/2018] [Indexed: 12/13/2022] Open
Abstract
Background The antitumoral effects of different Toll-like receptor (TLRs) agonists is mediated by activating immune responses to suppress tumors growth, although TLR ligands may also have a direct effect on tumoral cells. Given that TLR signaling induces hematopoietic cell differentiations this may serve as a novel differentiation therapeutic approach for AML. Methods We investigated the effects of agonists for the ten human TLRs on the proliferation, apoptosis, cell cycle and differentiation of ten different types of myeloid leukemia cell lines (HL-60, U-937, KG-1, KG-1a, K-562, Kasumi-1, EOL-1, NB4, MOLM-13 and HEL). Proliferation was measured using the CellTiter 96® Aqueous One Solution Cell Proliferation Assay (Promega). Staining and analysis with a flow cytometer was used to identify cell cycle progression and apoptosis. Differentiation was measured by staining cells with the EuroFlow™ antibody panel for AML and analyzed by flow cytometry. FlowJo software was used to analyze the cytometric data. In all experiments, statistical significance was determined by a two-tailed t test. Results The activation of particular TLRs on some cell lines can induce growth inhibition and Imiquimod (a TLR 7 agonist) was the most effective agonist in all leukemic cell lines examined. Imiquimod was able to induce apoptosis, as well as to induce cell cycle alteration and upregulation of myeloid differentiation markers on some of the cell lines tested. Conclusions Our results, together with the known efficacy of Imiquimod against many tumor entities, suggest that Imiquimod can be a potential alternative therapy to AML. This drug has a direct cytotoxic effect on leukemic cells, has the potential to induce differentiation, and can also stimulate the activation of cellular immune responses anti-AML.
Collapse
Affiliation(s)
- Eva Villamón
- 1Department of Hematology, University Hospital La Fe, Valencia, Spain
| | | | - Esperanza Such
- 1Department of Hematology, University Hospital La Fe, Valencia, Spain
| | | | - Daniel Gozalbo
- 2Departamento de Microbiología y Ecología, Universitat de València, Burjasot, Spain.,3Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Burjasot, Spain
| | - M Luisa Gil
- 2Departamento de Microbiología y Ecología, Universitat de València, Burjasot, Spain.,3Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Burjasot, Spain
| |
Collapse
|
35
|
Pei R, Si T, Lu Y, Zhou JX, Jiang L. Salvianolic acid A, a novel PI3K/Akt inhibitor, induces cell apoptosis and suppresses tumor growth in acute myeloid leukemia. Leuk Lymphoma 2017; 59:1959-1967. [PMID: 29164984 DOI: 10.1080/10428194.2017.1399314] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Salvianolic acid A (SAA), one of the main derivatives of Salvia miltiorrhiza, has been shown to possess anti-inflammatory and anti-thrombotic activities. Its role in inhibiting tumor growth, however, remains elusive. The aim of this study was to investigate the effect of SAA on acute myeloid leukemia (AML). Here, SAA showed a dose-dependent cell viability inhibition and apoptosis induction in AML cells. At the molecular level, SAA increased the expression of Bak and decreased the expression of Bcl-xL, following by PARP cleavage and caspase-3 activation. SAA also markedly attenuated Akt phosphorylation in AML cells. In a xenograft mouse model, SAA significantly suppressed the growth of AML tumors in vivo. Furthermore, SAA exhibited a more profound pro-apoptotic effect on primary AML cells than on bone marrow mononuclear cells from patients with benign diseases. Therefore, the pro-apoptotic and anti-tumor properties of SAA suggested its promising therapeutic value for AML.
Collapse
Affiliation(s)
- Renzhi Pei
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China.,b Department of Hematology , Yinzhou People's Hospital, Ningbo University School of Medicine , Ningbo , China
| | - Ting Si
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China.,b Department of Hematology , Yinzhou People's Hospital, Ningbo University School of Medicine , Ningbo , China
| | - Ying Lu
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China.,b Department of Hematology , Yinzhou People's Hospital, Ningbo University School of Medicine , Ningbo , China
| | - Jeff Xiwu Zhou
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China
| | - Lei Jiang
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China
| |
Collapse
|
36
|
Hemmati S, Haque T, Gritsman K. Inflammatory Signaling Pathways in Preleukemic and Leukemic Stem Cells. Front Oncol 2017; 7:265. [PMID: 29181334 PMCID: PMC5693908 DOI: 10.3389/fonc.2017.00265] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/20/2017] [Indexed: 12/15/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are a rare subset of bone marrow cells that usually exist in a quiescent state, only entering the cell cycle to replenish the blood compartment, thereby limiting the potential for errors in replication. Inflammatory signals that are released in response to environmental stressors, such as infection, trigger active cycling of HSCs. These inflammatory signals can also directly induce HSCs to release cytokines into the bone marrow environment, promoting myeloid differentiation. After stress myelopoiesis is triggered, HSCs require intracellular signaling programs to deactivate this response and return to steady state. Prolonged or excessive exposure to inflammatory cytokines, such as in prolonged infection or in chronic rheumatologic conditions, can lead to continued HSC cycling and eventual HSC loss. This promotes bone marrow failure, and can precipitate preleukemic states or leukemia through the acquisition of genetic and epigenetic changes in HSCs. This can occur through the initiation of clonal hematopoiesis, followed by the emergence preleukemic stem cells (pre-LSCs). In this review, we describe the roles of multiple inflammatory signaling pathways in the generation of pre-LSCs and in progression to myelodysplastic syndrome (MDS), myeloproliferative neoplasms, and acute myeloid leukemia (AML). In AML, activation of some inflammatory signaling pathways can promote the cycling and differentiation of LSCs, and this can be exploited therapeutically. We also discuss the therapeutic potential of modulating inflammatory signaling for the treatment of myeloid malignancies.
Collapse
Affiliation(s)
- Shayda Hemmati
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Tamanna Haque
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Oncology, Montefiore Medical Center, Bronx, NY, United States
| | - Kira Gritsman
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Oncology, Montefiore Medical Center, Bronx, NY, United States
| |
Collapse
|
37
|
Eriksson M, Peña-Martínez P, Ramakrishnan R, Chapellier M, Högberg C, Glowacki G, Orsmark-Pietras C, Velasco-Hernández T, Lazarević VL, Juliusson G, Cammenga J, Mulloy JC, Richter J, Fioretos T, Ebert BL, Järås M. Agonistic targeting of TLR1/TLR2 induces p38 MAPK-dependent apoptosis and NFκB-dependent differentiation of AML cells. Blood Adv 2017; 1:2046-2057. [PMID: 29296851 PMCID: PMC5728277 DOI: 10.1182/bloodadvances.2017006148] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 09/18/2017] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is associated with poor survival, and there is a strong need to identify disease vulnerabilities that might reveal new treatment opportunities. Here, we found that Toll-like receptor 1 (TLR1) and TLR2 are upregulated on primary AML CD34+CD38- cells relative to corresponding normal bone marrow cells. Activating the TLR1/TLR2 complex by the agonist Pam3CSK4 in MLL-AF9-driven human AML resulted in induction of apoptosis by p38 MAPK-dependent activation of Caspase 3 and myeloid differentiation in a NFκB-dependent manner. By using murine Trp53-/-MLL-AF9 AML cells, we demonstrate that p53 is dispensable for Pam3CSK4-induced apoptosis and differentiation. Moreover, murine AML1-ETO9a-driven AML cells also were forced into apoptosis and differentiation on TLR1/TLR2 activation, demonstrating that the antileukemic effects observed were not confined to MLL-rearranged AML. We further evaluated whether Pam3CSK4 would exhibit selective antileukemic effects. Ex vivo Pam3CSK4 treatment inhibited murine and human leukemia-initiating cells, whereas murine normal hematopoietic stem and progenitor cells (HSPCs) were relatively less affected. Consistent with these findings, primary human AML cells across several genetic subtypes of AML were more vulnerable for TLR1/TLR2 activation relative to normal human HSPCs. In the MLL-AF9 AML mouse model, treatment with Pam3CSK4 provided proof of concept for in vivo therapeutic efficacy. Our results demonstrate that TLR1 and TLR2 are upregulated on primitive AML cells and that agonistic targeting of TLR1/TLR2 forces AML cells into apoptosis by p38 MAPK-dependent activation of Caspase 3, and differentiation by activating NFκB, thus revealing a new putative strategy for therapeutically targeting AML cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Vladimir Lj Lazarević
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Gunnar Juliusson
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Jörg Cammenga
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - James C Mulloy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH; and
| | - Johan Richter
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | | | - Benjamin L Ebert
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
38
|
Chen S, Yu J, Lv X, Zhang L. HOXA9 is critical in the proliferation, differentiation, and malignancy of leukaemia cells both in vitro and in vivo. Cell Biochem Funct 2017; 35:433-440. [PMID: 28961318 DOI: 10.1002/cbf.3293] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/02/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Shibing Chen
- Department of Hematology; Yishui Center Hospital; Linyi Shandong Province China
| | - Juan Yu
- Department of Neurosurgery; Yishui Center Hospital; Linyi Shandong Province China
| | - Xin Lv
- Department of Hematology; Yishui Center Hospital; Linyi Shandong Province China
| | - Lijuan Zhang
- Department of Cardiology; Yishui Center Hospital; Linyi Shandong Province China
| |
Collapse
|
39
|
Gao D, Li W, Wang W, Cai Y, Wang Y, Luo X, Wei CC. Synergy of purine-scaffold TLR7 agonist with doxorubicin on systemic inhibition of lymphoma in mouse model. J Cancer 2017; 8:3183-3189. [PMID: 29158790 PMCID: PMC5665034 DOI: 10.7150/jca.20015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 08/30/2017] [Indexed: 11/25/2022] Open
Abstract
Chemo- and radio-therapy suffer from certain well-recognized drawbacks for lymphoma therapy. Passive immunotherapy with monoclonal antibody has improved outcome for patients with CD20+ B cell lymphoma, but not for T cell lymphoma. Therefore, novel treatment approaches are clearly required for T cell lymphoma. To date, the combined application targeting TLR7, 8 and 9 has established long-term antitumor immunity. We previously synthesized a purine-scaffold TLR7 agonist named GD5. Here, we report that the intratumoral administration of GD5 combined with doxorubicin (DOX), a conventional chemotherapeutic agent in T cell lymphoma. This combined treatment made mice to produce more cytokines in blood, and generate more potent cytotoxic T lymphocyte response, then result in effective eradication of both local and distant tumors in tumor-bearing mice. Our findings demonstrate the potential for enhancing the efficacy of the current standard DOX therapy through combination with TLR7 agonist GD5 to improve antitumor immune responses and provide durable remissions for T cell lymphoma.
Collapse
Affiliation(s)
- Dong Gao
- Shenzhen Hornetcorn Biotechnology Co., Ltd., Shenzhen, 518045, China
| | - Wang Li
- Shenzhen Hornetcorn Biotechnology Co., Ltd., Shenzhen, 518045, China
| | - Wanmin Wang
- Dalian Jinma Medical Technology Co., Ltd., Dalian, 116620, China
| | - Yongguang Cai
- The Fifth District of Chemotherapy, Department of Medical Oncology, Central Hospital of Guangdong Provincial Agricultural Reclamation, Zhanjiang, 524002, China
| | - Yuhuan Wang
- Shenzhen Hornetcorn Biotechnology Co., Ltd., Shenzhen, 518045, China
| | - Xiaoling Luo
- Shenzhen Hornetcorn Biotechnology Co., Ltd., Shenzhen, 518045, China
| | - Chih-Chang Wei
- Shenzhen Hornetcorn Biotechnology Co., Ltd., Shenzhen, 518045, China
| |
Collapse
|
40
|
Mesenchymal Stem Cells in Myeloid Malignancies: A Focus on Immune Escaping and Therapeutic Implications. Stem Cells Int 2017; 2017:6720594. [PMID: 28947904 PMCID: PMC5602646 DOI: 10.1155/2017/6720594] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/06/2017] [Accepted: 07/20/2017] [Indexed: 01/07/2023] Open
Abstract
The importance of the bone marrow microenvironment forming the so-called niche in physiologic hemopoiesis is largely known, and recent evidences support the presence of stromal alterations from the molecular to the cytoarchitectural level in hematologic malignancies. Various alterations in cell adhesion, metabolism, cytokine signaling, autophagy, and methylation patterns of tumor-derived mesenchymal stem cells have been demonstrated, contributing to the genesis of a leukemic permissive niche. This niche allows both the ineffective haematopoiesis typical of myelodysplastic syndromes and the differentiation arrest, proliferation advantage, and clone selection which is the hallmark of acute myeloid leukemia. Furthermore, the immune system, both adaptive and innate, encompassing mesenchymal-derived cells, has been shown to take part to the leukemic niche. Here, we critically review the state of art about mesenchymal stem cell role in myelodysplastic syndromes and acute myeloid leukemia, focusing on immune escaping mechanisms as a target for available and future anticancer therapies.
Collapse
|
41
|
Shekarian T, Valsesia-Wittmann S, Brody J, Michallet M, Depil S, Caux C, Marabelle A. Pattern recognition receptors: immune targets to enhance cancer immunotherapy. Ann Oncol 2017; 28:1756-1766. [DOI: 10.1093/annonc/mdx179] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
42
|
Chi H, Li C, Zhao FS, Zhang L, Ng TB, Jin G, Sha O. Anti-tumor Activity of Toll-Like Receptor 7 Agonists. Front Pharmacol 2017; 8:304. [PMID: 28620298 PMCID: PMC5450331 DOI: 10.3389/fphar.2017.00304] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/10/2017] [Indexed: 01/04/2023] Open
Abstract
Toll-like receptors (TLRs) are a class of pattern recognition receptors that play a bridging role in innate immunity and adaptive immunity. The activated TLRs not only induce inflammatory responses, but also elicit the development of antigen specific immunity. TLR7, a member of TLR family, is an intracellular receptor expressed on the membrane of endosomes. TLR7 can be triggered not only by ssRNA during viral infections, but also by immune modifiers that share a similar structure to nucleosides. Its powerful immune stimulatory action can be potentially used in the anti-tumor therapy. This article reviewed the anti-tumor activity and mechanism of TLR7 agonists that are frequently applied in preclinical and clinical investigations, and mainly focused on small synthetic molecules, including imiquimod, resiquimod, gardiquimod, and 852A, etc.
Collapse
Affiliation(s)
- Huju Chi
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science CentreShenzhen, China
| | - Chunman Li
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science CentreShenzhen, China
| | - Flora Sha Zhao
- School of Life Sciences, Faculty of Science, The Chinese University of Hong KongHong Kong, Hong Kong
| | - Li Zhang
- Department of Physiology and Neurology, University of ConnecticutStorrs, CT, United States
| | - Tzi Bun Ng
- Departmet of Biochemistry, Faculty of Science, The Chinese University of Hong KongHong Kong, Hong Kong
| | - Guangyi Jin
- Department of Pharmacy, Shenzhen University Health Science CentreShenzhen, China
| | - Ou Sha
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science CentreShenzhen, China
| |
Collapse
|
43
|
S100A9 induces differentiation of acute myeloid leukemia cells through TLR4. Blood 2017; 129:1980-1990. [PMID: 28137827 DOI: 10.1182/blood-2016-09-738005] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/20/2017] [Indexed: 12/22/2022] Open
Abstract
S100A8 and S100A9 are calcium-binding proteins predominantly expressed by neutrophils and monocytes and play key roles in both normal and pathological inflammation. Recently, both proteins were found to promote tumor progression through the establishment of premetastatic niches and inhibit antitumor immune responses. Although S100A8 and S100A9 have been studied in solid cancers, their functions in hematological malignancies remain poorly understood. However, S100A8 and S100A9 are highly expressed in acute myeloid leukemia (AML), and S100A8 expression has been linked to poor prognosis in AML. We identified a small subpopulation of cells expressing S100A8 and S100A9 in AML mouse models and primary human AML samples. In vitro and in vivo analyses revealed that S100A9 induces AML cell differentiation, whereas S100A8 prevents differentiation induced by S100A9 activity and maintains AML immature phenotype. Treatment with recombinant S100A9 proteins increased AML cell maturation, induced growth arrest, and prolonged survival in an AML mouse model. Interestingly, anti-S100A8 antibody treatment had effects similar to those of S100A9 therapy in vivo, suggesting that high ratios of S100A9 over S100A8 are required to induce differentiation. Our in vitro studies on the mechanisms/pathways involved in leukemic cell differentiation revealed that binding of S100A9 to Toll-like receptor 4 (TLR4) promotes activation of p38 mitogen-activated protein kinase, extracellular signal-regulated kinases 1 and 2, and Jun N-terminal kinase signaling pathways, leading to myelomonocytic and monocytic AML cell differentiation. These findings indicate that S100A8 and S100A9 are regulators of myeloid differentiation in leukemia and have therapeutic potential in myelomonocytic and monocytic AMLs.
Collapse
|
44
|
Monlish DA, Bhatt ST, Schuettpelz LG. The Role of Toll-Like Receptors in Hematopoietic Malignancies. Front Immunol 2016; 7:390. [PMID: 27733853 PMCID: PMC5039188 DOI: 10.3389/fimmu.2016.00390] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/15/2016] [Indexed: 12/02/2022] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors that shape the innate immune system by identifying pathogen-associated molecular patterns and host-derived damage-associated molecular patterns. TLRs are widely expressed on both immune cells and non-immune cells, including hematopoietic stem and progenitor cells, effector immune cell populations, and endothelial cells. In addition to their well-known role in the innate immune response to acute infection or injury, accumulating evidence supports a role for TLRs in the development of hematopoietic and other malignancies. Several hematopoietic disorders, including lymphoproliferative disorders and myelodysplastic syndromes, which possess a high risk of transformation to leukemia, have been linked to aberrant TLR signaling. Furthermore, activation of TLRs leads to the induction of a number of proinflammatory cytokines and chemokines, which can promote tumorigenesis by driving cell proliferation and migration and providing a favorable microenvironment for tumor cells. Beyond hematopoietic malignancies, the upregulation of a number of TLRs has been linked to promoting tumor cell survival, proliferation, and metastasis in a variety of cancers, including those of the colon, breast, and lung. This review focuses on the contribution of TLRs to hematopoietic malignancies, highlighting the known direct and indirect effects of TLR signaling on tumor cells and their microenvironment. In addition, the utility of TLR agonists and antagonists as potential therapeutics in the treatment of hematopoietic malignancies is discussed.
Collapse
Affiliation(s)
- Darlene A Monlish
- Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| | - Sima T Bhatt
- Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| | - Laura G Schuettpelz
- Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| |
Collapse
|
45
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
46
|
Wiedemann GM, Jacobi SJ, Chaloupka M, Krächan A, Hamm S, Strobl S, Baumgartner R, Rothenfusser S, Duewell P, Endres S, Kobold S. A novel TLR7 agonist reverses NK cell anergy and cures RMA-S lymphoma-bearing mice. Oncoimmunology 2016; 5:e1189051. [PMID: 27622045 DOI: 10.1080/2162402x.2016.1189051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/26/2016] [Accepted: 05/08/2016] [Indexed: 12/30/2022] Open
Abstract
Toll-like receptor 7 (TLR7) agonists are potent immune stimulants able to overcome cancer-associated immune suppression. Due to dose-limiting systemic toxicities, only the topically applied TLR7 agonist (imiquimod) has been approved for therapy of skin tumors. There is a need for TLR7-activating compounds with equivalent efficacy but less toxicity. SC1, a novel small molecule agonist for TLR7, is a potent type-1 interferon inducer, comparable to the reference TLR7 agonist resiquimod, yet with lower induction of proinflammatory cytokines. In vivo, SC1 activates NK cells in a TLR7-dependent manner. Mice bearing the NK cell-sensitive lymphoma RMA-S are cured by repeated s. c. administrations of SC1 as efficiently as by the administration of resiquimod. No relevant toxicities were observed. Mechanistically, SC1 reverses NK cell anergy and restores NK cell-mediated tumor cell killing in an IFN-α-dependent manner. TLR7 targeting by SC1-based compounds may form an attractive strategy to activate NK cell responses for cancer therapy.
Collapse
Affiliation(s)
- Gabriela Maria Wiedemann
- Center of Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich, Germany
| | - Severin Johannes Jacobi
- Center of Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich, Germany
| | - Michael Chaloupka
- Center of Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich, Germany
| | - Angelina Krächan
- Center of Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich, Germany
| | | | | | | | - Simon Rothenfusser
- Center of Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich, Germany
| | - Peter Duewell
- Center of Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPSM) and Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich, Germany
| |
Collapse
|
47
|
Alvarado AG, Lathia JD. Taking a Toll on Self-Renewal: TLR-Mediated Innate Immune Signaling in Stem Cells. Trends Neurosci 2016; 39:463-471. [PMID: 27155992 DOI: 10.1016/j.tins.2016.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023]
Abstract
Innate immunity has evolved as the front-line cellular defense mechanism to acutely sense and decisively respond to microenvironmental alterations. The Toll-like receptor (TLR) family activates signaling pathways in response to stimuli and is well-characterized in both resident and infiltrating immune cells during neural inflammation, injury, and degeneration. Innate immune signaling has also been observed in neural cells during development and disease, including in the stem and progenitor cells that build the brain and are responsible for its homeostasis. Recently, the activation of developmental programs in malignant brain tumors has emerged as a driver for growth via cancer stem cells. In this review we discuss how innate immune signaling interfaces with stem cell maintenance in the normal and neoplastic brain.
Collapse
Affiliation(s)
- Alvaro G Alvarado
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA; Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
48
|
Yuan XQ, Peng L, Zeng WJ, Jiang BY, Li GC, Chen XP. DNMT3A R882 Mutations Predict a Poor Prognosis in AML: A Meta-Analysis From 4474 Patients. Medicine (Baltimore) 2016; 95:e3519. [PMID: 27149454 PMCID: PMC4863771 DOI: 10.1097/md.0000000000003519] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
DNA (cytosine-5)-methyltransferase 3 alpha (DNMT3A) mutations were widely believed to be independently associated with inferior prognosis in acute myeloid leukemia (AML) patients. As dominant missense alterations in DNMT3A mutations, R882 mutations cause the focal hypomethylation phenotype. However, there remains debate on the influence of R882 mutations on AML prognosis. Thus, this meta-analysis aimed at further illustrating the prognostic power of DNMT3A R882 mutations in AML patients.Eligible studies were identified from 5 databases containing PubMed, Embase, Web of Science, Clinical Trials, and the Cochrane Library (up to October 25, 2015). Effects (hazard ratios [HRs] with 95% confidence interval [CI]) of relapse-free survival (RFS) and overall survival (OS) were pooled to estimate the prognostic power of mutant DNMT3A R882 in overall patients and subgroups of AML patients.Eight competent studies with 4474 AML patients including 694 with DNMT3A R882 mutations were included. AML patients with DNMT3A R882 mutations showed significant shorter RFS (HR = 1.40, 95% CI = 1.24-1.59, P < 0.001) and OS (HR = 1.47, 95% CI = 1.17-1.86, P = 0.001) in the overall population. DNMT3A R882 mutations predicted worse RFS and OS among the subgroups of patients under age 60 (RFS: HR = 1.44, 95% CI = 1.25-1.66, P < 0.001; OS: HR = 1.48, 95% CI = 1.15-1.90, P = 0.002), over age 60 (RFS: HR = 2.03, 95% CI = 1.40-2.93, P < 0.001; OS: HR = 1.85, 95% CI = 1.36-2.53, P < 0.001), cytogenetically normal (CN)-AML (RFS: HR = 1.52, 95% CI = 1.26-1.83, P < 0.001; OS: HR = 1.67, 95% CI = 1.16-2.41, P = 0.006), and non-CN-AML (RFS: HR = 1.96, 95% CI = 1.20-3.21, P = 0.006; OS: HR = 2.51, 95% CI = 1.52-4.15, P = 0.0038).DNMT3A R882 mutations possessed significant unfavorable prognostic influence on RFS and OS in AML patients.
Collapse
Affiliation(s)
- Xiao-Qing Yuan
- From the Department of Clinical Pharmacology, Xiangya Hospital; Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics (X-QY, W-JZ, X-PC); Cancer Research Institute, Central South University; Key Laboratory of Carcinogenesis, National Health and Family Planning Commission; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha (LP, B-YJ, G-CL); and Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, P.R. China (X-PC)
| | | | | | | | | | | |
Collapse
|
49
|
Toll-like receptor signaling in hematopoietic homeostasis and the pathogenesis of hematologic diseases. Front Med 2015; 9:288-303. [DOI: 10.1007/s11684-015-0412-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/10/2015] [Indexed: 02/07/2023]
|
50
|
Zhu J, He S, Du J, Wang Z, Li W, Chen X, Jiang W, Zheng D, Jin G. Local administration of a novel Toll-like receptor 7 agonist in combination with doxorubicin induces durable tumouricidal effects in a murine model of T cell lymphoma. J Hematol Oncol 2015; 8:21. [PMID: 25887995 PMCID: PMC4359787 DOI: 10.1186/s13045-015-0121-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/11/2015] [Indexed: 01/19/2023] Open
Abstract
Background Conventional chemotherapy and radiotherapy for the treatment of lymphoma have notable drawbacks, and passive immunotherapy using a monoclonal antibody is restricted to CD20-positive B cell lymphoma. Therefore, new treatment types are urgently required, especially for T cell lymphoma. One type of new antitumour therapy is the use of active immunotherapeutic agents, such as agonists of the Toll-like receptors (TLRs), which facilitate the induction of prolonged antitumour immune responses. Methods We have synthesised a novel TLR7 agonist called SZU-101 and investigated the systemic antitumour effect on a murine model of T cell lymphoma in vivo. Results Here, we report that the intratumoural administration of SZU-101 enhanced the effectiveness of a conventionally used chemotherapeutic agent, doxorubicin (DOX). SZU-101 administration improved tumour clearance in a murine model of T cell lymphoma. The novel combination of intratumourally administered SZU-101 and DOX generated strong cytokine production and enhanced the cytotoxic T lymphocyte response, leading to the eradication of both local and distant tumours in tumour-bearing mice. Conclusions These findings suggested that combined active immunotherapy can be developed as a promising treatment for T cell lymphoma, which may further improve the effectiveness of the current standard cyclophosphamide, DOX, vincristine and prednisone (CHOP) therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0121-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jiang Zhu
- Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China. .,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dong Feng RD East, Guangzhou, 510060, People's Republic of China.
| | - Shiping He
- Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China.
| | - Jie Du
- Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China.
| | - Zhulin Wang
- Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China. .,Shenzhen Engineering Lab of Synthetic Biology, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China.
| | - Wang Li
- Shenzhen Engineering Lab of Synthetic Biology, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China.
| | - Xianxiong Chen
- Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China.
| | - Wenqi Jiang
- Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China. .,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dong Feng RD East, Guangzhou, 510060, People's Republic of China.
| | - Duo Zheng
- Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China. .,Shenzhen Engineering Lab of Synthetic Biology, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China.
| | - Guangyi Jin
- Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China. .,Shenzhen Engineering Lab of Synthetic Biology, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, People's Republic of China.
| |
Collapse
|