1
|
Dasgupta A, May JN, Klinkenberg G, Besse HC, Buhl EM, Moeckel D, Mihyar R, Peña Q, Shalmani AA, Hark C, Rix A, Koletnik S, Metselaar J, Shi Y, Hennink WE, Storm G, van Vuurden D, Moonen C, Ries M, Schmid R, Kiessling F, Lammers T. Multidrug micelles and sonopermeation for chemotherapy co-delivery to brain tumors. J Control Release 2025; 380:818-828. [PMID: 39956394 DOI: 10.1016/j.jconrel.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 02/18/2025]
Abstract
Brain tumors are difficult to target and treat. The blood-brain barrier (BBB) limits drug delivery to pathological sites, and standard mono-chemotherapy typically results in suboptimal efficacy and development of drug resistance. We here set out to load a synergistic drug combination in polymeric micelles, and combined them with ultrasound- and microbubble-mediated BBB opening in glioma models in mice. Via high-throughput screening of various chemotherapy combinations in different glioma cell lines, valrubicin and panobinostat were identified as a synergic drug combination and co-loaded in mPEG-b-p(HPMAm-Bz)-based polymeric micelles. Intravenous administration of double-drug micelles showed good tolerability and resulted in significant tumor growth inhibition in mice with subcutaneous GL261 gliomas. In orthotopically inoculated patient-derived HSJD-DIPG-007 diffuse intrinsic pontine gliomas, notoriously known to have an intact BBB and poor drug responsiveness, we provide initial experimental evidence showing that multidrug micelles plus sonopermeation can help to improve treatment efficacy. Our work exemplifies that synergistic drug combinations can be efficiently co-loaded in polymeric micelles, and that advanced nanosonochemotherapy combination regimens hold promise for the treatment of hard-to-treat brain tumors.
Collapse
Affiliation(s)
- Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Jan-Niklas May
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Geir Klinkenberg
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Helena C Besse
- Center for Imaging Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Diana Moeckel
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Rahaf Mihyar
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Quim Peña
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | | | - Christopher Hark
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Susanne Koletnik
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Josbert Metselaar
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Yang Shi
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht University, 3584 CG Utrecht, the Netherlands; Department of Surgery, National University of Singapore, 119228 Singapore, Singapore
| | | | - Chrit Moonen
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Mario Ries
- Center for Imaging Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ruth Schmid
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen, Aachen, Germany.
| |
Collapse
|
2
|
Zrimšek M, Draganić K, Malzer A, Doblmayr V, Mišura K, de Freitas E Silva R, Matthews JD, Iannelli F, Wohlhaupter S, Pérez Malla CU, Fischer H, Schachner H, Schiefer AI, Sheibani-Tezerji R, Chiarle R, Turner SD, Ellmeier W, Seiser C, Egger G. HDAC1 acts as a tumor suppressor in ALK-positive anaplastic large cell lymphoma: implications for HDAC inhibitor therapy. Leukemia 2025:10.1038/s41375-025-02584-9. [PMID: 40175628 DOI: 10.1038/s41375-025-02584-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 03/03/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025]
Abstract
Histone deacetylases (HDACs) are frequently deregulated in cancer, and several HDAC inhibitors (HDACi) have gained approval for treating peripheral T cell lymphomas. Here, we investigated the effects of pharmacological or genetic HDAC inhibition on NPM::ALK positive anaplastic large cell lymphoma (ALCL) development to assess the potential use of HDACi for the treatment of this disease. Short-term systemic pharmacological inhibition of HDACs using the HDACi Entinostat in a premalignant ALCL mouse model postponed or even abolished lymphoma development, despite high expression of the NPM::ALK fusion oncogene. To further disentangle the effects of systemic HDAC inhibition from thymocyte intrinsic effects, conditional genetic deletions of HDAC1 and HDAC2 enzymes were employed. In sharp contrast, T cell-specific deletion of Hdac1 or Hdac2 in the ALCL mouse model significantly accelerated NPM::ALK-driven lymphomagenesis, with Hdac1 loss having a more pronounced effect. Integration of gene expression and chromatin accessibility data revealed that Hdac1 deletion selectively perturbed cell type-specific transcriptional programs, crucial for T cell differentiation and signaling. Moreover, multiple oncogenic signaling pathways, including PDGFRB signaling, were highly upregulated. Our findings underscore the tumor-suppressive function of HDAC1 and HDAC2 in T cells during ALCL development. Nevertheless, systemic pharmacological inhibition of HDACs could still potentially improve current therapeutic outcomes.
Collapse
Affiliation(s)
- Maša Zrimšek
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Kristina Draganić
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Anna Malzer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Verena Doblmayr
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Katarina Mišura
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Rafael de Freitas E Silva
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Fabio Iannelli
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | | | - Carlos Uziel Pérez Malla
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Heinz Fischer
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Helga Schachner
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Ana-Iris Schiefer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Raheleh Sheibani-Tezerji
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Roberto Chiarle
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suzanne Dawn Turner
- Department of Pathology, University of Cambridge, Cambridge, UK
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Christian Seiser
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria.
| |
Collapse
|
3
|
Yin T, Liu Y, Li C, Feng X, Lin Y, Qu Z. Characteristic analysis of adverse reactions of histone deacetylase inhibitors based on WHO-VigiAccess. Front Pharmacol 2025; 16:1563797. [PMID: 40170733 PMCID: PMC11959061 DOI: 10.3389/fphar.2025.1563797] [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: 01/20/2025] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Background This study assessed the adverse drug reactions (ADRs) associated with HDAC inhibitors using the VigiAccess database maintained by the World Health Organization (WHO). Furthermore, it compared the ADR profiles of three different drugs to identify the one with the lowest individualized risk for patients. Materials and methods Data on adverse events of HDAC Inhibitors was retrieved from WHO-VigiAccess on 6 January 2025. We obtained data on age, gender, reporting year, and continent. Descriptive data related were calculated using Excel 2021. In this study, we used Excel software to analyze the characteristics of those who were harmed due to adverse reactions. For each drug, the reporting rate of adverse reactions was calculated by dividing the number of adverse reaction symptoms of this drug by the total number of adverse reaction reports. We listed the top 20 most frequent adverse reaction symptoms as common adverse reactions. By counting the frequency and proportion of these common adverse reactions, we conducted a comparative analysis of the adverse reaction situations of different drugs and classified them according to different types. Result The WHO-VigiAccess database received 796, 1254, and 1658 ADR reports for Chidamide, Romidepsin, and Vorinostat respectively by 2024, with a total of 3,708. Gender distribution was relatively balanced (male:female ratio 0.81:1), and the 45-64 age group had the highest reporting rates, mostly from the Americas. Chidamide had higher rates in certain disorders, Romidepsin in others, and Vorinostat in specific ones. Common ADRs included thrombocytopenia etc., with some differences in rates among drugs. Serious ADR proportions were 0% for Chidamide, 2.27% for Romidepsin, and 1.02% for Vorinostat. 37 common signals were found, with Investigations having the most. Each drug had different ADR preferred terms (PTs) in renal/urinary and metabolism/nutrition disorders, with varying numbers of distinctive symptoms. Conclusion Current comparative observational studies of these inhibitors indicate that there are both common and specific adverse reactions reported in the ADR data received by the WHO for these medications. Clinicians should enhance the rational use of these drugs by considering the characteristics of the reported ADRs.
Collapse
Affiliation(s)
- Tongnan Yin
- Central Laboratory, Nanyang Central Hospital, Nanyang, China
| | - Yuyu Liu
- School of Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenwen Li
- Department of Dermatology, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Xinran Feng
- Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi’an, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Qu
- Department of Oncology, Nanyang Central Hospital, Nanyang, China
| |
Collapse
|
4
|
Shi Z, Li M, Zhang C, Li H, Zhang Y, Zhang L, Li X, Li L, Wang X, Fu X, Sun Z, Zhang X, Tian L, Zhang M, Chen WH, Li Z. Butyrate-producing Faecalibacterium prausnitzii suppresses natural killer/T-cell lymphoma by dampening the JAK-STAT pathway. Gut 2025; 74:557-570. [PMID: 39653411 DOI: 10.1136/gutjnl-2024-333530] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Natural killer/T-cell lymphoma (NKTCL) is a highly aggressive malignancy with a dismal prognosis, and gaps remain in understanding the determinants influencing disease outcomes. OBJECTIVE To characterise the gut microbiota feature and identify potential probiotics that could ameliorate the development of NKTCL. DESIGN This cross-sectional study employed shotgun metagenomic sequencing to profile the gut microbiota in two Chinese NKTCL cohorts, with validation conducted in an independent Korean cohort. Univariable and multivariable Cox proportional hazards analyses were applied to assess associations between identified marker species and patient outcomes. Tumour-suppressing effects were investigated using comprehensive in vivo and in vitro models. In addition, metabolomics, RNA sequencing, chromatin immunoprecipitation sequencing, Western blot analysis, immunohistochemistry and lentiviral-mediated gene knockdown system were used to elucidate the underlying mechanisms. RESULTS We first unveiled significant gut microbiota dysbiosis in NKTCL patients, prominently marked by a notable reduction in Faecalibacterium prausnitzii which correlated strongly with shorter survival among patients. Subsequently, we substantiated the antitumour properties of F. prausnitzii in NKTCL mouse models. Furthermore, F. prausnitzii culture supernatant demonstrated significant efficacy in inhibiting NKTCL cell growth. Metabolomics analysis revealed butyrate as a critical metabolite underlying these tumour-suppressing effects, validated in three human NKTCL cell lines and multiple tumour-bearing mouse models. Mechanistically, butyrate suppressed the activation of Janus kinase-signal transducer and activator of transcription pathway through enhancing histone acetylation, promoting the expression of suppressor of cytokine signalling 1. CONCLUSION These findings uncover a distinctive gut microbiota profile in NKTCL and provide a novel perspective on leveraging the therapeutic potential of F. prausnitzii to ameliorate this malignancy.
Collapse
Affiliation(s)
- Zhuangzhuang Shi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Min Li
- Department of Bioinformatics and Systems Biology, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Zhang
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
- Chinese PLA General Hospital and Medical School, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongwen Li
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yue Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Lei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Xin Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Ling Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Xinhua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Xiaorui Fu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Zhenchang Sun
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Li Tian
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Wei-Hua Chen
- Department of Bioinformatics and Systems Biology, Huazhong University of Science and Technology, Wuhan, China
- School of Biological Science, Jining Medical University, Rizhao, Shandong, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Das T, Bhar S, Ghosh D, Kabi B, Kar K, Chandra A. A promising future for breast cancer therapy with hydroxamic acid-based histone deacetylase inhibitors. Bioorg Chem 2025; 156:108169. [PMID: 39862739 DOI: 10.1016/j.bioorg.2025.108169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Histone deacetylases (HDACs) play a critical role in chromatin remodelling and modulating the activity of various histone proteins. Aberrant HDAC functions has been related to the progression of breast cancer (BC), making HDAC inhibitors (HDACi) promising small-molecule therapeutics for its treatment. Hydroxamic acid (HA) is a significant pharmacophore due to its strong metal-chelating ability, HDAC inhibition properties, MMP inhibition abilities, and more. They were found to increase the efficacy of the approved drugs when used in combination. In this review we presented bioinformatic analysis using available data from the Cancer Genome Atlas and Genotype-Tissue Expression databases, outlined the recent advancements in the application of HA-based HDACi for BC during preclinical investigation and clinical trials, tried to offer the rationale for targeting HDAC in BC with HA-based HDACi, summarised the challenges faced in the successful clinical application of HDACi, and proposed potential strategies to address these challenges, aiming to enhance treatment outcomes in BC. Abbreviations: ABCG2, ATP-binding cassette super-family G member 2; ABC, ATP-binding cassette; ADP, Adenosine diphosphate; APC, Antigen presenting cell; AML, Acute myeloid leukemia; ARH1, Aplysia ras homolog 1; BCRP, Breast cancer resistance protein; BRCA, Breast invasive carcinoma; Bax, B-cell lymphoma associated X; CK5, Cytokeratin 5; CK14, Cytokeratin 14; CK17, Cytokeratin 17; CoRESTMiDAC, Co-repressor for element-1-silencing transcription factor; CRM1, Chromosomal maintenance 1; CTCL, Cutaneous T-cell lymphoma; DNMT, DNA methyltransferase; DFS, Disease-free survival; ER, Oestrogen receptor; EMT, Epithelial-mesenchymal transition; FGFR1, Fibroblast growth factor receptor 1; GEPIA, Gene Expression Profiling Interactive Analysis; GTEx, Genotype tissue expression; HAT, Histone acetylase; HDAC, Histone deacetylase; HDF, Human dermal fibroblast; HER2, Human epidermal growth factor receptor 2; HDLP, Histone deacetylase-like protein; Hsp90, Heat shock protein 90; HSF1, Heat shock factor 1; HeLa, Henrietta Lacks; HER1, Human epidermal growth factor receptor 1; IARC, International Agency for Research on Cancer; IL-10, Interleukin-10; KAP1, KRAB associated protein 1; MDM2, Mouse double minute 2 homolog; MDR, Multidrug resistance; MCF-7, Michigan cancer foundation-7; MEF-2, Myocyte enhancer factor-2MMP- Matrix metalloproteinase; NAD, Nicotinamide adenine dinucleotide; NuRD, Nucleosome remodelling and deacetylation; NF- κ B, Nuclear factor kappa light chain enhancer of activated B cell; NES, Nuclear export signal; NLS, Nuclear localization signal; NCoR, Nuclear receptor corepressor; NCT, National clinical trial; OS, Overall survival; PR, Progesterone receptor; PI3K, Phosphoinositide 3-kinase; PAX3, Paired box gene 3; P-gp, P-glycoprotein; ROS, Reactive oxygen species; SIRT, Sirtuin; SMRT, Silencing mediator for retinoid and thyroid receptor; STAT3, Signal transducer and activator of transcription-3; SAR, Structure-activity relationship; SHP1, Src homology region 2 domain-containing phosphatase 1; SAHA, Suberoylanilide hydroxamic acid; SMEDDS, Self micro emulsifying drug delivery system; TNBC, Triple-negative breast cancer; TSA, Trichostatin A; ZBG, Zinc binding group.
Collapse
Affiliation(s)
- Tanima Das
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Sunandita Bhar
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Diya Ghosh
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Bikash Kabi
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Kanisha Kar
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Arpita Chandra
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India.
| |
Collapse
|
6
|
Dorando HK, Andrews JM, Borcherding NC, Quinn CC, Schmidt JA, Khatavkar OU, Aluri J, Harmon MT, Watkins MP, Frank A, Cooper MA, Musiek AC, Mehta-Shah N, Payton JE. Single Cell Resolution Tracking of Cutaneous T-Cell Lymphoma Reveals Clonal Evolution in Disease Progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637715. [PMID: 39990420 PMCID: PMC11844462 DOI: 10.1101/2025.02.11.637715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cutaneous T-cell lymphoma (CTCL) remains a challenging disease due to its significant heterogeneity, therapy resistance, and relentless progression. Multi-omics technologies offer the potential to provide uniquely precise views of disease progression and response to therapy. We present here a comprehensive multi-omics view of CTCL clonal evolution, incorporating exome, whole genome, epigenome, bulk-, single cell (sc) VDJ-, and scRNA-sequencing of 114 clinically annotated serial skin, peripheral blood, and lymph node samples from 35 CTCL patients. We leveraged this extensive dataset to define the molecular underpinnings of CTCL progression in individual patients at single cell resolution with the goal of identifying clinically useful biomarkers and therapeutic targets. Our studies identified a large number of recurrent progression-associated clonal genomic alterations; we highlight mutation of CCR4, PI3K signaling, and PD-1 checkpoint pathways as evasion tactics deployed by malignant T cells. We also identified a gain of function mutation in STAT3 (D661Y) and demonstrated by CUT&RUN-seq that it enhances binding to transcription start sites of genes in Rho GTPase pathways, which we previously reported to have activated chromatin and increased expression in HDACi-resistant CTCL. These data provide further support for a previously unrecognized role for Rho GTPase pathway dysregulation in CTCL progression. A striking number of progression-associated mutations occurred in chromatin methylation modifiers, including EZH2, suggesting that EZH1/2 inhibition may also benefit patients with CTCL. Knowledge of these molecular changes should be leveraged for improved disease monitoring, biomarker-informed clinical trial design, and new therapeutic strategies in this challenging and incurable cancer.
Collapse
|
7
|
Yang G, Yang S, Li J, Jiang P, Tian X, Wang X, Wei J, Zhang X, Liu J. Low-dose treatment with Epirubicin, a novel histone deacetylase 1 inhibitor, exerts anti-leukemic effects by inducing ferroptosis. Eur J Pharmacol 2024; 985:177058. [PMID: 39413949 DOI: 10.1016/j.ejphar.2024.177058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
AIMS Leukemia is hematopoietic stem cell malignant tumor with poor outcomes. Histone deacetylase 1 (HDAC1) is highly expressed in leukemia and current HDAC1 inhibitors have clinical limitations in leukemia therapy. Therefore, novel HDAC1 inhibitor is imperative to being found and its mechanism needs to be further explored. MATERIALS AND METHODS Novel HDAC1 inhibitors were discovered through drug virtual screening. CCK-8, EdU and soft agar assay were used to assess the anti-leukemic effect of the candidate HDAC1 inhibitor. ROS, lipid peroxidation, intracellular Fe2+ and LIP assay were employed to verify cell ferroptosis. Additionally, a xenograft model was performed to explore the efficacy and safety of the candidate HDAC1 inhibitor in vivo. RESULTS HDAC1 might be a promising therapeutic target for leukemia and Epirubicin (Epi) could be used as a potential HDAC1 inhibitor. Low-dose Epi exhibited good anti-leukemic effects by inhibiting cell proliferation, DNA synthesis and colony formation. Low-dose Epi could induce ferroptosis by triggering lipid peroxidation, which was better than that treated with current HDAC1 inhibitors Chidamide or Vorinostat, ROS generation and Fe2+ overload in leukemia cells. Mechanistically, low-dose Epi induced ferroptosis by targeting amino acid metabolism and iron metabolism. Similar results were found in a xenograft model in NOG mice with a good safety profile. CONCLUSION Our study demonstrated that Epi might be used as a HDAC1 inhibitor. Low-dose Epi could inhibit tumor progression by inducing cell ferroptosis in vitro and in vivo. Thus, Epi administration with lower concentration may be much more favorable and safer in the treatment with leukemia.
Collapse
Affiliation(s)
- Guancui Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China; Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Shijie Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jiarun Li
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Peijie Jiang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China; Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Xiaolong Tian
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xiaoqi Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jin Wei
- Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Xi Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 401329, China.
| | - Jinyi Liu
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| |
Collapse
|
8
|
Fischer A, Albert TK, Moreno N, Interlandi M, Mormann J, Glaser S, Patil P, de Faria FW, Richter M, Verma A, Balbach ST, Wagener R, Bens S, Dahlum S, Göbel C, Münter D, Inserte C, Graf M, Kremer E, Melcher V, Di Stefano G, Santi R, Chan A, Dogan A, Bush J, Hasselblatt M, Cheng S, Spetalen S, Fosså A, Hartmann W, Herbrüggen H, Robert S, Oyen F, Dugas M, Walter C, Sandmann S, Varghese J, Rossig C, Schüller U, Tzankov A, Pedersen MB, d'Amore FA, Mellgren K, Kontny U, Kancherla V, Veloza L, Missiaglia E, Fataccioli V, Gaulard P, Burkhardt B, Soehnlein O, Klapper W, de Leval L, Siebert R, Kerl K. Lack of SMARCB1 expression characterizes a subset of human and murine peripheral T-cell lymphomas. Nat Commun 2024; 15:8571. [PMID: 39362842 PMCID: PMC11452211 DOI: 10.1038/s41467-024-52826-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
Peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS) is a heterogeneous group of malignancies with poor outcome. Here, we identify a subgroup, PTCL-NOSSMARCB1-, which is characterized by the lack of the SMARCB1 protein and occurs more frequently in young patients. Human and murine PTCL-NOSSMARCB1- show similar DNA methylation profiles, with hypermethylation of T-cell-related genes and hypomethylation of genes involved in myeloid development. Single-cell analyses of human and murine tumors revealed a rich and complex network of interactions between tumor cells and an immunosuppressive and exhausted tumor microenvironment (TME). In a drug screen, we identified histone deacetylase inhibitors (HDACi) as a class of drugs effective against PTCL-NOSSmarcb1-. In vivo treatment of mouse tumors with SAHA, a pan-HDACi, triggered remodeling of the TME, promoting replenishment of lymphoid compartments and reversal of the exhaustion phenotype. These results provide a rationale for further exploration of HDACi combination therapies targeting PTCL-NOSSMARCB1- within the TME.
Collapse
MESH Headings
- Animals
- SMARCB1 Protein/genetics
- SMARCB1 Protein/metabolism
- Humans
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/drug therapy
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/pathology
- Mice
- Histone Deacetylase Inhibitors/pharmacology
- Tumor Microenvironment/genetics
- Tumor Microenvironment/drug effects
- DNA Methylation
- Gene Expression Regulation, Neoplastic
- Female
- Cell Line, Tumor
- Male
- Vorinostat/pharmacology
- Single-Cell Analysis
Collapse
Affiliation(s)
- Anja Fischer
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Thomas K Albert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Natalia Moreno
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Marta Interlandi
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Jana Mormann
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Selina Glaser
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Paurnima Patil
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Flavia W de Faria
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Mathis Richter
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Archana Verma
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Sebastian T Balbach
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Rabea Wagener
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Susanne Bens
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Sonja Dahlum
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Carolin Göbel
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Eppendorf (UKE), 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Daniel Münter
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Clara Inserte
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Monika Graf
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Eva Kremer
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Viktoria Melcher
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Gioia Di Stefano
- Pathological Anatomy Section, Careggi University Hospital, Florence, Italy
| | - Raffaella Santi
- Pathological Anatomy Section, Careggi University Hospital, Florence, Italy
| | - Alexander Chan
- Department of Pathology, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Ahmet Dogan
- Department of Pathology, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Jonathan Bush
- Division of Anatomical Pathology, British Columbia Children's Hospital and Women's Hospital and Health Center, Vancouver, BC, Canada
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, 48149, Münster, Germany
| | - Sylvia Cheng
- Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Signe Spetalen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Alexander Fosså
- Department of Oncology, Oslo University Hospital-Norwegian Radium Hospital, Oslo, Norway
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard-Domagk-Institut für Pathologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude D17, 48149, Münster, Germany
| | - Heidi Herbrüggen
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Stella Robert
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Florian Oyen
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Eppendorf (UKE), 20251, Hamburg, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Sarah Sandmann
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Eppendorf (UKE), 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), 20251, Hamburg, Germany
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Martin B Pedersen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Francesco A d'Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karin Mellgren
- Department of Pediatric Oncology and Hematology, Sahlgrenska University Hospital, The Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Udo Kontny
- Section of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatric and Adolescent Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Venkatesh Kancherla
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Luis Veloza
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Edoardo Missiaglia
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Virginie Fataccioli
- INSERM U955, Université Paris-Est, Créteil, France
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, INSERM U955, Université Paris Est Créteil, Créteil, France
| | - Philippe Gaulard
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, INSERM U955, Université Paris Est Créteil, Créteil, France
| | - Birgit Burkhardt
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Wolfram Klapper
- Department of Pathology, Haematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany.
| |
Collapse
|
9
|
Irimia R, Piccaluga PP. Histone Deacetylase Inhibitors for Peripheral T-Cell Lymphomas. Cancers (Basel) 2024; 16:3359. [PMID: 39409979 PMCID: PMC11482620 DOI: 10.3390/cancers16193359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Histone deacetylase inhibitors (HDACis) are being recognized as a potentially effective treatment approach for peripheral T-cell lymphomas (PTCLs), a heterogeneous group of aggressive malignancies with an unfavorable prognosis. Recent evidence has shown that HDACis are effective in treating PTCL, especially in cases where the disease has relapsed or is resistant to conventional treatments. Several clinical trials have demonstrated that HDACis, such as romidepsin and belinostat, can elicit long-lasting positive outcomes in individuals with PTCLs, either when used alone or in conjunction with conventional chemotherapy. They exert their anti-tumor effects by regulating gene expression through the inhibition of histone deacetylases, which leads to cell cycle arrest, induction of programmed cell death, and,the transformation of cancerous T cells, as demonstrated by gene expression profile studies. Importantly, besides clinical trials, real-world evidence indicated that the utilization of HDACis presents a significant and beneficial treatment choice for PTCLs. However, although HDACis showed potential effectiveness, they could not cure most patients. Therefore, new combinations with conventional drugs as well as new targeted agents are under investigation.
Collapse
Affiliation(s)
- Ruxandra Irimia
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, 030167 Bucharest, Romania;
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Pier Paolo Piccaluga
- Department of Medical and Surgical Sciences, School of Medicine, University of Bologna, 40138 Bologna, Italy
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Institute of Hematology and Medical Oncology “L&A Seràgnoli”, 40138 Bologna, Italy
| |
Collapse
|
10
|
Zong X, Yang Z, Zhou J, Jin Z, Wu D. Clinical trial: Chidamide plus CHOP improve the survival of newly diagnosed angioimmunoblastic T-cell lymphoma. Front Immunol 2024; 15:1430648. [PMID: 39229263 PMCID: PMC11368836 DOI: 10.3389/fimmu.2024.1430648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/25/2024] [Indexed: 09/05/2024] Open
Abstract
Background Angioimmunoblastic T-cell lymphoma (AITL) is known for its unfavorable survival prognosis. Chidamide has shown efficacy in relapsed/refractory AITL, but its efficacy in newly diagnosed AITL is uncertain. Objective This retrospective research aimed to evaluate the effectiveness and safety of chidamide when used with doxorubicin, cyclophosphamide, prednisone, and vincristine (CHOP) in comparison to CHOP by itself for individuals newly diagnosed with AITL, and to examine the impact of transplantation. Method This was an analysis that compared outcomes among patients who received chidamide + CHOP on a clinical trial vs. historical controls who received CHOP alone, enrolling a total of sixty-six treatment-naive AITL patients between April 2014 and November 2022. Among them, thirty-three received chidamide in addition to CHOP (chidamide group), while thirty-three received CHOP alone (control group). The clinical characteristics were balanced between the two groups. All patients were scheduled to undergo up to six courses of treatment before transplantation. Results The chidamide group had a significantly longer median overall survival (OS) compared to the control group, with a median OS that was not reached, as opposed to 20 months in the control group (p = 0.002). In the control group, the median progression-free survival (PFS) was 11 months, while in the chidamide group, it was 22 months (p = 0.080). In the high-risk group (IPI ≥ 3), the chidamide group demonstrated notably superior complete response (CR) and overall response rate (ORR) compared to the control cohort (p = 0.002, p = 0.034). The PFS and OS in the chidamide group were not reached, and there were significant differences compared to the control group (p = 0.007, p = 0.003). The median OS of the transplanted group was longer than the non-transplanted group (p = 0.004). On multivariate analysis, chidamide group reduced the hazards of death in the total cohort. Conclusion As the study was non-random and retrospective, Chidamide combined with chemotherapy should be tested in randomized trials given its potential to improve prognosis in treatment-naive AITL patients. Furthermore, autologous hematopoietic stem cell transplantation (auto-HSCT) has demonstrated enhanced overall survival in individuals with AITL. Clinical trial registration https://clinicaltrials.gov/, NCT03268889.
Collapse
Affiliation(s)
- Xiangping Zong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhen Yang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Zhou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhengming Jin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
11
|
De Wilde S, Graux C. Complete hematologic response in a patient with multiple pretreated angioimmunoblastic T-cell lymphoma after belinostat therapy followed by allogeneic stem cell transplantation: A case report. Clin Case Rep 2024; 12:e9159. [PMID: 38979088 PMCID: PMC11228349 DOI: 10.1002/ccr3.9159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024] Open
Abstract
Key Clinical Message Belinostat therapy followed by hematopoietic stem cell transplantation is a promising salvage strategy for heavily pretreated patients with peripheral T-cell lymphoma. Abstract Effective treatments for peripheral T-cell lymphoma in the relapsed and refractory (r/r) setting are limited. However, with the development and approval of innovative therapies, effective therapeutic options are becoming available for this patient population. This case report describes the treatment course of a patient with multiple r/r nodal follicular T-helper cell lymphoma of angioimmunoblastic type. Treatment with the histone deacetylase inhibitor belinostat as bridging, enabled allogeneic stem cell transplantation and resulted in a durable complete hematologic response for at least 21 months post-transplantation.
Collapse
|
12
|
Yao L, Wang Q, Ma W. Navigating the Immune Maze: Pioneering Strategies for Unshackling Cancer Immunotherapy Resistance. Cancers (Basel) 2023; 15:5857. [PMID: 38136402 PMCID: PMC10742031 DOI: 10.3390/cancers15245857] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer immunotherapy has ushered in a transformative era in oncology, offering unprecedented promise and opportunities. Despite its remarkable breakthroughs, the field continues to grapple with the persistent challenge of treatment resistance. This resistance not only undermines the widespread efficacy of these pioneering treatments, but also underscores the pressing need for further research. Our exploration into the intricate realm of cancer immunotherapy resistance reveals various mechanisms at play, from primary and secondary resistance to the significant impact of genetic and epigenetic factors, as well as the crucial role of the tumor microenvironment (TME). Furthermore, we stress the importance of devising innovative strategies to counteract this resistance, such as employing combination therapies, tailoring immune checkpoints, and implementing real-time monitoring. By championing these state-of-the-art methods, we anticipate a paradigm that blends personalized healthcare with improved treatment options and is firmly committed to patient welfare. Through a comprehensive and multifaceted approach, we strive to tackle the challenges of resistance, aspiring to elevate cancer immunotherapy as a beacon of hope for patients around the world.
Collapse
Affiliation(s)
- Liqin Yao
- Key Laboratory for Translational Medicine, The First Affiliated Hospital, Huzhou University, Huzhou 313000, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China;
| | - Wenxue Ma
- Department of Medicine, Moores Cancer Center, Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|