1
|
Reininger D, Fundel-Clemens K, Mayr CH, Wollin L, Laemmle B, Quast K, Nickolaus P, Herrmann FE. PDE4B inhibition by nerandomilast: Effects on lung fibrosis and transcriptome in fibrotic rats and on biomarkers in human lung epithelial cells. Br J Pharmacol 2024; 181:4766-4781. [PMID: 39183442 DOI: 10.1111/bph.17303] [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: 02/22/2024] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND AND PURPOSE The PDE4 family is considered a prime target for therapeutic intervention in several fibro-inflammatory diseases. We have investigated the molecular mechanisms of nerandomilast (BI 1015550), a preferential PDE4B inhibitor. EXPERIMENTAL APPROACH In addition to clinically relevant parameters of idiopathic pulmonary fibrosis (IPF; lung function measurement/high-resolution computed tomography scan/AI-Ashcroft score), whole-lung homogenates from a therapeutic male Wistar rat model of pulmonary fibrosis were analysed by next-generation sequencing (NGS). Data were matched with public domain data derived from human IPF samples to investigate how well the rat model reflected human IPF. We scored the top counter-regulated genes following treatment with nerandomilast in human single cells and validated disease markers discovered in the rat model using a human disease-relevant in vitro assay of IPF. KEY RESULTS Nerandomilast improved the decline of lung function parameters in bleomycin-treated animals. In the NGS study, most transcripts deregulated by bleomycin treatment were normalised by nerandomilast treatment. Most notably, a significant number of deregulated transcripts that were identified in human IPF disease were also found in the animal model and reversed by nerandomilast. Mapping to single-cell data revealed the strongest effects on mesenchymal, epithelial and endothelial cell populations. In a primary human epithelial cell culture system, several disease-related (bio)markers were inhibited by nerandomilast in a concentration-dependent manner. CONCLUSIONS AND IMPLICATIONS This study further supports the available knowledge about the anti-inflammatory/antifibrotic mechanisms of nerandomilast and provides novel insights into the mode of action and signalling pathways influenced by nerandomilast treatment of lung fibrosis.
Collapse
Affiliation(s)
- Dennis Reininger
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Katrin Fundel-Clemens
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Christoph H Mayr
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Lutz Wollin
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Baerbel Laemmle
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Karsten Quast
- Global Clinical Development & Operations, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Peter Nickolaus
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Franziska Elena Herrmann
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
2
|
Alayoubi AM, Khawaji ZY, Mohammed MA, Mercier FE. CRISPR-Cas9 system: a novel and promising era of genotherapy for beta-hemoglobinopathies, hematological malignancy, and hemophilia. Ann Hematol 2024; 103:1805-1817. [PMID: 37736806 DOI: 10.1007/s00277-023-05457-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023]
Abstract
Gene therapy represents a significant potential to revolutionize the field of hematology with applications in correcting genetic mutations, generating cell lines and animal models, and improving the feasibility and efficacy of cancer immunotherapy. Compared to different genetic engineering tools, clustered regularly interspaced short palindromic repeats (CRISPR) CRISPR-associated protein 9 (Cas9) emerged as an effective and versatile genetic editor with the ability to precisely modify the genome. The applications of genetic engineering in various hematological disorders have shown encouraging results. Monogenic hematological disorders can conceivably be corrected with single gene modification. Through the use of CRISPR-CAS9, restoration of functional red blood cells and hemostasis factors were successfully attained in sickle cell anemia, beta-thalassemia, and hemophilia disorders. Our understanding of hemato-oncology has been advanced via CRIPSR-CAS9 technology. CRISPR-CAS9 aided to build a platform of mutated genes responsible for cell survival and proliferation in leukemia. Therapeutic application of CRISPR-CAS9 when combined with chimeric antigen receptor (CAR) T cell therapy in multiple myeloma and acute lymphoblastic leukemia was feasible with attenuation of CAR T cell therapy pitfalls. Our review outlines the latest literature on the utilization of CRISPR-Cas9 in the treatment of beta-hemoglobinopathies and hemophilia disorders. We present the strategies that were employed and the findings of preclinical and clinical trials. Also, the review will discuss gene engineering in the field of hemato-oncology as a proper tool to facilitate and overcome the drawbacks of chimeric antigen receptor T cell therapy (CAR-T).
Collapse
Affiliation(s)
- Abdulfatah M Alayoubi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | | | | | - François E Mercier
- Divisions of Experimental Medicine & Hematology, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
3
|
Sauer J, Steixner-Kumar AA, Gabler S, Motyka M, Rippmann JF, Brosa S, Boettner D, Schönberger T, Lempp C, Frodermann V, Simon E, Krenkel O, Bahrami E. Diverse potential of secretome from natural killer cells and monocyte-derived macrophages in activating stellate cells. Front Immunol 2024; 15:1232070. [PMID: 38638443 PMCID: PMC11025356 DOI: 10.3389/fimmu.2024.1232070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 03/04/2024] [Indexed: 04/20/2024] Open
Abstract
Chronic liver diseases, such as non-alcoholic steatohepatitis (NASH)-induced cirrhosis, are characterized by an increasing accumulation of stressed, damaged, or dying hepatocytes. Hepatocyte damage triggers the activation of resident immune cells, such as Kupffer cells (KC), as well as the recruitment of immune cells from the circulation toward areas of inflammation. After infiltration, monocytes differentiate into monocyte-derived macrophages (MoMF) which are functionally distinct from resident KC. We herein aim to compare the in vitro signatures of polarized macrophages and activated hepatic stellate cells (HSC) with ex vivo-derived disease signatures from human NASH. Furthermore, to shed more light on HSC activation and liver fibrosis progression, we investigate the effects of the secretome from primary human monocytes, macrophages, and NK cells on HSC activation. Interleukin (IL)-4 and IL-13 treatment induced transforming growth factor beta 1 (TGF-β1) secretion by macrophages. However, the supernatant transfer did not induce HSC activation. Interestingly, PMA-activated macrophages showed strong induction of the fibrosis response genes COL10A1 and CTGF, while the supernatant of IL-4/IL-13-treated monocytes induced the upregulation of COL3A1 in HSC. The supernatant of PMA-activated NK cells had the strongest effect on COL10A1 induction in HSC, while IL-15-stimulated NK cells reduced the expression of COL1A1 and CTGF. These data indicate that other factors, aside from the well-known cytokines and chemokines, might potentially be stronger contributors to the activation of HSCs and induction of a fibrotic response, indicating a more diverse and complex role of monocytes, macrophages, and NK cells in liver fibrosis progression.
Collapse
Affiliation(s)
- Julia Sauer
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Svenja Gabler
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | | | - Stefan Brosa
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dennis Boettner
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Charlotte Lempp
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Eric Simon
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Oliver Krenkel
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Ehsan Bahrami
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
4
|
Jin Y, Wu H, Liu J, Cho WC, Song G. Application and progress of CRISPR/Cas9 gene editing in B-cell lymphoma: a narrative review. Transl Cancer Res 2024; 13:1584-1595. [PMID: 38617522 PMCID: PMC11009809 DOI: 10.21037/tcr-23-1146] [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: 07/05/2023] [Accepted: 01/23/2024] [Indexed: 04/16/2024]
Abstract
Background and Objective Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9) gene editing and CRISPR/Cas9 screening libraries are hot topics, and have high application values in the diagnosis and treatment of genetic diseases, and the improvement of prognosis. The major treatment of B-cell lymphoma is chemotherapy combined with biological therapy. Due to the individual specificity and the emergence of drug resistance, the therapeutic efficacy varies. The objective of this article is to explore potential targets to enhance therapeutic effects, optimize treatment plans, and improve the prognosis of patients with B-cell lymphoma. Methods We undertook a comprehensive, narrative review of the latest literature to define the current application and progress of CRISPR/Cas9 in B-cell lymphoma. Key Content and Findings The concepts of CRISPR/Cas9, the mechanism of gene editing, and the procedures of CRISPR/Cas9 screening libraries are investigated for candidate genes. We mainly focus on application and progress of CRISPR/Cas9 in B-cell lymphoma and screen out some genes, signaling pathways, and cytokines, which may become potential targets for clinical treatment. Conclusions CRISPR/Cas9 gene editing has great promise in the treatment of B-cell lymphoma. This article reviews some genes, signaling pathways, and cytokines related to the progression and prognosis of B-cell lymphoma to provide a strong theoretical basis.
Collapse
Affiliation(s)
- Ying Jin
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Haiyi Wu
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Jianzhao Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Dalian Medical University, Dalian, China
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Guoqi Song
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
5
|
Cai Z, You S, Liu Z, Song P, Zhao F, An J, Ding Y, He B, Zou MH. Selective deletion of E3 ubiquitin ligase FBW7 in VE-cadherin-positive cells instigates diffuse large B-cell lymphoma in mice in vivo. Cell Death Dis 2024; 15:212. [PMID: 38485719 PMCID: PMC10940678 DOI: 10.1038/s41419-024-06597-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024]
Abstract
During the maturation of hematopoietic stem/progenitor cells (HSPCs) to fully differentiated mature B lymphocytes, developing lymphocytes may undergo malignant transformation and produce B-cell lymphomas. Emerging evidence shows that through the endothelial-hematopoietic transition, specialized endothelial cells called the hemogenic endothelium can differentiate into HSPCs. However, the contribution of genetic defects in hemogenic endothelial cells to B-cell lymphomagenesis has not yet been investigated. Here, we report that mice with endothelial cell-specific deletion of Fbw7 spontaneously developed diffuse large B-cell lymphoma (DLBCL) following Bcl6 accumulation. Using lineage tracing, we showed that B-cell lymphomas in Fbw7 knockout mice were hemogenic endothelium-derived. Mechanistically, we found that FBW7 directly interacted with Bcl6 and promoted its proteasomal degradation. FBW7 expression levels are inversely correlated with BCL6 expression. Additionally, pharmacological disruption of Bcl6 abolished Fbw7 deletion-induced B-cell lymphomagenesis. We conclude that selective deletion of E3 ubiquitin ligase FBW7 in VE-cadherin positive endothelial cells instigates diffuse large B-cell lymphoma via upregulation of BCL6 stability. In addition, the mice with endothelial cell-specific deletion of Fbw7 provide a valuable preclinical platform for in vivo development and evaluation of novel therapeutic interventions for the treatment of DLBCL.
Collapse
Affiliation(s)
- Zhaohua Cai
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, 30303, USA
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Shaojin You
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, 30303, USA
| | - Zhixue Liu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, 30303, USA
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, 30303, USA
| | - Fujie Zhao
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, 30303, USA
| | - Junqing An
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, 30303, USA
| | - Ye Ding
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, 30303, USA
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
6
|
Bahrami E, Geiger T, Steixner-Kumar AA, Santacruz D, Viollet C, Dick A, Roth Y, Schlingeloff P, Schmidberger J, Haenle M, Kratzer W, Kitt K, Neubauer H, Simon E, Krenkel O, Werner M. An optimized protocol for isolation of hepatic leukocytes retrieved from murine and NASH liver biopsies. STAR Protoc 2023; 4:102597. [PMID: 37740914 PMCID: PMC10520930 DOI: 10.1016/j.xpro.2023.102597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/27/2023] [Accepted: 08/31/2023] [Indexed: 09/25/2023] Open
Abstract
Immune dysregulation and inflammation by hepatic-resident leukocytes is considered a key step in disease progression of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis toward cirrhosis and hepatocellular carcinoma. Here, we provide a protocol for isolation and characterization of liver-resident immune cells from fine-needle biopsies obtained from a rodent model and humans. We describe steps for isolating leukocytes, cell sorting, and RNA extraction and sequencing. We then detail procedures for low-input mRNA sequencing analyses.
Collapse
Affiliation(s)
- Ehsan Bahrami
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Tobias Geiger
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Diana Santacruz
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Coralie Viollet
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Alec Dick
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Yvonne Roth
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | | | - Mark Haenle
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Wolfgang Kratzer
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Kerstin Kitt
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Heike Neubauer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Eric Simon
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Oliver Krenkel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Markus Werner
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.
| |
Collapse
|
7
|
Weckerle J, Mayr CH, Fundel-Clemens K, Lämmle B, Boryn L, Thomas MJ, Bretschneider T, Luippold AH, Huber HJ, Viollet C, Rist W, Veyel D, Ramirez F, Klee S, Kästle M. Transcriptomic and Proteomic Changes Driving Pulmonary Fibrosis Resolution in Young and Old Mice. Am J Respir Cell Mol Biol 2023; 69:422-440. [PMID: 37411041 DOI: 10.1165/rcmb.2023-0012oc] [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: 01/09/2023] [Accepted: 07/06/2023] [Indexed: 07/08/2023] Open
Abstract
Bleomycin-induced pulmonary fibrosis in mice mimics major hallmarks of idiopathic pulmonary fibrosis. Yet in this model, it spontaneously resolves over time. We studied molecular mechanisms of fibrosis resolution and lung repair, focusing on transcriptional and proteomic signatures and the effect of aging. Old mice showed incomplete and delayed lung function recovery 8 weeks after bleomycin instillation. This shift in structural and functional repair in old bleomycin-treated mice was reflected in a temporal shift in gene and protein expression. We reveal gene signatures and signaling pathways that underpin the lung repair process. Importantly, the downregulation of WNT, BMP, and TGFβ antagonists Frzb, Sfrp1, Dkk2, Grem1, Fst, Fstl1, and Inhba correlated with lung function improvement. Those genes constitute a network with functions in stem cell pathways, wound, and pulmonary healing. We suggest that insufficient and delayed downregulation of those antagonists during fibrosis resolution in old mice explains the impaired regenerative outcome. Together, we identified signaling pathway molecules with relevance to lung regeneration that should be tested in-depth experimentally as potential therapeutic targets for pulmonary fibrosis.
Collapse
Affiliation(s)
| | | | | | - Bärbel Lämmle
- Global Computational Biology and Digital Sciences, and
| | | | | | - Tom Bretschneider
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | - Andreas H Luippold
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | | | | | - Wolfgang Rist
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | - Daniel Veyel
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | - Fidel Ramirez
- Global Computational Biology and Digital Sciences, and
| | - Stephan Klee
- Department of Immunology and Respiratory Disease Research
| | - Marc Kästle
- Department of Immunology and Respiratory Disease Research
| |
Collapse
|
8
|
Zhou M, Cui Y, Zuo S, Peng Q, Liu Y, Li X, Yang Y, He Q, Yu X, Zhou J, He Z, He Q. ZBTB40 is a telomere-associated protein and protects telomeres in human ALT cells. J Biol Chem 2023; 299:105053. [PMID: 37454741 PMCID: PMC10480536 DOI: 10.1016/j.jbc.2023.105053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023] Open
Abstract
Alternative lengthening of telomeres (ALTs) mechanism is activated in some somatic, germ cells, and human cancer cells. However, the key regulators and mechanisms of the ALT pathway remain elusive. Here we demonstrated that ZBTB40 is a novel telomere-associated protein and binds to telomeric dsDNA through its N-terminal BTB (BR-C, ttk and bab) or POZ (Pox virus and Zinc finger) domain in ALT cells. Notably, the knockout or knockdown of ZBTB40 resulted in the telomere dysfunction-induced foci and telomere lengthening in the ALT cells. The results also show that ZBTB40 is associated with ALT-associated promyelocytic leukemia nuclear bodies, and the loss of ZBTB40 induces the accumulation of the ALT-associated promyelocytic leukemia nuclear bodies in U2OS cells. Taken together, our results implicate that ZBTB40 is a key player of telomere protection and telomere lengthening regulation in human ALT cells.
Collapse
Affiliation(s)
- Mingqing Zhou
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yinghong Cui
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Shanru Zuo
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Qiyao Peng
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yucong Liu
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Xueguang Li
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yide Yang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Quanze He
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Xing Yu
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Junhua Zhou
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Zuping He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China.
| | - Quanyuan He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China.
| |
Collapse
|
9
|
Lee B, Pierpont T, August A, Richards K. Monoclonal antibodies binding to different epitopes of CD20 differentially sensitize DLBCL to different classes of chemotherapy. Front Oncol 2023; 13:1159484. [PMID: 37601699 PMCID: PMC10436104 DOI: 10.3389/fonc.2023.1159484] [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: 02/06/2023] [Accepted: 07/04/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Rituximab (R), an anti-CD20 monoclonal antibody (mAb) and the world's first approved antibody for oncology patients, was combined with the CHOP chemotherapy regimen and markedly improved the prognosis of all B- cell-derived lymphomas, the most common hematological malignancy worldwide. However, there is a 35% disease recurrence with no advancement in the first-line treatment since R was combined with the archetypal CHOP chemotherapy regimen nearly 30 years ago. There is evidence that R synergizes with chemotherapy, but the pharmacological interactions between R and CHOP or between newer anti-CD20 mAbs and CHOP remain largely unexplored. Methods We used in vitro models to score pharmacological interactions between R and CHOP across various lymphoma cell lines. We compared these pharmacological interactions to ofatumumab, a second-generation anti-CD20 mAb, and CHOP. Lastly, we used RNA-sequencing to characterize the transcriptional profiles induced by these two antibodies and potential molecular pathways that mediate their different effects. Results We discovered vast heterogeneity in the pharmacological interactions between R and CHOP in a way not predicted by the current clinical classification. We then discovered that R and ofatumumab differentially synergize with the cytotoxic and cytostatic capabilities of CHOP in separate distinct subsets of B-cell lymphoma cell lines, thereby expanding favorable immunochemotherapy interactions across a greater range of cell lines beyond those induced by R-CHOP. Lastly, we discovered these two mAbs differentially modulate genes enriched in the JNK and p38 MAPK family, which regulates apoptosis and proliferation. Discussion Our findings were completely unexpected because these mAbs were long considered to be biological and clinical equivalents but, in practice, may perform better than the other in a patient-specific manner. This finding may have immediate clinical significance because both immunochemotherapy combinations are already FDA-approved with no difference in toxicity across phase I, II, and III clinical trials. Therefore, this finding could inform a new precision medicine strategy to provide additional therapeutic benefit to patients with B-cell lymphoma using immunochemotherapy combinations that already meet the clinical standard of care.
Collapse
Affiliation(s)
- Brian Lee
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Tim Pierpont
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Kristy Richards
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
10
|
Efthymiou V, Ding L, Balaz M, Sun W, Balazova L, Straub LG, Dong H, Simon E, Ghosh A, Perdikari A, Keller S, Ghoshdastider U, Horvath C, Moser C, Hamilton B, Neubauer H, Wolfrum C. Inhibition of AXL receptor tyrosine kinase enhances brown adipose tissue functionality in mice. Nat Commun 2023; 14:4162. [PMID: 37443109 PMCID: PMC10344962 DOI: 10.1038/s41467-023-39715-8] [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: 08/10/2021] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The current obesity epidemic and high prevalence of metabolic diseases necessitate efficacious and safe treatments. Brown adipose tissue in this context is a promising target with the potential to increase energy expenditure, however no pharmacological treatments activating brown adipose tissue are currently available. Here, we identify AXL receptor tyrosine kinase as a regulator of adipose function. Pharmacological and genetic inhibition of AXL enhance thermogenic capacity of brown and white adipocytes, in vitro and in vivo. Mechanistically, these effects are mediated through inhibition of PI3K/AKT/PDE signaling pathway, resulting in induction of nuclear FOXO1 localization and increased intracellular cAMP levels via PDE3/4 inhibition and subsequent stimulation of the PKA-ATF2 pathway. In line with this, both constitutive Axl deletion as well as inducible adipocyte-specific Axl deletion protect animals from diet-induced obesity concomitant with increases in energy expenditure. Based on these data, we propose AXL receptor as a target for the treatment of obesity.
Collapse
Affiliation(s)
- Vissarion Efthymiou
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Joslin Diabetes Center, Section of Integrative Physiology and Metabolism, Research Division, Harvard Medical School, Boston, MA, USA
| | - Lianggong Ding
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Miroslav Balaz
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Laboratory of Cellular and Molecular Metabolism, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Wenfei Sun
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lucia Balazova
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Laboratory of Cellular and Molecular Metabolism, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Leon G Straub
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Institute of Child Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Karlsruhe, Germany
| | - Hua Dong
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Eric Simon
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Adhideb Ghosh
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Aliki Perdikari
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Svenja Keller
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Mechanisms of Inherited Kidney Diseases Group, Institute of Physiology, University of Zurich, 8057, Zurich, Switzerland
| | - Umesh Ghoshdastider
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Carla Horvath
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Caroline Moser
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Bradford Hamilton
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Heike Neubauer
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Christian Wolfrum
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland.
| |
Collapse
|
11
|
Simon E, Motyka M, Prins GH, Li M, Rust W, Kauschke S, Viollet C, Olinga P, Oldenburger A. Transcriptomic profiling of induced steatosis in human and mouse precision-cut liver slices. Sci Data 2023; 10:304. [PMID: 37208356 DOI: 10.1038/s41597-023-02220-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/09/2023] [Indexed: 05/21/2023] Open
Abstract
There is a high need for predictive human ex vivo models for non-alcoholic fatty liver disease (NAFLD). About a decade ago, precision-cut liver slices (PCLSs) have been established as an ex vivo assay for humans and other organisms. In the present study, we use transcriptomics by RNASeq to profile a new human and mouse PCLSs based assay for steatosis in NAFLD. Steatosis as quantified by an increase of triglycerides after 48 h in culture, is induced by incremental supplementation of sugars (glucose and fructose), insulin, and fatty acids (palmitate, oleate). We mirrored the experimental design for human vs. mouse liver organ derived PCLSs and profiled each organ at eight different nutrient conditions after 24 h and 48 h time in culture. Thus, the provided data allows a comprehensive analysis of the donor, species, time, and nutrient factor specific regulation of gene expression in steatosis, despite the heterogeneity of the human tissue samples. Exemplified this is demonstrated by ranking homologous gene pairs by convergent or divergent expression pattern across nutrient conditions.
Collapse
Affiliation(s)
- Eric Simon
- Global Computational Biology and Digital Science, Research Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany.
| | | | - Grietje H Prins
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen Research Institute of Pharmacy, A, Deusinglaan 1, 9713AV, Groningen, The Netherlands
| | - Mei Li
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen Research Institute of Pharmacy, A, Deusinglaan 1, 9713AV, Groningen, The Netherlands
| | - Werner Rust
- Global Computational Biology and Digital Science, Research Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Stefan Kauschke
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Coralie Viollet
- Global Computational Biology and Digital Science, Research Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen Research Institute of Pharmacy, A, Deusinglaan 1, 9713AV, Groningen, The Netherlands
| | - Anouk Oldenburger
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| |
Collapse
|
12
|
Karimi S, Shahabi F, Mubarak SMH, Arjmandi H, Hashemi ZS, Pourzardosht N, Zakeri A, Mahboobi M, Jahangiri A, Rahbar MR, Khalili S. Impact of SNPs, off-targets, and passive permeability on efficacy of BCL6 degrading drugs assigned by virtual screening and 3D-QSAR approach. Sci Rep 2022; 12:21091. [PMID: 36473934 PMCID: PMC9726907 DOI: 10.1038/s41598-022-25587-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
B-cell lymphoma 6 (BCL6) regulates various genes and is reported to be overexpressed in lymphomas and other malignancies. Thus, BCL6 inhibition or its tagging for degradation would be an amenable therapeutic approach. A library of 2500 approved drugs was employed to find BCL6 inhibitory molecules via virtual screening. Moreover, the 3D core structure of 170 BCL6 inhibitors was used to build a 3D QSAR model and predict the biological activity. The SNP database was analyzed to study the impact on the destabilization of BCL6/drug interactions. Structural similarity search and molecular docking analyses were used to assess the interaction between possible off-targets and BCL6 inhibitors. The tendency of drugs for passive membrane permeability was also analyzed. Lifitegrast (DB11611) had favorable binding properties and biological activity compared to the BI-3802. Missense SNPs were located at the essential interaction sites of the BCL6. Structural similarity search resulted in five BTB-domain containing off-target proteins. BI-3802 and Lifitegrast had similar chemical behavior and binding properties against off-target candidates. More interestingly, the binding affinity of BI-3802 (against off-targets) was higher than Lifitegrast. Energetically, Lifitegrast was less favorable for passive membrane permeability. The interaction between BCL6 and BI-3802 is more prone to SNP-derived variations. On the other hand, higher nonspecific binding of BI-3802 to off-target proteins could bring about higher undesirable properties. It should also be noted that energetically less desirable passive membrane translocation of Lifitegrast would demand drug delivery vehicles. However, further empirical evaluation of Lifitegrast would unveil its true potential.
Collapse
Affiliation(s)
- Solmaz Karimi
- grid.419305.a0000 0001 1943 2944Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Farzaneh Shahabi
- grid.411747.00000 0004 0418 0096Faculty of Advanced Technologies in Medical Sciences, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shaden M. H. Mubarak
- grid.442852.d0000 0000 9836 5198Department of Clinical Laboratory Science, Faculty of Pharmacy, University of Kufa, Najaf, Iraq
| | - Hanie Arjmandi
- grid.467532.10000 0004 4912 2930Faculty of Pharmacy, Islamic Azad University of Amol Branch, Amol, Iran
| | - Zahra Sadat Hashemi
- grid.417689.5ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Navid Pourzardosht
- grid.411874.f0000 0004 0571 1549Biochemistry Department, Guilan University of Medical Sciences, Rasht, Iran
| | - Alireza Zakeri
- grid.440791.f0000 0004 0385 049XDepartment of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Mahdieh Mahboobi
- grid.411521.20000 0000 9975 294XApplied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abolfazl Jahangiri
- grid.411521.20000 0000 9975 294XApplied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Rahbar
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Khalili
- grid.440791.f0000 0004 0385 049XDepartment of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
13
|
Zimmermann T, Thomas L, Baader-Pagler T, Haebel P, Simon E, Reindl W, Bajrami B, Rist W, Uphues I, Drucker DJ, Klein H, Santhanam R, Hamprecht D, Neubauer H, Augustin R. BI 456906: Discovery and preclinical pharmacology of a novel GCGR/GLP-1R dual agonist with robust anti-obesity efficacy. Mol Metab 2022; 66:101633. [PMID: 36356832 PMCID: PMC9679702 DOI: 10.1016/j.molmet.2022.101633] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/18/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE Obesity and its associated comorbidities represent a global health challenge with a need for well-tolerated, effective, and mechanistically diverse pharmaceutical interventions. Oxyntomodulin is a gut peptide that activates the glucagon receptor (GCGR) and glucagon-like peptide-1 receptor (GLP-1R) and reduces bodyweight by increasing energy expenditure and reducing energy intake in humans. Here we describe the pharmacological profile of the novel glucagon receptor (GCGR)/GLP-1 receptor (GLP-1R) dual agonist BI 456906. METHODS BI 456906 was characterized using cell-based in vitro assays to determine functional agonism. In vivo pharmacological studies were performed using acute and subchronic dosing regimens to demonstrate target engagement for the GCGR and GLP-1R, and weight lowering efficacy. RESULTS BI 456906 is a potent, acylated peptide containing a C18 fatty acid as a half-life extending principle to support once-weekly dosing in humans. Pharmacological doses of BI 456906 provided greater bodyweight reductions in mice compared with maximally effective doses of the GLP-1R agonist semaglutide. BI 456906's superior efficacy is the consequence of increased energy expenditure and reduced food intake. Engagement of both receptors in vivo was demonstrated via glucose tolerance, food intake, and gastric emptying tests for the GLP-1R, and liver nicotinamide N-methyltransferase mRNA expression and circulating biomarkers (amino acids, fibroblast growth factor-21) for the GCGR. The dual activity of BI 456906 at the GLP-1R and GCGR was supported using GLP-1R knockout and transgenic reporter mice, and an ex vivo bioactivity assay. CONCLUSIONS BI 456906 is a potent GCGR/GLP-1R dual agonist with robust anti-obesity efficacy achieved by increasing energy expenditure and decreasing food intake.
Collapse
Affiliation(s)
- Tina Zimmermann
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Leo Thomas
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Tamara Baader-Pagler
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Peter Haebel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Eric Simon
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Wolfgang Reindl
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Besnik Bajrami
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Wolfgang Rist
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Ingo Uphues
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, University of Toronto, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.
| | - Holger Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Rakesh Santhanam
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Dieter Hamprecht
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany; Boehringer Ingelheim Research Italia, Via Lorenzini 8, 20139 Milano, Italy.
| | - Heike Neubauer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| | - Robert Augustin
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400 Biberach an der Riβ, Germany.
| |
Collapse
|
14
|
Advances in CRISPR/Cas9. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9978571. [PMID: 36193328 PMCID: PMC9525763 DOI: 10.1155/2022/9978571] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/09/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
Abstract
CRISPR/Cas9 technology has become the most examined gene editing technology in recent years due to its simple design, yet low cost, high efficiency, and simple operation, which can also achieve simultaneous editing of multiple loci. It can also be carried out without using plasmids, saving lots of troubles caused by plasmids. CRISPR/Cas9 has shown great potential in the study of genes or genomic functions in microorganisms, plants, animals, and human beings. In this review, we will examine the history, structure, and basic mechanisms of the CRISPR/Cas9 system, describe its great value in precision medicine and sgRNA library screening, and dig its great potential in a new field: DNA information storage.
Collapse
|
15
|
Huckvale R, Harnden AC, Cheung KMJ, Pierrat OA, Talbot R, Box GM, Henley AT, de Haven Brandon AK, Hallsworth AE, Bright MD, Akpinar HA, Miller DSJ, Tarantino D, Gowan S, Hayes A, Gunnell EA, Brennan A, Davis OA, Johnson LD, de Klerk S, McAndrew C, Le Bihan YV, Meniconi M, Burke R, Kirkin V, van Montfort RLM, Raynaud FI, Rossanese OW, Bellenie BR, Hoelder S. Improved Binding Affinity and Pharmacokinetics Enable Sustained Degradation of BCL6 In Vivo. J Med Chem 2022; 65:8191-8207. [PMID: 35653645 PMCID: PMC9234961 DOI: 10.1021/acs.jmedchem.1c02175] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Indexed: 11/30/2022]
Abstract
The transcriptional repressor BCL6 is an oncogenic driver found to be deregulated in lymphoid malignancies. Herein, we report the optimization of our previously reported benzimidazolone molecular glue-type degrader CCT369260 to CCT373566, a highly potent probe suitable for sustained depletion of BCL6 in vivo. We observed a sharp degradation SAR, where subtle structural changes conveyed the ability to induce degradation of BCL6. CCT373566 showed modest in vivo efficacy in a lymphoma xenograft mouse model following oral dosing.
Collapse
Affiliation(s)
- Rosemary Huckvale
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Alice C. Harnden
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Kwai-Ming J. Cheung
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Olivier A. Pierrat
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rachel Talbot
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Gary M. Box
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Alan T. Henley
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | | | - Albert E. Hallsworth
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Michael D. Bright
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Hafize Aysin Akpinar
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Daniel S. J. Miller
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Dalia Tarantino
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Sharon Gowan
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Angela Hayes
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Emma A. Gunnell
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K.
| | - Alfie Brennan
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Owen A. Davis
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Louise D. Johnson
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Selby de Klerk
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Craig McAndrew
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Yann-Vaï Le Bihan
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K.
| | - Mirco Meniconi
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rosemary Burke
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Vladimir Kirkin
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rob L. M. van Montfort
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K.
| | - Florence I. Raynaud
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Olivia W. Rossanese
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Benjamin R. Bellenie
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Swen Hoelder
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| |
Collapse
|
16
|
Dai M, Radhakrishnan S, Li R, Tan R, Yan K, Fan G, Liu M. Targeted Protein Degradation: An Important Tool for Drug Discovery for "Undruggable" Tumor Transcription Factors. Technol Cancer Res Treat 2022; 21:15330338221095950. [PMID: 35466792 PMCID: PMC9047787 DOI: 10.1177/15330338221095950] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Conventional small-molecule drugs (SMDs) are compounds characterized by low
molecular weight, high cell permeability, and high selectivity. In clinical
translation, SMDs are regarded as good candidates for oral drug formulation. SMD
inhibitors play an important role in cancer treatment; however, resistance and
low effectiveness have been major bottlenecks in clinical application.
Generally, only 20% of cell proteins can potentially be targeted and have been
developed as SMDs; thus, some types of tumor targets are considered
“undruggable.” Among these are transcription factors (TFs), an important class
of proteins that regulate the occurrence, formation, and development of tumors.
It is difficult for SMDs and macromolecular drugs to identify bioactive sites in
TFs and hence for use as pharmacological inhibitors in targeting TF proteins.
For this reason, technologies that enable targeted protein degradation, such as
proteolysis-targeting chimera or molecular glues, could serve as a potential
tool to solve these conundrums.
Collapse
Affiliation(s)
- Mengyuan Dai
- Department of Gynecological Oncology, 89674Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Sridhar Radhakrishnan
- Cancer Science Institute of Singapore, 37580National University of Singapore, Singapore, Singapore
| | - Rui Li
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, 598782Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Kuo Yan
- Institute of Cell and Neurobiology, Charité Medical University, Berlin, Germany
| | - Gang Fan
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,477382The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Miao Liu
- Department of Pathology, 1861Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Pearce AC, Bamford MJ, Barber R, Bridges A, Convery MA, Demetriou C, Evans S, Gobbetti T, Hirst DJ, Holmes DS, Hutchinson JP, Jayne S, Lezina L, McCabe MT, Messenger C, Morley J, Musso MC, Scott-Stevens P, Manso AS, Schofield J, Slocombe T, Somers D, Walker AL, Wyce A, Zhang XP, Wagner SD. GSK137, a potent small-molecule BCL6 inhibitor with in vivo activity, suppresses antibody responses in mice. J Biol Chem 2021; 297:100928. [PMID: 34274316 PMCID: PMC8350397 DOI: 10.1016/j.jbc.2021.100928] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
B-cell lymphoma 6 (BCL6) is a zinc finger transcriptional repressor possessing a BTB-POZ (BR-C, ttk, and bab for BTB; pox virus and zinc finger for POZ) domain, which is required for homodimerization and association with corepressors. BCL6 has multiple roles in normal immunity, autoimmunity, and some types of lymphoma. Mice bearing disrupted BCL6 loci demonstrate suppressed high-affinity antibody responses to T-dependent antigens. The corepressor binding groove in the BTB-POZ domain is a potential target for small compound-mediated therapy. Several inhibitors targeting this binding groove have been described, but these compounds have limited or absent in vivo activity. Biophysical studies of a novel compound, GSK137, showed an in vitro pIC50 of 8 and a cellular pIC50 of 7.3 for blocking binding of a peptide derived from the corepressor silencing mediator for retinoid or thyroid hormone receptors to the BCL6 BTB-POZ domain. The compound has good solubility (128 μg/ml) and permeability (86 nM/s). GSK137 caused little change in cell viability or proliferation in four BCL6-expressing B-cell lymphoma lines, although there was modest dose-dependent accumulation of G1 phase cells. Pharmacokinetic studies in mice showed a profile compatible with achieving good levels of target engagement. GSK137, administered orally, suppressed immunoglobulin G responses and reduced numbers of germinal centers and germinal center B cells following immunization of mice with the hapten trinitrophenol. Overall, we report a novel small-molecule BCL6 inhibitor with in vivo activity that inhibits the T-dependent antigen immune response.
Collapse
Affiliation(s)
| | - Mark J Bamford
- GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | - Ruth Barber
- Leicester Drug Discovery and Diagnostics, University of Leicester, Leicester, UK
| | - Angela Bridges
- GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | | | - Constantinos Demetriou
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematological Research Unit, University of Leicester, Leicester, UK
| | - Sian Evans
- Leicester Drug Discovery and Diagnostics, University of Leicester, Leicester, UK
| | | | - David J Hirst
- GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | | | | | - Sandrine Jayne
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematological Research Unit, University of Leicester, Leicester, UK
| | - Larissa Lezina
- Leicester Drug Discovery and Diagnostics, University of Leicester, Leicester, UK; Leicester Cancer Research Centre and Ernest and Helen Scott Haematological Research Unit, University of Leicester, Leicester, UK
| | | | | | - Joanne Morley
- GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | | | | | - Ana Sousa Manso
- Leicester Drug Discovery and Diagnostics, University of Leicester, Leicester, UK; Leicester Cancer Research Centre and Ernest and Helen Scott Haematological Research Unit, University of Leicester, Leicester, UK
| | - Jennifer Schofield
- Leicester Drug Discovery and Diagnostics, University of Leicester, Leicester, UK
| | - Tom Slocombe
- GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | - Don Somers
- GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | - Ann L Walker
- GlaxoSmithKline, Medicines Research Centre, Stevenage, UK
| | | | | | - Simon D Wagner
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematological Research Unit, University of Leicester, Leicester, UK.
| |
Collapse
|
18
|
CD276 is an important player in macrophage recruitment into the tumor and an upstream regulator for PAI-1. Sci Rep 2021; 11:14849. [PMID: 34290311 PMCID: PMC8295264 DOI: 10.1038/s41598-021-94360-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
More than 70% of colorectal, prostate, ovarian, pancreatic and breast cancer specimens show expression of CD276 (B7–H3), a potential immune checkpoint family member. Several studies have shown that high CD276 expression in cancer cells correlates with a poor clinical prognosis. This has been associated with the presence of lower tumor infiltrating leukocytes. Among those, tumor-associated macrophages can comprise up to 50% of the tumor mass and are thought to support tumor growth through various mechanisms. However, a lack of information on CD276 function and interaction partner(s) impedes rigorous evaluation of CD276 as a therapeutic target in oncology. Therefore, we aimed to understand the relevance of CD276 in tumor-macrophage interaction by employing a 3D spheroid coculture system with human cells. Our data show a role for tumor-expressed CD276 on the macrophage recruitment into the tumor spheroid, and also in regulation of the extracellular matrix modulator PAI-1. Furthermore, our experiments focusing on macrophage-expressed CD276 suggest that the antibody-dependent CD276 engagement triggers predominantly inhibitory signaling networks in human macrophages.
Collapse
|
19
|
An Overview on Diffuse Large B-Cell Lymphoma Models: Towards a Functional Genomics Approach. Cancers (Basel) 2021; 13:cancers13122893. [PMID: 34207773 PMCID: PMC8226720 DOI: 10.3390/cancers13122893] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Lymphoma research is a paradigm of integrating basic and applied research within the fields of molecular marker-based diagnosis and therapy. In recent years, major advances in next-generation sequencing have substantially improved the understanding of the genomics underlying diffuse large B-cell lymphoma (DLBCL), the most frequent type of B-cell lymphoma. This review addresses the various approaches that have helped unveil the biology and intricate alterations in this pathology, from cell lines to more sophisticated last-generation experimental models, such as organoids. We also provide an overview of the most recent findings in the field, their potential relevance for designing targeted therapies and the corresponding applicability to personalized medicine. Abstract Lymphoma research is a paradigm of the integration of basic and clinical research within the fields of diagnosis and therapy. Clinical, phenotypic, and genetic data are currently used to predict which patients could benefit from standard treatment. However, alternative therapies for patients at higher risk from refractoriness or relapse are usually empirically proposed, based on trial and error, without considering the genetic complexity of aggressive B-cell lymphomas. This is primarily due to the intricate mosaic of genetic and epigenetic alterations in lymphomas, which are an obstacle to the prediction of which drug will work for any given patient. Matching a patient’s genes to drug sensitivity by directly testing live tissues comprises the “precision medicine” concept. However, in the case of lymphomas, this concept should be expanded beyond genomics, eventually providing better treatment options for patients in need of alternative therapeutic approaches. We provide an overview of the most recent findings in diffuse large B-cell lymphomas genomics, from the classic functional models used to study tumor biology and the response to experimental treatments using cell lines and mouse models, to the most recent approaches with spheroid/organoid models. We also discuss their potential relevance and applicability to daily clinical practice.
Collapse
|
20
|
Cui D, Franz A, Fillon SA, Jannetti L, Isambert T, Fundel-Clemens K, Huber HJ, Viollet C, Ghanem A, Niwa A, Weigle B, Pflanz S. High-Yield Human Induced Pluripotent Stem Cell-Derived Monocytes and Macrophages Are Functionally Comparable With Primary Cells. Front Cell Dev Biol 2021; 9:656867. [PMID: 33937256 PMCID: PMC8080307 DOI: 10.3389/fcell.2021.656867] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/12/2021] [Indexed: 12/17/2022] Open
Abstract
Macrophages are pivotal effectors of host immunity and regulators of tissue homeostasis. Understanding of human macrophage biology has been hampered by the lack of reliable and scalable models for cellular and genetic studies. Human induced pluripotent stem cell (hiPSC)-derived monocytes and macrophages, as an unlimited source of subject genotype-specific cells, will undoubtedly play an important role in advancing our understanding of macrophage biology and implication in human diseases. In this study, we present a fully optimized differentiation protocol of hiPSC-derived monocytes and granulocyte-macrophage colony-stimulating factor (GM-CSF) or macrophage colony-stimulating factor (M-CSF). We present characterization of iPSC-derived myeloid lineage cells at phenotypic, functional, and transcriptomic levels, in comparison with corresponding subsets of peripheral blood-derived cells. We also highlight the application of hiPSC-derived monocytes and macrophages as a gene-editing platform for functional validation in research and drug screening, and the study also provides a reference for cell therapies.
Collapse
Affiliation(s)
- Di Cui
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cancer Immunology and Immune Modulation, Biberach an der Riss, Germany
| | - Alexandra Franz
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cancer Immunology and Immune Modulation, Biberach an der Riss, Germany
| | - Sophie A Fillon
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cancer Immunology and Immune Modulation, Biberach an der Riss, Germany
| | - Linda Jannetti
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cancer Immunology and Immune Modulation, Biberach an der Riss, Germany
| | - Timo Isambert
- Boehringer Ingelheim Pharma GmbH & Co. KG, Medicinal Chemistry, Biberach an der Riss, Germany
| | - Katrin Fundel-Clemens
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Computational Biology and Digital Sciences, Biberach an der Riss, Germany
| | - Heinrich J Huber
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Computational Biology and Digital Sciences, Biberach an der Riss, Germany
| | - Coralie Viollet
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Computational Biology and Digital Sciences, Biberach an der Riss, Germany
| | - Alexander Ghanem
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cancer Immunology and Immune Modulation, Biberach an der Riss, Germany
| | - Akira Niwa
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Bernd Weigle
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cancer Immunology and Immune Modulation, Biberach an der Riss, Germany
| | - Stefan Pflanz
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cancer Immunology and Immune Modulation, Biberach an der Riss, Germany.,Boehringer Ingelheim Pharma GmbH & Co. KG, Venture Fund, Ridgefield, CT, United States
| |
Collapse
|
21
|
Ai Y, Hwang L, MacKerell AD, Melnick A, Xue F. Progress toward B-Cell Lymphoma 6 BTB Domain Inhibitors for the Treatment of Diffuse Large B-Cell Lymphoma and Beyond. J Med Chem 2021; 64:4333-4358. [PMID: 33844535 DOI: 10.1021/acs.jmedchem.0c01686] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
B-cell lymphoma 6 (BCL6) is a master regulator of germinal center formation that produce antibody-secreting plasma cells and memory B-cells for sustained immune responses. The BTB domain of BCL6 (BCL6BTB) forms a homodimer that mediates transcriptional repression by recruiting its corepressor proteins to form a biologically functional transcriptional complex. The protein-protein interaction (PPI) between the BCL6BTB and its corepressors has emerged as a therapeutic target for the treatment of DLBCL and a number of other human cancers. This Perspective provides an overview of recent advances in the development of BCL6BTB inhibitors from reversible inhibitors, irreversible inhibitors, to BCL6 degraders. Inhibitor design and medicinal chemistry strategies for the development of novel compounds will be provided. The binding mode of new inhibitors to BCL6BTB are highlighted. Also, the in vitro and in vivo assays used for the evaluation of new compounds will be discussed.
Collapse
Affiliation(s)
- Yong Ai
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Lucia Hwang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Ari Melnick
- Department of Hematology and Oncology, Weill Cornell Medical College, New York, New York 10021, United States.,Department of Pharmacology, Weill Cornell Medical College, New York, New York 10021, United States
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| |
Collapse
|
22
|
Tsai CC, Su YC, Bamodu OA, Chen BJ, Tsai WC, Cheng WH, Lee CH, Hsieh SM, Liu ML, Fang CL, Lin HT, Chen CL, Yeh CT, Lee WH, Ho CL, Lai SW, Tzeng HE, Hsieh YY, Chang CL, Zheng YM, Liu HW, Yen Y, Whang-Peng J, Chao TY. High-Grade B-Cell Lymphoma (HGBL) with MYC and BCL2 and/or BCL6 Rearrangements Is Predominantly BCL6-Rearranged and BCL6-Expressing in Taiwan. Cancers (Basel) 2021; 13:cancers13071620. [PMID: 33807449 PMCID: PMC8059140 DOI: 10.3390/cancers13071620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 01/15/2023] Open
Abstract
Simple Summary This study highlights the epidemiological, cytogenetic and clinical difference between patients with multiple hit diffuse large B-cell lymphoma in Taiwan and those from western countries. Unlike in the West, the majority of patients with multiple hit lymphoma in Taiwan harbor a BCL6 rearrangement. Almost three in every five BCL6-rearranged double hit lymphoma cases in Taiwan are non-GCB phenotype, indicating, at least in part, that the preferential screening for double hit with BCL6 rearrangement may be a clinically-informative modality for patients with non-GCB phenotype DLBCL in Taiwan. This also suggests the need for a different treatment approach than is obtained in the West where BCL6 double hit lymphomas are seemingly GCB. Consistent with our present findings, mandatory screening for BCL6-rearrangement in suspected DLBCL cases in Taiwan may aid early diagnosis, therapy decision, and clinical outcome forecast. Abstract This study investigated the epidemiological and clinical peculiarities of BCL2 and BCL6 rearrangement in patients with high grade B-cell lymphoma (HGBL) from Taiwan, compared with data from Western countries. Two hundred and eighty-two DLBCL cases from Taipei Medical University-affiliated hospitals (n = 179) and Tri-Service General Hospital (n = 103) were enrolled for this study. From the 282, 47 (16.7%) had MYC translocation; 24 of these harbored concurrent BCL2 and/or BCL6 translocation (double-hit, DH or triple-hit, TH). Twelve DH-HGBL cases had simultaneous MYC and BCL6 translocations, 8 harbored MYC and BCL2 rearrangement, while the remaining 4 patients exhibited TH. Together, 66.7% of DH/TH-HGBL patients were BCL6 rearrangement positive. Among these BCL6-rearranged DH/TH-HGBL patients, only 6 (37.5%) overexpressed MYC and BCL6 proteins simultaneously, indicating that MYC-BCL6 co-overexpression may not be plausible surrogate biomarker for screening BCL6-rearranged DH-HGBL. By the end of year 5, all patients with TH-HGBL, BCL2 DH-HGBL and all but one BCL6 DH-HGBL cases had expired or were lost to follow-up. Progression-free survival (PFS) was longer for the non-DH/TH-HGBL group compared with the DH/TH-HGBL group. While the patients with BCL2 DH-HGBL were lost to follow-up by day 800, their remaining TH-HGBL and BCL6 DH-HGBL peers exhibited very poor PFS, regardless of age strata. More so, patients with BCL6 rearrangement were 5.5-fold more likely associated with extranodal involvement compared with their BCL2-rearranged peers. Moreover, ~60.0% of the BCL6-rearranged DH-HGBL cases were non-GCB, suggesting that including screening for BCL6 rearrangement in patients with the non-GCB phenotype may aid medical decision-making and therapeutic strategy. Contrary to contemporary data from western countries, 2 in every 3 patients with DH/TH-HGBL in Taiwan harbor BCL6 rearrangement. Consistent with present findings, we recommend mandatory screening for BCL6 rearrangement in patients with aggressive HGBL in Taiwan.
Collapse
Affiliation(s)
- Cheng-Chih Tsai
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-C.T.); (Y.-C.S.); (O.A.B.); (W.-H.C.); (C.-T.Y.); (Y.-Y.H.); (H.-W.L.)
| | - Yung-Cheng Su
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-C.T.); (Y.-C.S.); (O.A.B.); (W.-H.C.); (C.-T.Y.); (Y.-Y.H.); (H.-W.L.)
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei City 115, Taiwan
| | - Oluwaseun Adebayo Bamodu
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-C.T.); (Y.-C.S.); (O.A.B.); (W.-H.C.); (C.-T.Y.); (Y.-Y.H.); (H.-W.L.)
- Department of Medical Research and Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Bo-Jung Chen
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (B.-J.C.); (C.-H.L.); (M.-L.L.); (W.-H.L.)
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan;
| | - Wei-Hong Cheng
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-C.T.); (Y.-C.S.); (O.A.B.); (W.-H.C.); (C.-T.Y.); (Y.-Y.H.); (H.-W.L.)
| | - Chii-Hong Lee
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (B.-J.C.); (C.-H.L.); (M.-L.L.); (W.-H.L.)
- Department of Anatomic Pathology, Taipei Institute of Pathology, Taipei City 103, Taiwan
| | - Shu-Min Hsieh
- Department of Clinical Pathology, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan;
| | - Mei-Ling Liu
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (B.-J.C.); (C.-H.L.); (M.-L.L.); (W.-H.L.)
| | - Chia-Lang Fang
- Department of Pathology, Taipei Medical University Hospital, Taipei City 110, Taiwan; (C.-L.F.); (C.-L.C.)
| | - Huan-Tze Lin
- Division of Hematology and Oncology, Department of Medicine, Taipei Medical University Hospital, Taipei City 110, Taiwan; (H.-T.L.); (H.-E.T.)
| | - Chi-Long Chen
- Department of Pathology, Taipei Medical University Hospital, Taipei City 110, Taiwan; (C.-L.F.); (C.-L.C.)
- Department of Pathology, School of Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Chi-Tai Yeh
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-C.T.); (Y.-C.S.); (O.A.B.); (W.-H.C.); (C.-T.Y.); (Y.-Y.H.); (H.-W.L.)
- Department of Medical Research and Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Wei-Hwa Lee
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (B.-J.C.); (C.-H.L.); (M.-L.L.); (W.-H.L.)
| | - Ching-Liang Ho
- Division of Hematology-Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan; (C.-L.H.); (S.-W.L.)
| | - Shiue-Wei Lai
- Division of Hematology-Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan; (C.-L.H.); (S.-W.L.)
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Huey-En Tzeng
- Division of Hematology and Oncology, Department of Medicine, Taipei Medical University Hospital, Taipei City 110, Taiwan; (H.-T.L.); (H.-E.T.)
- Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City 110, Taiwan;
| | - Yao-Yu Hsieh
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-C.T.); (Y.-C.S.); (O.A.B.); (W.-H.C.); (C.-T.Y.); (Y.-Y.H.); (H.-W.L.)
| | - Chia-Lun Chang
- Department of Medicine, Division of Hematology and Oncology, Taipei Medical University-Wan-Fang Hospital, Taipei City 116, Taiwan; (C.-L.C.); (Y.-M.Z.); (J.W.-P.)
| | - Yu-Mei Zheng
- Department of Medicine, Division of Hematology and Oncology, Taipei Medical University-Wan-Fang Hospital, Taipei City 116, Taiwan; (C.-L.C.); (Y.-M.Z.); (J.W.-P.)
| | - Hui-Wen Liu
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-C.T.); (Y.-C.S.); (O.A.B.); (W.-H.C.); (C.-T.Y.); (Y.-Y.H.); (H.-W.L.)
| | - Yun Yen
- Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City 110, Taiwan;
| | - Jacqueline Whang-Peng
- Department of Medicine, Division of Hematology and Oncology, Taipei Medical University-Wan-Fang Hospital, Taipei City 116, Taiwan; (C.-L.C.); (Y.-M.Z.); (J.W.-P.)
- Taipei Cancer Center, Taipei Medical University, Taipei City 110, Taiwan
| | - Tsu-Yi Chao
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-C.T.); (Y.-C.S.); (O.A.B.); (W.-H.C.); (C.-T.Y.); (Y.-Y.H.); (H.-W.L.)
- Division of Hematology-Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan; (C.-L.H.); (S.-W.L.)
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei City 110, Taiwan
- Correspondence: ; Tel.: +886-2-2249-0088 (ext. 8402); Fax: +886-2-6639-7181
| |
Collapse
|
23
|
Small-molecule-induced polymerization triggers degradation of BCL6. Nature 2020; 588:164-168. [PMID: 33208943 PMCID: PMC7816212 DOI: 10.1038/s41586-020-2925-1] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 09/03/2020] [Indexed: 12/23/2022]
Abstract
Effective and sustained inhibition of non-enzymatic oncogenic driver proteins is a major pharmacological challenge. The clinical success of thalidomide analogues demonstrates the therapeutic efficacy of drug-induced degradation of transcription factors and other cancer targets1-3, but a substantial subset of proteins are resistant to targeted degradation using existing approaches4,5. Here we report an alternative mechanism of targeted protein degradation, in which a small molecule induces the highly specific, reversible polymerization of a target protein, followed by its sequestration into cellular foci and subsequent degradation. BI-3802 is a small molecule that binds to the Broad-complex, Tramtrack and Bric-à-brac (BTB) domain of the oncogenic transcription factor B cell lymphoma 6 (BCL6) and leads to the proteasomal degradation of BCL66. We use cryo-electron microscopy to reveal how the solvent-exposed moiety of a BCL6-binding molecule contributes to a composite ligand-protein surface that engages BCL6 homodimers to form a supramolecular structure. Drug-induced formation of BCL6 filaments facilitates ubiquitination by the SIAH1 E3 ubiquitin ligase. Our findings demonstrate that a small molecule such as BI-3802 can induce polymerization coupled to highly specific protein degradation, which in the case of BCL6 leads to increased pharmacological activity compared to the effects induced by other BCL6 inhibitors. These findings open new avenues for the development of therapeutic agents and synthetic biology.
Collapse
|