1
|
Sarkozy C, Tessoulin B, Chiron D. Unraveling MCL biology to understand resistance and identify vulnerabilities. Blood 2025; 145:696-707. [PMID: 38551811 DOI: 10.1182/blood.2023022351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/15/2024] [Indexed: 02/14/2025] Open
Abstract
ABSTRACT Mantle cell lymphoma (MCL) is a rare (5%-7%), aggressive B-cell non-Hodgkin lymphoma with well-defined hallmarks (eg, cyclin D1, SOX11), and its expansion is highly dependent on the tumor microenvironment (TME). Parallel drastic progress in the understanding of lymphomagenesis and improved treatments led to a paradigm shift in this B-cell malignancy with now prolonged disease-free survival after intensive chemotherapy and anti-CD20-based maintenance. However, this toxic strategy is not applicable in frail or older patients, and a small but significant part of the cases present a refractory disease representing unmet medical needs. Importantly, the field has recently seen the rapid emergence of targeted and immune-based strategies with effective combinations relying on biological rationales to overcome malignant plasticity and intratumor heterogeneity. In this review, we expose how unraveling the biology of MCL allows to better understand the therapeutic resistances and to identify neo-vulnerabilities in tumors, which are essential to offer efficient novel strategies for high-risk patients. We first highlight the tumor intrinsic resistance mechanisms and associated Achilles heels within various pathways, such as NF-κB, mitochondrial apoptosis, DNA repair, and epigenetic regulators. We then place the tumor in its complex ecosystem to decipher the dialog with the multiple TME components and show how the resulting protumoral signals could be disrupted with innovative therapeutic strategies. Finally, we discuss how these progresses could be integrated into a personalized approach in MCL.
Collapse
Affiliation(s)
- Clémentine Sarkozy
- Service d'Hématologie, Institut Curie, Saint Cloud, France
- Laboratoire d'Imagerie Translationnelle en Oncologie, U1288 Inserm/Institut Curie Centre de Recherche, Paris, France
| | - Benoit Tessoulin
- Service d'Hématologie, Centre Hospitalier Universitaire Nantes, Nantes, France
- reMoVE-B, Nantes Université, INSERM, Centre National de la Recherche Scientifique, Université d'Angers, CRCI2NA, Nantes, France
| | - David Chiron
- reMoVE-B, Nantes Université, INSERM, Centre National de la Recherche Scientifique, Université d'Angers, CRCI2NA, Nantes, France
| |
Collapse
|
2
|
Zou D, Feng S, Hu B, Guo M, Lv Y, Ma R, Du Y, Feng J. Bromodomain proteins as potential therapeutic targets for B-cell non-Hodgkin lymphoma. Cell Biosci 2024; 14:143. [PMID: 39580422 PMCID: PMC11585172 DOI: 10.1186/s13578-024-01326-1] [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: 06/26/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND B-cell non-Hodgkin lymphoma (B-NHL) is the most common type of lymphoma and is significantly heterogeneous among various subtypes. Despite of considerable advancements in treatment strategies for B-NHL, the prognosis of relapsed/refractory patients remains poor. MAIN TEXT It has been indicated that epigenetic dysregulation is critically associated with the pathogenesis of most hematological malignancies, resulting in the clinical targeting of epigenetic modifications. Bromodomain (BRD) proteins are essential epigenetic regulators which contain eight subfamilies, including BRD and extra-terminal domain (BET) family, histone acetyltransferases (HATs) and HAT-related proteins, transcriptional coactivators, transcriptional mediators, methyltransferases, helicases, ATP-dependent chromatin-remodeling complexes, and nuclear-scaffolding proteins. Most pre-clinical and clinical studies on B-NHL have focused predominantly on the BET family and the use of BET inhibitors as mono-treatment or co-treatment with other anti-tumor drugs. Furthermore, preclinical models of B-NHL have revealed that BET degraders are more active than BET inhibitors. Moreover, with the development of BET inhibitors and degraders, non-BET BRD protein inhibitors have also been designed and have shown antitumor activities in B-NHL preclinical models. This review summarized the mechanism of BRD proteins and the recent progress of BRD protein-related drugs in B-NHL. This study aimed to collect the most recent evidences and summarize possibility on whether BRD proteins can serve as therapeutic targets for B-NHL. CONCLUSION In summary, BRD proteins are critical epigenetic regulatory factors and may be potential therapeutic targets for B-NHL.
Collapse
Affiliation(s)
- Dan Zou
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Sitong Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Bowen Hu
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Mengya Guo
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yan Lv
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Rong Ma
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yuxin Du
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| | - Jifeng Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| |
Collapse
|
3
|
Sartori G, Tarantelli C, Spriano F, Gaudio E, Cascione L, Mascia M, Barreca M, Arribas AJ, Licenziato L, Golino G, Ferragamo A, Pileri S, Damia G, Zucca E, Stathis A, Politz O, Wengner AM, Bertoni F. The ATR inhibitor elimusertib exhibits anti-lymphoma activity and synergizes with the PI3K inhibitor copanlisib. Br J Haematol 2024; 204:191-205. [PMID: 38011941 DOI: 10.1111/bjh.19218] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023]
Abstract
The DNA damage response (DDR) is the cellular process of preserving an intact genome and is often deregulated in lymphoma cells. The ataxia telangiectasia and Rad3-related (ATR) kinase is a crucial factor of DDR in the response to DNA single-strand breaks. ATR inhibitors are agents that have shown considerable clinical potential in this context. We characterized the activity of the ATR inhibitor elimusertib (BAY 1895344) in a large panel of lymphoma cell lines. Furthermore, we evaluated its activity combined with the clinically approved PI3K inhibitor copanlisib in vitro and in vivo. Elimusertib exhibits potent anti-tumour activity across various lymphoma subtypes, which is associated with the expression of genes related to replication stress, cell cycle regulation and, as also sustained by CRISPR Cas9 experiments, CDKN2A loss. In several tumour models, elimusertib demonstrated widespread anti-tumour activity stronger than ceralasertib, another ATR inhibitor. This activity is present in both DDR-proficient and DDR-deficient lymphoma models. Furthermore, a combination of ATR and PI3K inhibition by treatment with elimusertib and copanlisib has in vitro and in vivo anti-tumour activity, providing a potential new treatment option for lymphoma patients.
Collapse
Affiliation(s)
- Giulio Sartori
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Chiara Tarantelli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Filippo Spriano
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Eugenio Gaudio
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Luciano Cascione
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Michele Mascia
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Marilia Barreca
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Alberto J Arribas
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Luca Licenziato
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Gaetanina Golino
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Adele Ferragamo
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Stefano Pileri
- Division of Diagnostic Haematopathology, European Institute of Oncology, Milan, Italy
| | - Giovanna Damia
- Laboratory of Molecular Pharmacology, Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Emanuele Zucca
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, USI, Lugano, Switzerland
| | - Oliver Politz
- Bayer AG, Pharmaceuticals, Research & Development, Berlin, Germany
| | - Antje M Wengner
- Bayer AG, Pharmaceuticals, Research & Development, Berlin, Germany
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| |
Collapse
|
4
|
Wang ZQ, Zhang ZC, Wu YY, Pi YN, Lou SH, Liu TB, Lou G, Yang C. Bromodomain and extraterminal (BET) proteins: biological functions, diseases, and targeted therapy. Signal Transduct Target Ther 2023; 8:420. [PMID: 37926722 PMCID: PMC10625992 DOI: 10.1038/s41392-023-01647-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/23/2023] [Accepted: 09/12/2023] [Indexed: 11/07/2023] Open
Abstract
BET proteins, which influence gene expression and contribute to the development of cancer, are epigenetic interpreters. Thus, BET inhibitors represent a novel form of epigenetic anticancer treatment. Although preliminary clinical trials have shown the anticancer potential of BET inhibitors, it appears that these drugs have limited effectiveness when used alone. Therefore, given the limited monotherapeutic activity of BET inhibitors, their use in combination with other drugs warrants attention, including the meaningful variations in pharmacodynamic activity among chosen drug combinations. In this paper, we review the function of BET proteins, the preclinical justification for BET protein targeting in cancer, recent advances in small-molecule BET inhibitors, and preliminary clinical trial findings. We elucidate BET inhibitor resistance mechanisms, shed light on the associated adverse events, investigate the potential of combining these inhibitors with diverse therapeutic agents, present a comprehensive compilation of synergistic treatments involving BET inhibitors, and provide an outlook on their future prospects as potent antitumor agents. We conclude by suggesting that combining BET inhibitors with other anticancer drugs and innovative next-generation agents holds great potential for advancing the effective targeting of BET proteins as a promising anticancer strategy.
Collapse
Affiliation(s)
- Zhi-Qiang Wang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Zhao-Cong Zhang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Yu-Yang Wu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ya-Nan Pi
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Sheng-Han Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tian-Bo Liu
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Ge Lou
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China.
| | - Chang Yang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China.
| |
Collapse
|
5
|
Spriano F, Sartori G, Tarantelli C, Barreca M, Golino G, Rinaldi A, Napoli S, Mascia M, Scalise L, Arribas AJ, Cascione L, Zucca E, Stathis A, Gaudio E, Bertoni F. Pharmacologic screen identifies active combinations with BET inhibitors and LRRK2 as a novel putative target in lymphoma. EJHAEM 2022; 3:764-774. [PMID: 36051080 PMCID: PMC9422027 DOI: 10.1002/jha2.535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 11/26/2022]
Abstract
Inhibitors of the Bromo- and Extra-Terminal domain (BET) family proteins have strong preclinical antitumor activity in multiple tumor models, including lymphomas. Limited single-agent activity has been reported in the clinical setting. Here, we have performed a pharmacological screening to identify compounds that can increase the antitumor activity of BET inhibitors in lymphomas. The germinal center B-cell like diffuse large B-cell lymphoma (DLBCL) cell lines OCI-LY-19 and WSU-DLCL2 were exposed to 348 compounds given as single agents at two different concentrations and in combination with the BET inhibitor birabresib. The combination partners included small molecules targeting important biologic pathways such as PI3K/AKT/MAPK signaling and apoptosis, approved anticancer agents, kinase inhibitors, epigenetic compounds. The screening identified a series of compounds leading to a stronger antiproliferative activity when given in combination than as single agents: the histone deacetylase (HDAC) inhibitors panobinostat and dacinostat, the mTOR (mechanistic target of rapamycin) inhibitor everolimus, the ABL/SRC (ABL proto-oncogene/SRC proto oncogene) inhibitor dasatinib, the AKT1/2/3 inhibitor MK-2206, the JAK2 inhibitor TG101209. The novel finding was the benefit given by the addition of the LRRK2 inhibitor LRRK2-IN-1, which was validated in vitro and in vivo. Genetic silencing demonstrated that LRRK2 sustains the proliferation of lymphoma cells, a finding paired with the association between high expression levels and inferior outcome in DLBCL patients. We identified combinations that can improve the response to BET inhibitors in lymphomas, and LRRK2 as a gene essential for lymphomas and as putative novel target for this type of tumors.
Collapse
Affiliation(s)
- Filippo Spriano
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Giulio Sartori
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Chiara Tarantelli
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Marilia Barreca
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
- Department of BiologicalChemical and Pharmaceutical Sciences and Technologies (STEBICEF)University of PalermoPalermoItaly
| | - Gaetanina Golino
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Andrea Rinaldi
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Sara Napoli
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Michele Mascia
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Lorenzo Scalise
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Alberto J. Arribas
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
- SIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Luciano Cascione
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
- SIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Emanuele Zucca
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
- Department of OncologyOncology Institute of Southern SwitzerlandEnte Ospedaliero CantonaleBellinzonaSwitzerland
| | - Anastasios Stathis
- Department of OncologyOncology Institute of Southern SwitzerlandEnte Ospedaliero CantonaleBellinzonaSwitzerland
- Faculty of Biomedical SciencesUniversità della Svizzera ItalianaLuganoSwitzerland
| | - Eugenio Gaudio
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Francesco Bertoni
- Institute of Oncology ResearchFaculty of Biomedical SciencesUniversità della Svizzera ItalianaBellinzonaSwitzerland
- Department of OncologyOncology Institute of Southern SwitzerlandEnte Ospedaliero CantonaleBellinzonaSwitzerland
| |
Collapse
|
6
|
Wang LW, Jiang S, Yuan YH, Duan J, Mao ND, Hui Z, Bai R, Xie T, Ye XY. Recent Advances in Synergistic Antitumor Effects Exploited from the Inhibition of Ataxia Telangiectasia and RAD3-Related Protein Kinase (ATR). Molecules 2022; 27:molecules27082491. [PMID: 35458687 PMCID: PMC9029554 DOI: 10.3390/molecules27082491] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/27/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
As one of the key phosphatidylinositol 3-kinase-related kinases (PIKKs) family members, ataxia telangiectasia and RAD3-related protein kinase (ATR) is crucial in maintaining mammalian cell genomic integrity in DNA damage response (DDR) and repair pathways. Dysregulation of ATR has been found across different cancer types. In recent years, the inhibition of ATR has been proven to be effective in cancer therapy in preclinical and clinical studies. Importantly, tumor-specific alterations such as ATM loss and Cyclin E1 (CCNE1) amplification are more sensitive to ATR inhibition and are being exploited in synthetic lethality (SL) strategy. Besides SL, synergistic anticancer effects involving ATRi have been reported in an increasing number in recent years. This review focuses on the recent advances in different forms of synergistic antitumor effects, summarizes the pharmacological benefits and ongoing clinical trials behind the biological mechanism, and provides perspectives for future challenges and opportunities. The hope is to draw awareness to the community that targeting ATR should have great potential in developing effective anticancer medicines.
Collapse
Affiliation(s)
- Li-Wei Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Songwei Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Ying-Hui Yuan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Jilong Duan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Nian-Dong Mao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Zi Hui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (R.B.); (T.X.); (X.-Y.Y.); Tel.: +86-571-28860236 (X.-Y.Y.)
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (R.B.); (T.X.); (X.-Y.Y.); Tel.: +86-571-28860236 (X.-Y.Y.)
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (R.B.); (T.X.); (X.-Y.Y.); Tel.: +86-571-28860236 (X.-Y.Y.)
| |
Collapse
|
7
|
Vann KR, Pal D, Smith AL, Sahar NE, Krishnaiah M, El-Gamal D, Kutateladze TG. Combinatorial inhibition of BTK, PI3K-AKT and BRD4-MYC as a strategy for treatment of mantle cell lymphoma. MOLECULAR BIOMEDICINE 2022; 3:2. [PMID: 35031886 PMCID: PMC8760370 DOI: 10.1186/s43556-021-00066-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/17/2021] [Indexed: 11/10/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a subtype of non-Hodgkin's lymphoma characterized by poor prognosis. The complexity of MCL pathogenesis arises from aberrant activities of diverse signaling pathways, including BTK, PI3K-AKT-mTOR and MYC-BRD4. Here, we report that MCL-related signaling pathways can be altered by a single small molecule inhibitor, SRX3305. Binding and kinase activities along with resonance changes in NMR experiments reveal that SRX3305 targets both bromodomains of BRD4 and is highly potent in inhibition of the PI3K isoforms α, γ and δ, as well as BTK and the drug-resistant BTK mutant. Preclinical investigations herein reveal that SRX3305 perturbs the cell cycle, promotes apoptosis in MCL cell lines and shows dose dependent anti-proliferative activity in both MCL and drug-resistant MCL cells. Our findings underscore the effectiveness of novel multi-action small molecule inhibitors for potential treatment of MCL.
Collapse
Affiliation(s)
- Kendra R Vann
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Dhananjaya Pal
- Molecular Targeted Therapeutics Laboratory, Levine Cancer Institute, Charlotte, NC, USA
- Division of Hematology and Oncology, Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Audrey L Smith
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Namood-E Sahar
- Division of Hematology and Oncology, Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Maddeboina Krishnaiah
- Molecular Targeted Therapeutics Laboratory, Levine Cancer Institute, Charlotte, NC, USA
| | - Dalia El-Gamal
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tatiana G Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
8
|
BET Proteins as Attractive Targets for Cancer Therapeutics. Int J Mol Sci 2021; 22:ijms222011102. [PMID: 34681760 PMCID: PMC8538173 DOI: 10.3390/ijms222011102] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/04/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Transcriptional dysregulation is a hallmark of cancer and can be an essential driver of cancer initiation and progression. Loss of transcriptional control can cause cancer cells to become dependent on certain regulators of gene expression. Bromodomain and extraterminal domain (BET) proteins are epigenetic readers that regulate the expression of multiple genes involved in carcinogenesis. BET inhibitors (BETis) disrupt BET protein binding to acetylated lysine residues of chromatin and suppress the transcription of various genes, including oncogenic transcription factors. Phase I and II clinical trials demonstrated BETis’ potential as anticancer drugs against solid tumours and haematological malignancies; however, their clinical success was limited as monotherapies. Emerging treatment-associated toxicities, drug resistance and a lack of predictive biomarkers limited BETis’ clinical progress. The preclinical evaluation demonstrated that BETis synergised with different classes of compounds, including DNA repair inhibitors, thus supporting further clinical development of BETis. The combination of BET and PARP inhibitors triggered synthetic lethality in cells with proficient homologous recombination. Mechanistic studies revealed that BETis targeted multiple essential homologous recombination pathway proteins, including RAD51, BRCA1 and CtIP. The exact mechanism of BETis’ anticancer action remains poorly understood; nevertheless, these agents provide a novel approach to epigenome and transcriptome anticancer therapy.
Collapse
|
9
|
Shorstova T, Foulkes WD, Witcher M. Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer 2021; 124:1478-1490. [PMID: 33723398 PMCID: PMC8076232 DOI: 10.1038/s41416-021-01321-0] [Citation(s) in RCA: 220] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/12/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
The transcriptional upregulation of oncogenes is a driving force behind the progression of many tumours. However, until a decade ago, the concept of 'switching off' these oncogenic pathways represented a formidable challenge. Research has revealed that members of the bromo- and extra-terminal domain (BET) motif family are key activators of oncogenic networks in a spectrum of cancers; their function depends on their recruitment to chromatin through two bromodomains (BD1 and BD2). The advent of potent inhibitors of BET proteins (BETi), which target either one or both bromodomains, represents an important step towards the goal of suppressing oncogenic networks within tumours. Here, we discuss the biology of BET proteins, advances in BETi design and highlight potential biomarkers predicting their activity. We also outline the logic of incorporating BETi into combination therapies to enhance its efficacy. We suggest that understanding mechanisms of activity, defining predictive biomarkers and identifying potent synergies represents a roadmap for clinical success using BETi.
Collapse
Affiliation(s)
- Tatiana Shorstova
- grid.414980.00000 0000 9401 2774Departments of Oncology and Experimental Medicine, McGill University, Lady Davis Institute and Segal Cancer Centre, Jewish General Hospital, Montreal, QC Canada
| | - William D. Foulkes
- grid.414980.00000 0000 9401 2774Departments of Oncology and Human Genetics, McGill University, Lady Davis Institute and Segal Cancer Centre, Jewish General Hospital, Montreal, QC Canada
| | - Michael Witcher
- grid.414980.00000 0000 9401 2774Departments of Oncology and Experimental Medicine, McGill University, Lady Davis Institute and Segal Cancer Centre, Jewish General Hospital, Montreal, QC Canada
| |
Collapse
|
10
|
Genomic profiling of platinum-resistant ovarian cancer: The road into druggable targets. Semin Cancer Biol 2020; 77:29-41. [PMID: 33161141 DOI: 10.1016/j.semcancer.2020.10.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023]
Abstract
Ovarian cancer is the most lethal gynecologic cancer. High-grade serous carcinoma (HGSC) is the most frequent histologic subtype and while it is a highly platinum-sensitive cancer at initial treatment, nearly 90 % of stage IIIC patients recur in 5 years and eventually become resistant to platinum treatment. Historically, the definition of platinum-resistant disease is based on the time interval between last platinum therapy and recurrence shorter than 6 months. Nowadays the use of sophisticated imaging techniques and serum markers to detect recurrence makes the accuracy of this clinical definition less clear and even more debatable as we begin to better understand the molecular landscape of HGSC and markers of platinum resistance and sensitivity. HGSC is characterized by a low frequency of recurrent mutations, great genomic instability with widespread copy number variations, universal TP53 mutations, and homologous recombination deficiency in more than 50 % of cases. Platinum agents form DNA adducts and intra- and inter-strand cross-links in the DNA. Most of DNA repair pathways are involved at some point in the repair of platinum induced DNA damaging, most notably homologous recombination, Fanconi Anemia, and nucleotide excision repair pathways. Mechanisms of platinum resistance are related mostly to the limitation of platinum-DNA adduct formation by changing cellular pharmacology, and to the prevention of cell death after DNA damage due to alterations in DNA repair pathways and cell cycle regulation. Understanding these mechanisms of sensitivity and resistance may help to define the utility of platinum re-challenge in each situation and guide new therapeutic opportunities. Moreover, the discovery of mechanisms of synthetic lethality related to alterations in DNA repair and cell cycle regulation pathways has opened up a new avenue for drug therapy in the last decade. In the present article, we review pathways involved in platinum-induced DNA damage repair and their relationship with genomic alterations present in HGSC. Moreover, we report new treatment strategies that are underway to target these alterations.
Collapse
|
11
|
Carrassa L, Colombo I, Damia G, Bertoni F. Targeting the DNA damage response for patients with lymphoma: Preclinical and clinical evidences. Cancer Treat Rev 2020; 90:102090. [DOI: 10.1016/j.ctrv.2020.102090] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
|
12
|
Klener P. Mantle cell lymphoma: insights into therapeutic targets at the preclinical level. Expert Opin Ther Targets 2020; 24:1029-1045. [PMID: 32842810 DOI: 10.1080/14728222.2020.1813718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Mantle cell lymphoma (MCL) is a chronically relapsing B-cell non-Hodgkin lymphoma characterized by recurrent molecular-cytogenetic aberrations that lead to deregulation of DNA damage response, cell cycle progression, epigenetics, apoptosis, proliferation, and motility. In the last 10 years, clinical approval of several innovative drugs dramatically changed the landscape of treatment options in the relapsed/refractory (R/R) MCL, which translated into significantly improved survival parameters. AREAS COVERED Here, up-to-date knowledge on the biology of MCL together with currently approved and clinically tested frontline and salvage therapies are reviewed. In addition, novel therapeutic targets in MCL based on the scientific reports published in Pubmed are discussed. EXPERT OPINION Bruton tyrosine-kinase inhibitors, NFkappaB inhibitors, BCL2 inhibitors, and immunomodulary agents in combination with monoclonal antibodies and genotoxic drugs have the potential to induce long-term remissions in majority of newly diagnosed MCL patients. Several other classes of anti-tumor drugs including phosphoinositole-3-kinase, cyclin-dependent kinase or DNA damage response kinase inhibitors have demonstrated promising anti-lymphoma efficacy in R/R MCL. Most importantly, adoptive immunotherapy with genetically modified T-cells carrying chimeric antigen receptor represents a potentially curative treatment approach even in the patients with chemotherapy and ibrutinib-refractory disease.
Collapse
Affiliation(s)
- Pavel Klener
- First Department of Internal Medicine- Hematology, University General Hospital and First Faculty of Medicine, Charles University , Prague, Czech Republic.,Institute of Pathological Physiology, First Faculty of Medicine, Charles University , Prague, Czech Republic
| |
Collapse
|
13
|
Spriano F, Stathis A, Bertoni F. Targeting BET bromodomain proteins in cancer: The example of lymphomas. Pharmacol Ther 2020; 215:107631. [PMID: 32693114 DOI: 10.1016/j.pharmthera.2020.107631] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022]
Abstract
The Bromo- and Extra-Terminal domain (BET) family proteins act as "readers" of acetylated histones and they are important transcription regulators. BRD2, BRD3, BRD4 and BRDT, part of the BET family, are important in different tumors, where upregulation or translocation often occurs. The potential of targeting BET proteins as anti-cancer treatment originated with data obtained with a first series of compounds, and there are now several data supporting BET inhibition in both solid tumors and hematological malignancies. Despite very positive preclinical data in different tumor types, the clinical results have been so far moderate. Using lymphoma as an example to review the data produced in the laboratory and in the context of the early clinical trials, we discuss the modalities to make BET targeting more efficient both generating novel generation of compounds and by exploring the combination with small molecules affecting various signaling pathways, BCL2, or DNA damage response signaling, but also with additional epigenetic agents and with immunotherapy. We also discuss the mechanisms of resistance and the toxicity profiles so far reported.
Collapse
Affiliation(s)
- Filippo Spriano
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Faculty of Biomedical Sciences, USI, Lugano, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, Bellinzona, Switzerland.
| |
Collapse
|
14
|
Priebe V, Sartori G, Napoli S, Chung EYL, Cascione L, Kwee I, Arribas AJ, Mensah AA, Rinaldi A, Ponzoni M, Zucca E, Rossi D, Efremov D, Lenz G, Thome M, Bertoni F. Role of ETS1 in the Transcriptional Network of Diffuse Large B Cell Lymphoma of the Activated B Cell-Like Type. Cancers (Basel) 2020; 12:cancers12071912. [PMID: 32679859 PMCID: PMC7409072 DOI: 10.3390/cancers12071912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 01/08/2023] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is a heterogenous disease that has been distinguished into at least two major molecular entities, the germinal center-like B cell (GCB) DLBCL and activated-like B cell (ABC) DLBCL, based on transcriptome expression profiling. A recurrent ch11q24.3 gain is observed in roughly a fourth of DLBCL cases resulting in the overexpression of two ETS transcription factor family members, ETS1 and FLI1. Here, we knocked down ETS1 expression by siRNA and analyzed expression changes integrating them with ChIP-seq data to identify genes directly regulated by ETS1. ETS1 silencing affected expression of genes involved in B cell signaling activation, B cell differentiation, cell cycle, and immune processes. Integration of RNA-Seq (RNA sequencing) data and ChIP-Seq (chromatin immunoprecipitation sequencing) identified 97 genes as bona fide, positively regulated direct targets of ETS1 in ABC-DLBCL. Among these was the Fc receptor for IgM, FCMR (also known as FAIM3 or Toso), which showed higher expression in ABC- than GCB-DLBCL clinical specimens. These findings show that ETS1 is contributing to the lymphomagenesis in a subset of DLBCL and identifies FCMR as a novel target of ETS1, predominantly expressed in ABC-DLBCL.
Collapse
Affiliation(s)
- Valdemar Priebe
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Giulio Sartori
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Sara Napoli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Elaine Yee Lin Chung
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Luciano Cascione
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Ivo Kwee
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
- Dalle Molle Institute for Artificial Intelligence (IDSIA), 6928 Manno, Switzerland
| | - Alberto Jesus Arribas
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Afua Adjeiwaa Mensah
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Andrea Rinaldi
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Maurilio Ponzoni
- San Raffaele Scientific Institute, Vita Salute University, 20132 Milan, Italy;
| | - Emanuele Zucca
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland
| | - Davide Rossi
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland
| | - Dimitar Efremov
- Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy;
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany;
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland;
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland
- Correspondence: ; Tel.: +41-91-8200-367; Fax: +41-91-8200-397
| |
Collapse
|
15
|
Sarkozy C, Ribrag V. Novel agents for mantle cell lymphoma: molecular rational and clinical data. Expert Opin Investig Drugs 2020; 29:555-566. [PMID: 32321318 DOI: 10.1080/13543784.2020.1760245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Mantle cell lymphoma (MCL) is an aggressive B cell non-Hodgkin lymphoma (NHL) that is characterized by the translocation t(11;14)(q13;q32) and a poor response to rituximab-anthracycline-based chemotherapy. Intensive regimens offer durable response, but a subgroup of MCL patients will not be eligible for those regimens and hence are candidates for less toxic, novel therapies based on a more tailored personalized approach. AREAS COVERED This article examines the molecular landscape of MCL, drug resistance mechanisms, and the data on emerging targeted therapies. EXPERT OPINION DNA damage pathway, ATM mutation, TP53, and epigenetic abnormalities are key drivers of MCL. sBCL2, PARP, ATR, CDK inhibitors or epigenetic modifiers are among the most promising drugs under investigation in clinical trials. The genomic landscape of MCL suggests two types of disease based on the presence of ATM or TP53 alterations which should be the framework of future molecular driven strategies. Among novel drugs, those interacting with the DNA damage response pathway offer the most effective rational for their use in MCL.
Collapse
Affiliation(s)
- Clémentine Sarkozy
- Centre National de la Recherche UMR 5286, Centre de Recherche en Cancérologie de lyon, INSERM Unité Mixte de Recherche (UMR)-S1052 , Lyon, France
| | | |
Collapse
|
16
|
Gonzalez-Santamarta M, Quinet G, Reyes-Garau D, Sola B, Roué G, Rodriguez MS. Resistance to the Proteasome Inhibitors: Lessons from Multiple Myeloma and Mantle Cell Lymphoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:153-174. [PMID: 32274756 DOI: 10.1007/978-3-030-38266-7_6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Since its introduction in the clinics in early 2000s, the proteasome inhibitor bortezomib (BTZ) significantly improved the prognosis of patients with multiple myeloma (MM) and mantle cell lymphoma (MCL), two of the most challenging B cell malignancies in western countries. However, relapses following BTZ therapy are frequent, while primary resistance to this agent remains a major limitation for further development of its therapeutic potential. In the present chapter, we recapitulate the molecular mechanisms associated with intrinsic and acquired resistance to BTZ learning from MM and MCL experience, including mutations of crucial genes and activation of prosurvival signalling pathways inherent to malignant B cells. We also outline the preclinical and clinical evaluations of some potential druggable targets associated to BTZ resistance, considering the most meaningful findings of the past 10 years. Although our understanding of BTZ resistance is far from being completed, recent discoveries are contributing to develop new approaches to treat relapsed MM and MCL patients.
Collapse
Affiliation(s)
| | | | - Diana Reyes-Garau
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona (Barcelona), Spain
| | - Brigitte Sola
- Normandie University, INSERM UMR1245, UNICAEN, Caen, France
| | - Gaël Roué
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona (Barcelona), Spain
| | | |
Collapse
|
17
|
Letson C, Padron E. Non-canonical transcriptional consequences of BET inhibition in cancer. Pharmacol Res 2019; 150:104508. [PMID: 31698067 DOI: 10.1016/j.phrs.2019.104508] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/12/2019] [Accepted: 10/21/2019] [Indexed: 01/01/2023]
Abstract
Inhibition of the bromo and extra-terminal domain (BET) protein family in preclinical studies has demonstrated that BET proteins are critical for cancer progression and important therapeutic targets. Downregulation of the MYC oncogene, CDK6, BCL2 and FOSL1 are just a few examples of the effects of BET inhibitors that can lead to cell cycle arrest and apoptosis in cancer cells. However, BET inhibitors have had little success in the clinic as a single agent, and there are an increasing number of reports of resistance to BET inhibition emerging after sustained treatment of cancer cells in vitro. Here we summarize the non-canonical consequences of BET inhibition in cancer, and discuss how these may both lead to resistance and inform rational combinations that could greatly enhance the clinical application of these inhibitors.
Collapse
Affiliation(s)
- Christopher Letson
- Moffitt Cancer Center: 12902 USF Magnolia Drive, Tampa, FL 33612, United States.
| | - Eric Padron
- Moffitt Cancer Center: 12902 USF Magnolia Drive, Tampa, FL 33612, United States.
| |
Collapse
|
18
|
Spriano F, Chung EYL, Gaudio E, Tarantelli C, Cascione L, Napoli S, Jessen K, Carrassa L, Priebe V, Sartori G, Graham G, Selvanathan SP, Cavalli A, Rinaldi A, Kwee I, Testoni M, Genini D, Ye BH, Zucca E, Stathis A, Lannutti B, Toretsky JA, Bertoni F. The ETS Inhibitors YK-4-279 and TK-216 Are Novel Antilymphoma Agents. Clin Cancer Res 2019; 25:5167-5176. [PMID: 31182435 DOI: 10.1158/1078-0432.ccr-18-2718] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/18/2019] [Accepted: 05/31/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Transcription factors are commonly deregulated in cancer, and they have been widely considered as difficult to target due to their nonenzymatic mechanism of action. Altered expression levels of members of the ETS-transcription factors are often observed in many different tumors, including lymphomas. Here, we characterized two small molecules, YK-4-279 and its clinical derivative, TK-216, targeting ETS factors via blocking the protein-protein interaction with RNA helicases, for their antilymphoma activity. EXPERIMENTAL DESIGN The study included preclinical in vitro activity screening on a large panel of cell lines, both as single agent and in combination; validation experiments on in vivo models; and transcriptome and coimmunoprecipitation experiments. RESULTS YK-4-279 and TK-216 demonstrated an antitumor activity across several lymphoma cell lines, which we validated in vivo. We observed synergistic activity when YK-4-279 and TK-216 were combined with the BCL2 inhibitor venetoclax and with the immunomodulatory drug lenalidomide. YK-4-279 and TK-216 interfere with protein interactions of ETS family members SPIB, in activated B-cell-like type diffuse large B-cell lymphomas, and SPI1, in germinal center B-cell-type diffuse large B-cell lymphomas. CONCLUSIONS The ETS inhibitor YK-4-279 and its clinical derivative TK-216 represent a new class of agents with in vitro and in vivo antitumor activity in lymphomas. Although their detailed mechanism of action needs to be fully defined, in DLBCL they might act by targeting subtype-specific essential transcription factors.
Collapse
Affiliation(s)
- Filippo Spriano
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Elaine Yee Lin Chung
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Eugenio Gaudio
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Chiara Tarantelli
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Luciano Cascione
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sara Napoli
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | | | - Laura Carrassa
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Valdemar Priebe
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Giulio Sartori
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Garrett Graham
- Departments of Oncology and Pediatrics, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Saravana P Selvanathan
- Departments of Oncology and Pediatrics, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Andrea Cavalli
- Università della Svizzera italiana, Institute of Biomedical Research, Bellinzona, Switzerland
| | - Andrea Rinaldi
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Ivo Kwee
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Dalle Molle Institute for Artificial Intelligence, Manno, Switzerland
| | - Monica Testoni
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - Davide Genini
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland
| | - B Hilda Ye
- Department of Cell Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, New York
| | - Emanuele Zucca
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | | | | | - Jeffrey A Toretsky
- Departments of Oncology and Pediatrics, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Francesco Bertoni
- Università della Svizzera italiana, Institute of Oncology Research, Bellinzona, Switzerland.
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Pharmacological inhibition of Bromodomain and Extra-Terminal (BET) domain proteins is a very exciting epigenetic therapeutic modality. Due to the central role of BET proteins in transcription regulation, their inhibition heavily affects lymphoma cells and BET inhibitors show a clear preclinical antitumor activity as single agents and in combination, paired with early reports of clinical activity. RECENT FINDINGS Relevant data have been recently presented on the mechanism of action of the BET inhibitors, on modalities to improve their activity in lymphomas, and their clinical evaluation. SUMMARY There are now plenty of preclinical data sustaining BET proteins as therapeutic targets in lymphomas. Newer compounds and combinations with other agents may be pursued in the future aiming also to identify those patients that they most likely benefit from BET inhibition.
Collapse
|
20
|
Boi M, Todaro M, Vurchio V, Yang SN, Moon J, Kwee I, Rinaldi A, Pan H, Crescenzo R, Cheng M, Cerchietti L, Elemento O, Riveiro ME, Cvitkovic E, Bertoni F, Inghirami G. Therapeutic efficacy of the bromodomain inhibitor OTX015/MK-8628 in ALK-positive anaplastic large cell lymphoma: an alternative modality to overcome resistant phenotypes. Oncotarget 2016; 7:79637-79653. [PMID: 27793034 PMCID: PMC5346742 DOI: 10.18632/oncotarget.12876] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/19/2016] [Indexed: 01/08/2023] Open
Abstract
Anaplastic large cell lymphomas (ALCL) represent a peripheral T-cell lymphoma subgroup, stratified based on the presence or absence of anaplastic lymphoma kinase (ALK) chimeras. Although ALK-positive ALCLs have a more favorable outcome than ALK-negative ALCL, refractory and/or relapsed forms are common and novel treatments are needed. Here we investigated the therapeutic potential of a novel bromodomain inhibitor, OTX015/MK-8628 in ALK-positive ALCLs.The effects of OTX015 on a panel of ALK+ ALCL cell lines was evaluated in terms of proliferation, cell cycle and downstream signaling, including gene expression profiling analyses. Synergy was tested with combination targeted therapies.Bromodomain inhibition with OTX015 led primarily to ALCL cell cycle arrest in a dose-dependent manner, along with downregulation of MYC and its downstream regulated genes. MYC overexpression did not compensate this OTX015-mediated phenotype. Transcriptomic analysis of OTX015-treated ALCL cells identified a gene signature common to various hematologic malignancies treated with bromodomain inhibitors, notably large cell lymphoma. OTX015-modulated genes included transcription factors (E2F2, NFKBIZ, FOS, JUNB, ID1, HOXA5 and HOXC6), members of multiple signaling pathways (ITK, PRKCH, and MKNK2), and histones (clusters 1-3). Combination of OTX015 with the Bruton's tyrosine kinase (BTK) inhibitor ibrutinib led to cell cycle arrest then cell death, and combination with suboptimal doses of the ALK inhibitor CEP28122 caused cell cycle arrest. When OTX015 was associated with GANT61, a selective GLI1/2 inhibitor, C1156Y-resistant ALK ALCL growth was impaired.These findings support OTX015 clinical trials in refractory ALCL in combination with inhibitors of interleukin-2-inducible kinase or SHH/GLI1.
Collapse
MESH Headings
- Acetanilides/pharmacology
- Anaplastic Lymphoma Kinase
- Antineoplastic Agents/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Biomarkers, Tumor/genetics
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm/drug effects
- Drug Synergism
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Heterocyclic Compounds, 3-Ring/pharmacology
- Humans
- Inhibitory Concentration 50
- Lymphoma, Large-Cell, Anaplastic/drug therapy
- Lymphoma, Large-Cell, Anaplastic/genetics
- Lymphoma, Large-Cell, Anaplastic/pathology
- Phenotype
- Receptor Protein-Tyrosine Kinases/genetics
- Signal Transduction/drug effects
- Time Factors
- Transcriptome
Collapse
Affiliation(s)
- Michela Boi
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino, Italy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maria Todaro
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino, Italy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Valentina Vurchio
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino, Italy
| | - Shao Ning Yang
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - John Moon
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ivo Kwee
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
- Dalle Molle Institute for Artificial Intelligence (IDSIA), Manno, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Andrea Rinaldi
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
| | - Heng Pan
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
- Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ramona Crescenzo
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino, Italy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Mangeng Cheng
- In Vitro Pharmacology, Merck Research Laboratory, Boston, MA, USA
| | - Leandro Cerchietti
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Olivier Elemento
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
- Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Esteban Cvitkovic
- Oncology Therapeutic Development, Clichy, France
- Oncoethix SA (Now Oncoethix GmbH, A Wholly Owned Subsidiary of Merck Sharp & Dohme Corp.), Lucerne, Switzerland
| | - Francesco Bertoni
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
- IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Giorgio Inghirami
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino, Italy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Pathology, and NYU Cancer Center, New York University School of Medicine, New York, NY, USA
| | | |
Collapse
|