101
|
Fan F, Podar K. The Role of AP-1 Transcription Factors in Plasma Cell Biology and Multiple Myeloma Pathophysiology. Cancers (Basel) 2021; 13:2326. [PMID: 34066181 PMCID: PMC8151277 DOI: 10.3390/cancers13102326] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (MM) is an incurable hematologic malignancy characterized by the clonal expansion of malignant plasma cells within the bone marrow. Activator Protein-1 (AP-1) transcription factors (TFs), comprised of the JUN, FOS, ATF and MAF multigene families, are implicated in a plethora of physiologic processes and tumorigenesis including plasma cell differentiation and MM pathogenesis. Depending on the genetic background, the tumor stage, and cues of the tumor microenvironment, specific dimeric AP-1 complexes are formed. For example, AP-1 complexes containing Fra-1, Fra-2 and B-ATF play central roles in the transcriptional control of B cell development and plasma cell differentiation, while dysregulation of AP-1 family members c-Maf, c-Jun, and JunB is associated with MM cell proliferation, survival, drug resistance, bone marrow angiogenesis, and bone disease. The present review article summarizes our up-to-date knowledge on the role of AP-1 family members in plasma cell differentiation and MM pathophysiology. Moreover, it discusses novel, rationally derived approaches to therapeutically target AP-1 TFs, including protein-protein and protein-DNA binding inhibitors, epigenetic modifiers and natural products.
Collapse
Affiliation(s)
- Fengjuan Fan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China;
| | - Klaus Podar
- Department of Internal Medicine II, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr.-Karl-Dorrek-Strasse 30, 3500 Krems an der Donau, Austria
| |
Collapse
|
102
|
Bohl SR, Schmalbrock LK, Bauhuf I, Meyer T, Dolnik A, Szyska M, Blätte TJ, Knödler S, Röhner L, Miller D, Kull M, Langer C, Döhner H, Letai A, Damm F, Heckl D, Bullinger L, Krönke J. Comprehensive CRISPR-Cas9 screens identify genetic determinants of drug responsiveness in multiple myeloma. Blood Adv 2021; 5:2391-2402. [PMID: 33950175 PMCID: PMC8114551 DOI: 10.1182/bloodadvances.2020003541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
The introduction of new drugs in the past years has substantially improved outcome in multiple myeloma (MM). However, the majority of patients eventually relapse and become resistant to one or multiple drugs. While the genetic landscape of relapsed/ resistant multiple myeloma has been elucidated, the causal relationship between relapse-specific gene mutations and the sensitivity to a given drug in MM has not systematically been evaluated. To determine the functional impact of gene mutations, we performed combined whole-exome sequencing (WES) of longitudinal patient samples with CRISPR-Cas9 drug resistance screens for lenalidomide, bortezomib, dexamethasone, and melphalan. WES of longitudinal samples from 16 MM patients identified a large number of mutations in each patient that were newly acquired or evolved from a small subclone (median 9, range 1-55), including recurrent mutations in TP53, DNAH5, and WSCD2. Focused CRISPR-Cas9 resistance screens against 170 relapse-specific mutations functionally linked 15 of them to drug resistance. These included cereblon E3 ligase complex members for lenalidomide, structural genes PCDHA5 and ANKMY2 for dexamethasone, RB1 and CDK2NC for bortezomib, and TP53 for melphalan. In contrast, inactivation of genes involved in the DNA damage repair pathway, including ATM, FANCA, RAD54B, and BRCC3, enhanced susceptibility to cytotoxic chemotherapy. Resistance patterns were highly drug specific with low overlap and highly correlated with the treatment-dependent clonal evolution in patients. The functional association of specific genetic alterations with drug sensitivity will help to personalize treatment of MM in the future.
Collapse
Affiliation(s)
- Stephan R Bohl
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Laura K Schmalbrock
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Imke Bauhuf
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Tatjana Meyer
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Anna Dolnik
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin Szyska
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tamara J Blätte
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sarah Knödler
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Linda Röhner
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Denise Miller
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Miriam Kull
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Christian Langer
- Department of Hematology, Internal Oncology and Palliative Care, Kempten Hospital, Kempten, Germany; and
| | - Hartmut Döhner
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Frederik Damm
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dirk Heckl
- Department of Hematology and Oncology Children's Hospital, Halle University Hospital, Halle, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jan Krönke
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
103
|
Current and emerging therapies for primary central nervous system lymphoma. Biomark Res 2021; 9:32. [PMID: 33957995 PMCID: PMC8101140 DOI: 10.1186/s40364-021-00282-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/07/2021] [Indexed: 12/26/2022] Open
Abstract
Primary central nervous system (CNS) lymphoma (PCNSL) is a rare type of extranodal lymphoma exclusively involving the CNS at the onset, with diffuse large B-cell lymphoma (DLBCL) as the most common histological subtype. As PCNSL is a malignancy arising in an immune-privileged site, suboptimal delivery of systemic agents into tumor tissues results in poorer outcomes in PCNSL than in non-CNS DLBCLs. Commonly used regimens for PCNSL include high-dose methotrexate-based chemotherapy with rituximab for induction therapy and intensive chemotherapy followed by autologous hematopoietic stem cell transplantation or whole-brain radiotherapy for consolidation therapy. Targeted agents against the B-cell receptor signaling pathway, microenvironment immunomodulation and blood-brain barrier (BBB) permeabilization appear to be promising in treating refractory/relapsed patients. Chimeric antigen receptor-T cells (CAR-T cells) have been shown to penetrate the BBB as a potential tool to manipulate this disease entity while controlling CAR-T cell-related encephalopathy syndrome. Future approaches may stratify patients according to age, performance status, molecular biomarkers and cellular bioinformation. This review summarizes the current therapies and emerging agents in clinical development for PCNSL treatment.
Collapse
|
104
|
Meermeier EW, Welsh SJ, Sharik ME, Du MT, Garbitt VM, Riggs DL, Shi CX, Stein CK, Bergsagel M, Chau B, Wheeler ML, Bezman N, Wang F, Strop P, Leif Bergsagel P, Chesi M. Tumor burden limits bispecific antibody efficacy through T cell exhaustion averted by concurrent cytotoxic therapy. Blood Cancer Discov 2021; 2:354-369. [PMID: 34258584 DOI: 10.1158/2643-3230.bcd-21-0038] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BCMA-CD3-targeting bispecific antibodies (BsAb) are a recently developed immunotherapy class which shows potent tumor killing activity in multiple myeloma (MM). Here, we investigated a murine BCMA-CD3-targeting BsAb in the immunocompetent Vk*MYC and its IMiD-sensitive derivative Vk*MYChCRBN models of MM. The BCMA-CD3 BsAb was safe and efficacious in a subset of mice, but failed in those with high-tumor burden, consistent with clinical reports of BsAb in leukemia. The combination of BCMA-CD3 BsAb with pomalidomide expanded lytic T cells and improved activity even in IMiD resistant high-tumor burden cases. Yet, survival was only marginally extended due to acute toxicity and T cell exhaustion, which impaired T cell persistence. In contrast, the combination with cyclophosphamide was safe and allowed for a tempered pro-inflammatory response associated with long-lasting complete remission. Concurrent cytotoxic therapy with BsAb actually improved T cell persistence and function, offering a promising approach to patients with a large tumor burden.
Collapse
Affiliation(s)
- Erin W Meermeier
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Seth J Welsh
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Meaghen E Sharik
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Megan T Du
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Victoria M Garbitt
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Daniel L Riggs
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Chang-Xin Shi
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Caleb K Stein
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Marco Bergsagel
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Bryant Chau
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, CA, 94063
| | - Matthew L Wheeler
- Tumor Microenvironment Thematic Research Center, Bristol Myers Squibb, 700 Bay Road, Redwood City, CA, 94063
| | - Natalie Bezman
- Tumor Microenvironment Thematic Research Center, Bristol Myers Squibb, 700 Bay Road, Redwood City, CA, 94063
| | - Feng Wang
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, CA, 94063
| | - Pavel Strop
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, CA, 94063
| | - P Leif Bergsagel
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| | - Marta Chesi
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ, 85259
| |
Collapse
|
105
|
Yan J, Sun S, Zhang W, Li P, Zheng Z. Combined scaffold hopping, molecular screening with dynamic simulation to screen potent CRBN ligands. J Cell Biochem 2021; 122:1207-1215. [PMID: 33938033 DOI: 10.1002/jcb.29941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/07/2021] [Accepted: 04/11/2021] [Indexed: 11/09/2022]
Abstract
Thalidomide and its derivatives lenalidomide and pomalidomide, known as immunomodulatory drugs, (IMiDs) bind directly to cereblon (CRBN), a substrate receptor of an E3 ubiquitin ligase, resulting in the rapid ubiquitination and degradation of the substrate protein. With the discovery of the protein degradation mechanism of IMiDs, targeted protein degradation mediated by IMiDs via CRBN emerged and developed rapidly for the advantages of overcoming drug resistance and targeting undruggable. To date, almost all CRBN ligands are derived from thalidomide and there are few structural differences between them. Hence, we employed an accurate, effective, and rational approach to screen novel and potential CRBN ligands. In this study, we have built a molecular library by scaffold hopping with thalidomide. ADMET screening, virtual screening, and visual inspection screening were performed step-by-step to screen the molecular library and five molecules were hit. Furthermore, docking analysis and a period of 150 ns molecular dynamic (MD) simulation were performed to validate the accuracy of our screen. The docking results showed that molecular A (-10.42 kcal/mol), molecular B (-9.73 kcal/mol), molecular C (-9.25 kcal/mol), molecular D (-9.09 kcal/mol), and molecular E (-10.16 kcal/mol) have lower binding energy than thalidomide (-5.42 kcal/mol), lenalidomide (-5.74 kcal/mol), and pomalidomide (-5.51 kcal/mol). In the MD simulation, all the five screened molecules form key interactions with the active site amino acid residues (Trp380, Trp386, and Trp400) as well as the three marketed IMiDs. Besides, we found and explained that Pro352 was positive for ligand binding to CRBN and Glu377 in reverse, which has not been reported before. We believe that our findings and those five molecules can serve as further optimization of CRBN ligands and development of proteolysis targeting chimeras.
Collapse
Affiliation(s)
- Jian Yan
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Shiyang Sun
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wenjuan Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Pengyun Li
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhibing Zheng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
106
|
Mynott RL, Wallington-Beddoe CT. Drug and Solute Transporters in Mediating Resistance to Novel Therapeutics in Multiple Myeloma. ACS Pharmacol Transl Sci 2021; 4:1050-1065. [PMID: 34151200 DOI: 10.1021/acsptsci.1c00074] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Indexed: 02/06/2023]
Abstract
Multiple myeloma remains an incurable malignancy of plasma cells. Novel therapies, notably proteasome inhibitors and immunomodulatory drugs, have improved the survival of multiple myeloma patients; however, patients either present with, or develop resistance to, these therapies. Resistance to traditional chemotherapeutic agents can be caused by cellular drug efflux via adenosine triphosphate (ATP)-binding cassette (ABC) transporters, but it is still not clear whether these transporters mediate resistance to proteasome inhibitors and immunomodulatory drugs in multiple myeloma. Solute carrier (SLC) transporters also play a role in cancer drug resistance due to changes in cell homeostasis caused by their abnormal expression and changes in the solutes they transport. In this review, we evaluate resistance to novel therapies used to treat multiple myeloma, as mediated by drug and solute transporters.
Collapse
Affiliation(s)
- Rachel L Mynott
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Craig T Wallington-Beddoe
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia.,Flinders Medical Centre, Bedford Park, South Australia 5042, Australia.,Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, South Australia 5000, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| |
Collapse
|
107
|
Cai QQ, Gao XM, Le J, Zhao H, Cai H, Cao XX, Li J. Cereblon expression is a prognostic marker in newly diagnosed POEMS syndrome treated with lenalidomide plus dexamethasone. Ann Hematol 2021; 100:1547-1552. [PMID: 33839882 DOI: 10.1007/s00277-021-04517-9] [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: 02/17/2021] [Accepted: 04/05/2021] [Indexed: 11/27/2022]
Abstract
POEMS syndrome is a rare plasma cell disorder. Lenalidomide has recently emerged as a therapeutic option for POEMS syndrome. Cereblon has been identified as the direct target of lenalidomide, and high cereblon expression is associated with better response and outcome to lenalidomide therapy in multiple myeloma patients. Here, we analyzed the predictive value of cereblon, IKZF1, and IKZF3 in CD138+ selected plasma cells from forty-one newly diagnosed POEMS syndrome patients treated with lenalidomide in combination with dexamethasone at both gene and protein levels. We found that patients with high cereblon expression tended to achieve better hematologic response compared to those with low expression (p = 0.024 for gene expression; p = 0.01 for protein expression). Multivariate Cox regression analysis revealed high cereblon mRNA expression as an independent prognostic marker for longer progression-free survival (hazard ratio 0.542; 95% CI 0.337-0.871; p = 0.011). In conclusion, our results emphasized the role of cereblon mRNA expression as a unique biomarker for predicting the clinical response and outcome of lenalidomide-based therapy in newly diagnosed POEMS syndrome patients.
Collapse
Affiliation(s)
- Qian-Qian Cai
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Xue-Min Gao
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jing Le
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Hao Zhao
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hao Cai
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xin-Xin Cao
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jian Li
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
108
|
Clinical Applications of Genomic Alterations in ATLL: Predictive Markers and Therapeutic Targets. Cancers (Basel) 2021; 13:cancers13081801. [PMID: 33918793 PMCID: PMC8068906 DOI: 10.3390/cancers13081801] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary In this review paper, we aim to summarize recent findings of genomic alterations found in adult T-cell leukemia/lymphoma (ATLL), which is an incurable disease induced by a virus; human T-cell leukemia virus type 1 (HTLV-1). Genomic alterations of ATLL have been comprehensively analyzed and the identified alterations and HTLV-1 infection synergistically act for ATLL development. As HTLV-1 is an endemic disease, ATLL frequently occurs in the endemic areas. Current clinicogenomic analyses suggest the existence of regional difference in ATLL pathophysiology. From a clinical perspective, several studies identified alterations that act as predictive markers and that a part of the alterations can be targetable in ATLL. The alterations can be leveraged to improve ATLL prognosis. Abstract Adult T-cell leukemia/lymphoma (ATLL) is a peripheral T-cell lymphoma (PTCL) caused by human T-cell leukemia virus type 1 (HTLV-1). Recent comprehensive genomic analyses have revealed the genomic landscape. One of the important findings of genomic alterations in ATLL is that almost all alterations are subclonal, suggesting that therapeutic strategies targeting a genomic alteration will result in partial effects. Among the identified alterations, genes involved in T-cell receptor signaling and immune escape mechanisms, such as PLCG1, CARD11, and PD-L1 (also known as CD274), are characteristic of ATLL alterations. From a geographic perspective, ATLL patients in Caribbean islands tend to be younger than those in Japan and the landscape differs between the two areas. Additionally, young Japanese ATLL patients frequently have CD28 fusions, compared with unselected Japanese cases. From a clinical perspective, PD-L1 amplification is an independent prognostic factor among every subtype of ATLL case. Recently, genomic analysis using deep sequencing identified a pre-ATLL clone with ATLL-common mutations in HTLV-1 carriers before development, indicating that genomic analysis can stratify cases based on the risks of development and mortality. In addition to genomic alterations, targetable super-enhancers have been identified in ATLL. These data can be leveraged to improve the prognosis of ATLL.
Collapse
|
109
|
Emerging Therapeutic Strategies to Overcome Drug Resistance in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13071686. [PMID: 33918370 PMCID: PMC8038312 DOI: 10.3390/cancers13071686] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Multiple myeloma is a deadly blood cancer, but fortunately drug development has substantially prolonged the lifespan of patients to average more than a decade after diagnosis with optimal therapy. As a result, the population of patients living with multiple myeloma has grown considerably. Through its course, patients suffer repeated relapses for which they require new lines of treatment. Currently, the key drug classes for treatment are immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies. The goal of this review is to summarize the understanding of the problem of resistance to these drugs, which is ultimately responsible for patient fatality. In addition, we will focus on how new agents that are promising in clinical trials overcome resistance. Abstract Multiple myeloma is a malignant plasma cell neoplasm that remains incurable and is ultimately fatal when patients acquire multi-drug resistance. Thus, advancing our understanding of the mechanisms behind drug resistance in multi-relapsed patients is critical for developing better strategies to extend their lifespan. Here, we review the understanding of resistance to the three key drug classes approved for multiple myeloma treatment: immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies. We consider how the complex, heterogenous biology of multiple myeloma may influence the acquisition of drug resistance and reflect on the gaps in knowledge where additional research is needed to improve our treatment approaches. Fortunately, many agents are currently being evaluated preclinically and in clinical trials that have the potential to overcome or delay drug resistance, including next-generation immunomodulatory drugs and proteasome inhibitors, novel small molecule drugs, chimeric antigen receptor T cells, antibody-drug conjugates, and bispecific antibodies. For each class, we discuss the potential of these strategies to overcome resistance through modifying agents within each class or new classes without cross-resistance to currently available drugs.
Collapse
|
110
|
Hirano M, Imai Y, Kaito Y, Murayama T, Sato K, Ishida T, Yamamoto J, Ito T, Futami M, Ri M, Yasui H, Denda T, Tanaka Y, Ota Y, Nojima M, Kamikubo Y, Gotoh N, Iida S, Handa H, Tojo A. Small-molecule HDAC and Akt inhibitors suppress tumor growth and enhance immunotherapy in multiple myeloma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:110. [PMID: 33757580 PMCID: PMC7989023 DOI: 10.1186/s13046-021-01909-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/14/2021] [Indexed: 12/29/2022]
Abstract
Background Multiple myeloma (MM) is an incurable disease. The acquisition of resistance to drugs, including immunomodulatory drugs (IMiDs), has a negative effect on its prognosis. Cereblon (CRBN) is a key mediator of the bioactivities of IMiDs such as lenalidomide. Moreover, genetic alteration of CRBN is frequently detected in IMiD-resistant patients and is considered to contribute to IMiD resistance. Thus, overcoming resistance to drugs, including IMiDs, is expected to improve clinical outcomes. Here, we examined potential mechanisms of a histone deacetylase (HDAC) inhibitor and Akt inhibitor in relapsed/refractory MM patients. Methods We established lenalidomide-resistant cells by knocking down CRBN with RNAi-mediated downregulation or knocking out CRBN using CRISPR-Cas9 in MM cells. Additionally, we derived multi-drug (bortezomib, doxorubicin, or dexamethasone)-resistant cell lines and primary cells from relapsed/refractory MM patients. The effects of HDAC and Akt inhibitors on these drug-resistant MM cells were then observed with a particular focus on whether HDAC inhibitors enhance immunotherapy efficacy. We also investigated the effect of lenalidomide on CRBN-deficient cells. Results The HDAC inhibitor suppressed the growth of drug-resistant MM cell lines and enhanced the antibody-dependent cellular cytotoxicity (ADCC) of therapeutic antibodies by upregulating natural killer group 2D (NKG2D) ligands in MM cells. CRBN-deficient cells showed lenalidomide-induced upregulation of phosphorylated glycogen synthase kinase-3 (p-GSK-3) and c-Myc phosphorylation. Moreover, HDAC and Akt inhibitors downregulated c-Myc by blocking GSK-3 phosphorylation. HDAC and Akt inhibitors also exhibited synergistic cytotoxic and c-Myc-suppressive effects. The dual HDAC and PI3K inhibitor, CUDC-907, exhibited cytotoxic and immunotherapy-enhancing effects in MM cells, including multi-drug-resistant lines and primary cells from lenalidomide-resistant patients. Conclusions The combination of an HDAC and an Akt inhibitor represents a promising approach for the treatment of relapsed/refractory MM. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01909-7.
Collapse
Affiliation(s)
- Mitsuhito Hirano
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Imai
- Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Yuta Kaito
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takahiko Murayama
- Division of Cancer Cell Biology, Cancer Research Institute of Kanazawa University, Kanazawa, Japan
| | - Kota Sato
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Tadao Ishida
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Junichi Yamamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo, Japan.,Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | - Takumi Ito
- Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | - Muneyoshi Futami
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaki Ri
- Department of Hematology & Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroshi Yasui
- Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Project Division of Fundamental Study on Cutting Edge of Genome Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tamami Denda
- Department of Pathology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yukihisa Tanaka
- Department of Pathology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yasunori Ota
- Department of Pathology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masanori Nojima
- Center for Translational Research/Division of Advanced Medicine Promotion The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yasuhiko Kamikubo
- Laboratory of Oncology and Strategic Innovation, Laboratory Science, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute of Kanazawa University, Kanazawa, Japan
| | - Shinsuke Iida
- Department of Hematology & Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroshi Handa
- Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | - Arinobu Tojo
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
111
|
Paradzik T, Bandini C, Mereu E, Labrador M, Taiana E, Amodio N, Neri A, Piva R. The Landscape of Signaling Pathways and Proteasome Inhibitors Combinations in Multiple Myeloma. Cancers (Basel) 2021; 13:1235. [PMID: 33799793 PMCID: PMC8000754 DOI: 10.3390/cancers13061235] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma is a malignancy of terminally differentiated plasma cells, characterized by an extreme genetic heterogeneity that poses great challenges for its successful treatment. Due to antibody overproduction, MM cells depend on the precise regulation of the protein degradation systems. Despite the success of PIs in MM treatment, resistance and adverse toxic effects such as peripheral neuropathy and cardiotoxicity could arise. To this end, the use of rational combinatorial treatments might allow lowering the dose of inhibitors and therefore, minimize their side-effects. Even though the suppression of different cellular pathways in combination with proteasome inhibitors have shown remarkable anti-myeloma activities in preclinical models, many of these promising combinations often failed in clinical trials. Substantial progress has been made by the simultaneous targeting of proteasome and different aspects of MM-associated immune dysfunctions. Moreover, targeting deranged metabolic hubs could represent a new avenue to identify effective therapeutic combinations with PIs. Finally, epigenetic drugs targeting either DNA methylation, histone modifiers/readers, or chromatin remodelers are showing pleiotropic anti-myeloma effects alone and in combination with PIs. We envisage that the positive outcome of patients will probably depend on the availability of more effective drug combinations and treatment of early MM stages. Therefore, the identification of sensitive targets and aberrant signaling pathways is instrumental for the development of new personalized therapies for MM patients.
Collapse
Affiliation(s)
- Tina Paradzik
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
| | - Cecilia Bandini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
| | - Elisabetta Mereu
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
| | - Maria Labrador
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
| | - Elisa Taiana
- Department of Oncology and Hemato-oncology, University of Milano, 20122 Milano, Italy; (E.T.); (A.N.)
- Hematology Unit, Fondazione Cà Granda IRCCS, Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Antonino Neri
- Department of Oncology and Hemato-oncology, University of Milano, 20122 Milano, Italy; (E.T.); (A.N.)
- Hematology Unit, Fondazione Cà Granda IRCCS, Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Roberto Piva
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
- Città Della Salute e della Scienza Hospital, 10126 Torino, Italy
| |
Collapse
|
112
|
D'Souza C, Prince HM, Neeson PJ. Understanding the Role of T-Cells in the Antimyeloma Effect of Immunomodulatory Drugs. Front Immunol 2021; 12:632399. [PMID: 33746969 PMCID: PMC7973099 DOI: 10.3389/fimmu.2021.632399] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
Immunomodulatory drugs (IMiDs) are effective treatments for patients with multiple myeloma. IMiDs have pleotropic effects including targeting the myeloma cells directly, and improving the anti-myeloma immune response. In the absence of myeloma cells, lenalidomide and pomalidomide induce CD4+ T cell secretion of IL-2 and indirect activation of Natural Killer (NK) cells. In the context of T cell receptor ligation, IMiDs enhance T cell proliferation, cytokine release and Th1 responses, both in vivo and in vitro. Furthermore, combination treatment of IMiDs and myeloma-targeting monoclonal antibodies eg. daratumumab (anti-CD38) and elotuzumab (anti-SLAMF7), checkpoint inhibitors, or bispecific T cell engagers showed synergistic effects, mainly via enhanced T and NK cell dependent cellular toxicity and T cell proliferation. Conversely, the corticosteroid dexamethasone can impair the immune modulatory effects of IMiDs, indicating that careful choice of myeloma drugs in combination with IMiDs is key for the best anti-myeloma therapeutic efficacy. This review presents an overview of the role for T cells in the overall anti-myeloma effects of immunomodulatory drugs.
Collapse
Affiliation(s)
- Criselle D'Souza
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - H Miles Prince
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Clinical Hematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
113
|
The paradoxical pharmacological mechanisms of lenalidomide and bortezomib in the treatment of multiple myeloma. Anticancer Drugs 2021; 32:227-232. [PMID: 33534410 DOI: 10.1097/cad.0000000000001041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The combination of bortezomib (Velcade, PS-341) and lenalidomide (Revlimid) for the treatment of multiple myeloma was proved by USA Food and Drug Administration in 2006. Lenalidomide prevents the proliferation of multiple myeloma cells through binding to cereblon and promoting the ubiquitinational degradation of IKZF1 (Ikaros)/IKZF3 (Aiolos). However, the proteasome inhibitor bortezomib would inhibit the ubiquitinational degradation of IKZF1/IKZF3. How bortezomib could not block the antiproliferative effect of lenalidomide on multiple myeloma cells, which is the paradoxical pharmacological mechanisms in multiple myeloma. In this review, we summarized recent advances in molecular mechanisms underlying the combination of bortezomib and lenalidomide for the treatment multiple myeloma, discussed the paradoxical pharmacological mechanisms of lenalidomide and bortezomib in the treatment of multiple myeloma.
Collapse
|
114
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021. [DOI: 10.37349/etat.2020.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3University of Montpellier, UFR Medicine, 34093 Montpellier, France 4 Institut Universitaire de France (IUF), 75000 Paris France
| |
Collapse
|
115
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:65-106. [PMID: 36046090 PMCID: PMC9400753 DOI: 10.37349/etat.2021.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/06/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3UFR Medicine, University of Montpellier, 34093 Montpellier, France 4Institut Universitaire de France (IUF), 75000 Paris, France
| |
Collapse
|
116
|
Role of MBD3-SOX2 axis in residual myeloma following pomalidomide. Leukemia 2021; 35:3319-3323. [PMID: 33603141 DOI: 10.1038/s41375-021-01145-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/17/2020] [Accepted: 01/21/2021] [Indexed: 01/18/2023]
|
117
|
Cherniawsky HM, Kukreti V, Reece D, Masih-Khan E, McCurdy A, Jimenez-Zepeda VH, Sebag M, Song K, White D, Stakiw J, LeBlanc R, Reiman A, Louzada M, Aslam M, Kotb R, Gul E, Atenafu E, Venner CP. The impact of lenalidomide maintenance on second-line chemotherapy in transplant eligible patients with multiple myeloma. Eur J Haematol 2021; 106:673-681. [PMID: 33539037 DOI: 10.1111/ejh.13596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/01/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To understand the impact of therapy sequencing on progression-free (PFS) and overall survival (OS) for the treatment of multiple myeloma (MM). The use of daily, low-dose, lenalidomide maintenance (LM) has raised concern for fostering resistance, preventing its use in the relapsed setting. METHODS We conducted a retrospective analysis of survival outcomes from the Canadian Myeloma Research Group Database. Patients were grouped based on receipt of LM after autologous stem cell transplant and receipt of lenalidomide in second-line therapy, 575 patients were included. RESULTS Patients treated with LM had statistically similar 2nd PFS when re-exposed to lenalidomide in second-line therapy compared to those receiving non-lenalidomide-containing regimens (10.2 vs 14.0 months, P =.53). This cohort also had the longest 2nd OS, 18 months longer than patients treated with LM who did not receive lenalidomide at relapse (55.3 vs 37 months, P =.004). Patients treated with LM also demonstrated deeper responses to second-line therapy than their non-LM counterparts. CONCLUSION Our data suggest that patients progressing on LM who receive lenalidomide-containing therapy at first relapse have comparable 2nd PFS and better 2nd OS compared to non-lenalidomide-containing second-line regimens. Identification of patients mostly likely to benefit from further lenalidomide-containing therapy is paramount.
Collapse
Affiliation(s)
| | - Vishal Kukreti
- Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Donna Reece
- Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Esther Masih-Khan
- Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.,Canadian Myeloma Research Group, Toronto, ON, Canada
| | - Arleigh McCurdy
- Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | | | - Michael Sebag
- Departments of Medicine and Oncology, Division of Hematology, McGill University, Montreal, QC, Canada
| | - Kevin Song
- BC Cancer Agency, Vancouver General Hospital, Vancouver, BC, Canada
| | - Darrell White
- Queen Elizabeth II Health Sciences Centre. Dalhousie University, Halifax, NS, Canada
| | - Julie Stakiw
- Saskatoon Cancer Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Richard LeBlanc
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC, Canada
| | - Anthony Reiman
- Department of Oncology, Saint John Regional Hospital, Saint John, NB, Canada
| | | | | | - Rami Kotb
- Cancer Care Manitoba, Winnipeg, MB, Canada
| | - Engin Gul
- Canadian Myeloma Research Group, Toronto, ON, Canada
| | | | | |
Collapse
|
118
|
Grover SP, Hisada YM, Kasthuri RS, Reeves BN, Mackman N. Cancer Therapy-Associated Thrombosis. Arterioscler Thromb Vasc Biol 2021; 41:1291-1305. [PMID: 33567864 DOI: 10.1161/atvbaha.120.314378] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Steven P Grover
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Yohei M Hisada
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Raj S Kasthuri
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Brandi N Reeves
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| |
Collapse
|
119
|
Heider M, Eichner R, Stroh J, Morath V, Kuisl A, Zecha J, Lawatscheck J, Baek K, Garz AK, Rudelius M, Deuschle FC, Keller U, Lemeer S, Verbeek M, Götze KS, Skerra A, Weber WA, Buchner J, Schulman BA, Kuster B, Fernández-Sáiz V, Bassermann F. The IMiD target CRBN determines HSP90 activity toward transmembrane proteins essential in multiple myeloma. Mol Cell 2021; 81:1170-1186.e10. [PMID: 33571422 PMCID: PMC7980223 DOI: 10.1016/j.molcel.2020.12.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 11/28/2020] [Accepted: 12/30/2020] [Indexed: 12/23/2022]
Abstract
The complex architecture of transmembrane proteins requires quality control (QC) of folding, membrane positioning, and trafficking as prerequisites for cellular homeostasis and intercellular communication. However, it has remained unclear whether transmembrane protein-specific QC hubs exist. Here we identify cereblon (CRBN), the target of immunomodulatory drugs (IMiDs), as a co-chaperone that specifically determines chaperone activity of HSP90 toward transmembrane proteins by means of counteracting AHA1. This function is abrogated by IMiDs, which disrupt the interaction of CRBN with HSP90. Among the multiple transmembrane protein clients of CRBN-AHA1-HSP90 revealed by cell surface proteomics, we identify the amino acid transporter LAT1/CD98hc as a determinant of IMiD activity in multiple myeloma (MM) and present an Anticalin-based CD98hc radiopharmaceutical for MM radio-theranostics. These data establish the CRBN-AHA1-HSP90 axis in the biogenesis of transmembrane proteins, link IMiD activity to tumor metabolism, and nominate CD98hc and LAT1 as attractive diagnostic and therapeutic targets in MM. CRBN functions as a transmembrane protein-specific co-chaperone of HSP90 Disruption of CRBN-HSP90 interaction determines the anti-tumor activity of IMiDs The CD98hc/LAT1 complex is a central target of IMiDs in multiple myeloma CD98hc-Anticalin is a theranostic tool in multiple myeloma
Collapse
Affiliation(s)
- Michael Heider
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany
| | - Ruth Eichner
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany
| | - Jacob Stroh
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany
| | - Volker Morath
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Anna Kuisl
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany
| | - Jana Zecha
- Department of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jannis Lawatscheck
- Center for Integrated Protein Science at the Department of Chemistry, Technical University of Munich, 85748 Garching, Germany
| | - Kheewoong Baek
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Anne-Kathrin Garz
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Ludwig-Maximilians University, 80337 Munich, Germany
| | | | - Ulrich Keller
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Department of Hematology, Oncology and Tumor Immunology (Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, 12200 Berlin, Germany
| | - Simone Lemeer
- Department of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Mareike Verbeek
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Katharina S Götze
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technical University of Munich, 85354 Freising, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Johannes Buchner
- Center for Integrated Protein Science at the Department of Chemistry, Technical University of Munich, 85748 Garching, Germany
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Bernhard Kuster
- Department of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Vanesa Fernández-Sáiz
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany.
| | - Florian Bassermann
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
120
|
Brownsey DK, Rowley BC, Gorobets E, Gelfand BS, Derksen DJ. Rapid synthesis of pomalidomide-conjugates for the development of protein degrader libraries. Chem Sci 2021; 12:4519-4525. [PMID: 34163717 PMCID: PMC8179520 DOI: 10.1039/d0sc05442a] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Current methods for the preparation of heterobifunctional pomalidomide-conjugates rely on methods that are often low yielding and produce intractable byproducts. Herein we describe our strategy for the reliable and succinct preparation of pomalidomide-linkers which is essential to the formation of these conjugates. We present the preparation of 18 pomalidomide-linkers in high yield compared to current literature methods. Our findings show that secondary amines consistently afford greater yields than their primary counterparts, a trend that we were able to exploit in the synthesis of several new pomalidomide homo-dimers in enhanced yields compared to similar literature syntheses. This trend was further utilised to develop the first one-pot synthesis of JQ1-pomalidomide conjugates in yields up to 62%, providing a method that is suited to rapid preparation of conjugate libraries as is frequently required for the development of new protein degraders. Current methods for the preparation of heterobifunctional pomalidomide-conjugates rely on methods that are often low yielding and produce intractable byproducts. Herein we describe our strategy for the succinct preparation of pomalidomide-linkers.![]()
Collapse
Affiliation(s)
- Duncan K Brownsey
- Department of Chemistry, University of Calgary Calgary T2N 1N4 AB Canada
| | - Ben C Rowley
- Department of Chemistry, University of Calgary Calgary T2N 1N4 AB Canada
| | - Evgueni Gorobets
- Department of Chemistry, University of Calgary Calgary T2N 1N4 AB Canada
| | - Benjamin S Gelfand
- Department of Chemistry, University of Calgary Calgary T2N 1N4 AB Canada
| | - Darren J Derksen
- Department of Chemistry, University of Calgary Calgary T2N 1N4 AB Canada
| |
Collapse
|
121
|
Koduri V, Duplaquet L, Lampson BL, Wang AC, Sabet AH, Ishoey M, Paulk J, Teng M, Harris IS, Endress JE, Liu X, Dasilva E, Paulo JA, Briggs KJ, Doench JG, Ott CJ, Zhang T, Donovan KA, Fischer ES, Gygi SP, Gray NS, Bradner J, Medin JA, Buhrlage SJ, Oser MG, Kaelin WG. Targeting oncoproteins with a positive selection assay for protein degraders. SCIENCE ADVANCES 2021; 7:7/6/eabd6263. [PMID: 33547076 PMCID: PMC7864573 DOI: 10.1126/sciadv.abd6263] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 12/17/2020] [Indexed: 06/12/2023]
Abstract
Most intracellular proteins lack hydrophobic pockets suitable for altering their function with drug-like small molecules. Recent studies indicate that some undruggable proteins can be targeted by compounds that can degrade them. For example, thalidomide-like drugs (IMiDs) degrade the critical multiple myeloma transcription factors IKZF1 and IKZF3 by recruiting them to the cereblon E3 ubiquitin ligase. Current loss of signal ("down") assays for identifying degraders often exhibit poor signal-to-noise ratios, narrow dynamic ranges, and false positives from compounds that nonspecifically suppress transcription or translation. Here, we describe a gain of signal ("up") assay for degraders. In arrayed chemical screens, we identified novel IMiD-like IKZF1 degraders and Spautin-1, which, unlike the IMiDs, degrades IKZF1 in a cereblon-independent manner. In a pooled CRISPR-Cas9-based screen, we found that CDK2 regulates the abundance of the ASCL1 oncogenic transcription factor. This methodology should facilitate the identification of drugs that directly or indirectly degrade undruggable proteins.
Collapse
Affiliation(s)
- Vidyasagar Koduri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Leslie Duplaquet
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Benjamin L Lampson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Adam C Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Amin H Sabet
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Mette Ishoey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Joshiawa Paulk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Mingxing Teng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Isaac S Harris
- Ludwig Cancer Center, Boston, MA 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jennifer E Endress
- Ludwig Cancer Center, Boston, MA 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoxi Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Linde Program in Chemical Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ethan Dasilva
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Linde Program in Chemical Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kimberly J Briggs
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christopher J Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Tinghu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - James Bradner
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Jeffrey A Medin
- Departments of Pediatrics and Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sara J Buhrlage
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Linde Program in Chemical Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Matthew G Oser
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
122
|
Shrestha P, Davis DA, Jaeger HK, Stream A, Aisabor AI, Yarchoan R. Pomalidomide restores immune recognition of primary effusion lymphoma through upregulation of ICAM-1 and B7-2. PLoS Pathog 2021; 17:e1009091. [PMID: 33411730 PMCID: PMC7817053 DOI: 10.1371/journal.ppat.1009091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 01/20/2021] [Accepted: 10/23/2020] [Indexed: 01/08/2023] Open
Abstract
Pomalidomide (Pom) is an immunomodulatory drug that has efficacy against Kaposi’s sarcoma, a tumor caused by Kaposi’s sarcoma-associated herpesvirus (KSHV). Pom also induces direct cytotoxicity in primary effusion lymphoma (PEL), a B-cell malignancy caused by KSHV, in part through downregulation of IRF4, cMyc, and CK1α as a result of its interaction with cereblon, a cellular E3 ubiquitin ligase. Additionally, Pom can reverse KSHV-induced downregulation of MHCI and co-stimulatory immune surface molecules ICAM-1 and B7-2 on PELs. Here, we show for the first time that Pom-induced increases in ICAM-1 and B7-2 on PEL cells lead to an increase in both T-cell activation and NK-mediated cytotoxicity against PEL. The increase in T-cell activation can be prevented by blocking ICAM-1 and/or B7-2 on the PEL cell surface, suggesting that both ICAM-1 and B7-2 are important for T-cell co-stimulation by PELs. To gain mechanistic insights into Pom’s effects on surface markers, we generated Pom-resistant (PomR) PEL cells, which showed about 90% reduction in cereblon protein level and only minimal changes in IRF4 and cMyc upon Pom treatment. Pom no longer upregulated ICAM-1 and B7-2 on the surface of PomR cells, nor did it increase T-cell and NK-cell activation. Cereblon-knockout cells behaved similarly to the pomR cells upon Pom-treatment, suggesting that Pom’s interaction with cereblon is necessary for these effects. Further mechanistic studies revealed PI3K signaling pathway as being important for Pom-induced increases in these molecules. These observations provide a rationale for the study of Pom as therapy in treating PEL and other KSHV-associated tumors. Primary effusion lymphoma (PEL) is an aggressive B-cell lymphoma caused by Kaposi’s sarcoma-associated herpesvirus (KSHV). KSHV encodes various genes that enable infected cells to evade recognition and elimination by the immune system. PEL cells are poorly recognized by T-cells and NK cells, partly due to KSHV-induced downregulation of immune stimulatory surface molecules ICAM-1 and B7-2. We previously found that a cereblon-binding immunomodulatory drug pomalidomide (Pom) can restore the levels of these markers on PELs. Here, we show that the increases in ICAM-1 and B7-2 induced by Pom leads to a functional increase in the recognition and killing of PELs by both T-cells and NK cells. Further, exposure of both the PEL cells and T-cells to Pom lead to an even higher T-cell stimulation providing strong evidence that Pom could help PEL patients by providing specific immune-stimulatory effect. We further perform mechanistic studies and show that Pom’s cellular binding partner cereblon as well as the PI3K pathway are important for Pom-mediated increases in these surface markers.
Collapse
Affiliation(s)
- Prabha Shrestha
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - David A. Davis
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Hannah K. Jaeger
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Alexandra Stream
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Ashley I. Aisabor
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
123
|
Shirasaki R, Matthews GM, Gandolfi S, de Matos Simoes R, Buckley DL, Raja Vora J, Sievers QL, Brüggenthies JB, Dashevsky O, Poarch H, Tang H, Bariteau MA, Sheffer M, Hu Y, Downey-Kopyscinski SL, Hengeveld PJ, Glassner BJ, Dhimolea E, Ott CJ, Zhang T, Kwiatkowski NP, Laubach JP, Schlossman RL, Richardson PG, Culhane AC, Groen RWJ, Fischer ES, Vazquez F, Tsherniak A, Hahn WC, Levy J, Auclair D, Licht JD, Keats JJ, Boise LH, Ebert BL, Bradner JE, Gray NS, Mitsiades CS. Functional Genomics Identify Distinct and Overlapping Genes Mediating Resistance to Different Classes of Heterobifunctional Degraders of Oncoproteins. Cell Rep 2021; 34:108532. [PMID: 33406420 DOI: 10.1016/j.celrep.2020.108532] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 06/14/2019] [Accepted: 11/25/2020] [Indexed: 12/15/2022] Open
Abstract
Heterobifunctional proteolysis-targeting chimeric compounds leverage the activity of E3 ligases to induce degradation of target oncoproteins and exhibit potent preclinical antitumor activity. To dissect the mechanisms regulating tumor cell sensitivity to different classes of pharmacological "degraders" of oncoproteins, we performed genome-scale CRISPR-Cas9-based gene editing studies. We observed that myeloma cell resistance to degraders of different targets (BET bromodomain proteins, CDK9) and operating through CRBN (degronimids) or VHL is primarily mediated by prevention of, rather than adaptation to, breakdown of the target oncoprotein; and this involves loss of function of the cognate E3 ligase or interactors/regulators of the respective cullin-RING ligase (CRL) complex. The substantial gene-level differences for resistance mechanisms to CRBN- versus VHL-based degraders explains mechanistically the lack of cross-resistance with sequential administration of these two degrader classes. Development of degraders leveraging more diverse E3 ligases/CRLs may facilitate sequential/alternating versus combined uses of these agents toward potentially delaying or preventing resistance.
Collapse
Affiliation(s)
- Ryosuke Shirasaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Geoffrey M Matthews
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sara Gandolfi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Ricardo de Matos Simoes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Dennis L Buckley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joseline Raja Vora
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Quinlan L Sievers
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Johanna B Brüggenthies
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Haley Poarch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Huihui Tang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Megan A Bariteau
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michal Sheffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Yiguo Hu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Sondra L Downey-Kopyscinski
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paul J Hengeveld
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Brian J Glassner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Eugen Dhimolea
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Christopher J Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tinghu Zhang
- Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicholas P Kwiatkowski
- Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jacob P Laubach
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Robert L Schlossman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Aedin C Culhane
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Richard W J Groen
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Eric S Fischer
- Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joan Levy
- Multiple Myeloma Research Foundation, Norwalk, CT, USA
| | | | - Jonathan D Licht
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | | | - Lawrence H Boise
- Department of Hematology and Medical Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nathanael S Gray
- Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
124
|
Fiskus W, Mill CP, Perera D, Birdwell C, Deng Q, Yang H, Lara BH, Jain N, Burger J, Ferrajoli A, Davis JA, Saenz DT, Jin W, Coarfa C, Crews CM, Green MR, Khoury JD, Bhalla KN. BET proteolysis targeted chimera-based therapy of novel models of Richter Transformation-diffuse large B-cell lymphoma. Leukemia 2021; 35:2621-2634. [PMID: 33654205 PMCID: PMC8410602 DOI: 10.1038/s41375-021-01181-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/19/2021] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
Richter Transformation (RT) develops in CLL as an aggressive, therapy-resistant, diffuse large B cell lymphoma (RT-DLBCL), commonly clonally-related (CLR) to the concomitant CLL. Lack of available pre-clinical human models has hampered the development of novel therapies for RT-DLBCL. Here, we report the profiles of genetic alterations, chromatin accessibility and active enhancers, gene-expressions and anti-lymphoma drug-sensitivity of three newly established, patient-derived, xenograft (PDX) models of RT-DLBCLs, including CLR and clonally-unrelated (CLUR) to concomitant CLL. The CLR and CLUR RT-DLBCL cells display active enhancers, higher single-cell RNA-Seq-determined mRNA, and protein expressions of IRF4, TCF4, and BCL2, as well as increased sensitivity to BET protein inhibitors. CRISPR knockout of IRF4 attenuated c-Myc levels and increased sensitivity to a BET protein inhibitor. Co-treatment with BET inhibitor or BET-PROTAC and ibrutinib or venetoclax exerted synergistic in vitro lethality in the RT-DLBCL cells. Finally, as compared to each agent alone, combination therapy with BET-PROTAC and venetoclax significantly reduced lymphoma burden and improved survival of immune-depleted mice engrafted with CLR-RT-DLBCL. These findings highlight a novel, potentially effective therapy for RT-DLBCL.
Collapse
Affiliation(s)
- Warren Fiskus
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Christopher P. Mill
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Dimuthu Perera
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Christine Birdwell
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Qing Deng
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Haopeng Yang
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Bernardo H. Lara
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Nitin Jain
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Jan Burger
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Alessandra Ferrajoli
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - John A. Davis
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Dyana T. Saenz
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Wendy Jin
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Cristian Coarfa
- grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Craig M. Crews
- grid.47100.320000000419368710Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT USA ,grid.47100.320000000419368710Department of Chemistry, Yale University, New Haven, CT USA ,grid.47100.320000000419368710Department of Pharmacology, Yale University, New Haven, CT USA
| | - Michael R. Green
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Joseph D. Khoury
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Kapil N. Bhalla
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
125
|
Matyskiela ME, Zhu J, Baughman JM, Clayton T, Slade M, Wong HK, Danga K, Zheng X, Labow M, LeBrun L, Lu G, Chamberlain PP, Thompson JW. Cereblon Modulators Target ZBTB16 and Its Oncogenic Fusion Partners for Degradation via Distinct Structural Degrons. ACS Chem Biol 2020; 15:3149-3158. [PMID: 33206504 DOI: 10.1021/acschembio.0c00674] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
There is a growing interest in using targeted protein degradation as a therapeutic modality in view of its potential to expand the druggable proteome. One avenue to using this modality is via molecular glue based Cereblon E3 Ligase Modulating Drug compounds. Here, we report the identification of the transcription factor ZBTB16 as a Cereblon neosubstrate. We also report two new Cereblon modulators, CC-3060 and CC-647, that promote ZBTB16 degradation. Unexpectedly, CC-3060 and CC-647 target ZBTB16 for degradation by primarily engaging distinct structural degrons on different zinc finger domains. The reciprocal fusion proteins, ZBTB16-RARα and RARα-ZBTB16, which cause a rare acute promyelocytic leukemia, contain these same structural degrons and can be targeted for proteasomal degradation with Cereblon modulator treatment. Thus, a targeted protein degradation approach via Cereblon modulators may represent a novel therapeutic strategy in acute promyelocytic leukemia where ZBTB16/RARA rearrangements are critical disease drivers.
Collapse
Affiliation(s)
- Mary E Matyskiela
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| | - Jinyi Zhu
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| | - Joshua M Baughman
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| | - Thomas Clayton
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| | - Michelle Slade
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| | - Hon Kit Wong
- Bristol Myers Squibb CompanyCambridgeMassachusetts02140United States
| | - Kristina Danga
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| | - Xinde Zheng
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| | - Mark Labow
- Bristol Myers Squibb CompanyCambridgeMassachusetts02140United States
| | - Laurie LeBrun
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| | - Gang Lu
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| | | | - Joel W Thompson
- Bristol Myers Squibb Company, San Diego, California 92121, United States
| |
Collapse
|
126
|
Zhou L, Yu W, Jayabalan DS, Niesvizky R, Jaffrey SR, Huang X, Xu G. Caspase-8 Inhibition Prevents the Cleavage and Degradation of E3 Ligase Substrate Receptor Cereblon and Potentiates Its Biological Function. Front Cell Dev Biol 2020; 8:605989. [PMID: 33392195 PMCID: PMC7773819 DOI: 10.3389/fcell.2020.605989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/20/2020] [Indexed: 01/02/2023] Open
Abstract
Cereblon (CRBN), a substrate receptor of cullin 4-RING E3 ligase (CRL4), mediates the ubiquitination and degradation of constitutive substrates and immunomodulatory drug-induced neo-substrates including MEIS2, c-Jun, CLC1, IKZF1/3, CK1α, and SALL4. It has been reported that CRBN itself could be degraded through the ubiquitin-proteasome system by its associated or other cullin-RING E3 ligases, thus influencing its biological functions. However, it is unknown whether the CRBN stability and its biological function could be modulated by caspases. In this study, using model cell lines, we found that activation of the death receptor using tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) leads to the decreased CRBN protein level. Through pharmacological inhibition and activation of caspase-8 (CASP-8), we disclosed that CASP-8 regulates CRBN cleavage in cell lines. Site mapping experiments revealed that CRBN is cleaved after Asp9 upon CASP-8 activation, resulting in the reduced stability. Using myeloma as a model system, we further revealed that either inhibition or genetic depletion of CASP-8 enhances the anti-myeloma activity of lenalidomide (Len) by impairing CRBN cleavage, leading to the attenuated IKZF1 and IKZF3 protein levels and the reduced viability of myeloma cell lines and primary myeloma cells from patients. The present study discovered that the stability of the substrate receptor of an E3 ligase can be modulated by CASP-8 and suggested that administration of CASP-8 inhibitors enhances the overall effectiveness of Len-based combination therapy in myeloma.
Collapse
Affiliation(s)
- Liang Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Wenjun Yu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - David S Jayabalan
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Ruben Niesvizky
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Samie R Jaffrey
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Xiangao Huang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Guoqiang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
127
|
Leissing TM, Luh LM, Cromm PM. Structure driven compound optimization in targeted protein degradation. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 37:73-82. [PMID: 34895657 DOI: 10.1016/j.ddtec.2020.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 06/14/2023]
Abstract
Small molecule induced protein degradation has created tremendous excitement in drug discovery within recent years. Not being confined to target inhibition and being able to remove disease-causing protein targets via engagement and subsequent ubiquitination has provided scientists with a powerful tool to expand the druggable space. At the center of this approach sits the ternary complex formed between an E3 ubiquitin ligase, the small molecule degrader, and the target protein. A productive ternary complex is pivotal for a ubiquitin to be transferred to a surface lysine of the target protein resulting in poly-ubiquitination which enables recognition and finally degradation by the proteasome. As understanding the ternary complex means understanding the degradation process, many efforts are put into obtaining structural information of the ternary complex and getting a snapshot of the underlying conformations and molecular contacts. Locking this transient trimeric intermediate in a crystalline state has proven to be very demanding but the obtained results have tremendously improved our understanding of small molecule degraders. This review discusses target protein degradation from a structural perspective and highlights the evolution of certain degraders based on the obtained structural insights.
Collapse
Affiliation(s)
| | - Laura M Luh
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Philipp M Cromm
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany.
| |
Collapse
|
128
|
Nadeem O, Tai YT, Anderson KC. Immunotherapeutic and Targeted Approaches in Multiple Myeloma. Immunotargets Ther 2020; 9:201-215. [PMID: 33117743 PMCID: PMC7569026 DOI: 10.2147/itt.s240886] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/22/2020] [Indexed: 12/20/2022] Open
Abstract
The multiple myeloma (MM) therapeutic landscape has evolved significantly with the approval of numerous novel agents, including next generation proteasome inhibitors (PIs), immunomodulatory agents (IMIDs), and monoclonal antibodies (MoABs) targeting CD38 and SLAMF7. While these discoveries have led to an unprecedented improval in patient outcomes, the disease still remains incurable. Immunotherapeutic approaches have shown substantial promise in recent studies of chimeric antigen receptor T-cell (CAR T-cell) therapy, bispecific antibodies, and antibody drug conjugates targeting B-cell maturation antigen (BCMA). This review will highlight these novel and targeted therapies in MM, with particular focus on PIs, IMIDs, MoAb and BCMA-directed immunotherapy.
Collapse
Affiliation(s)
- Omar Nadeem
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yu-Tzu Tai
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
129
|
Martinez-Høyer S, Karsan A. Mechanisms of lenalidomide sensitivity and resistance. Exp Hematol 2020; 91:22-31. [PMID: 32976949 DOI: 10.1016/j.exphem.2020.09.196] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022]
Abstract
The discovery that the immunomodulatory imide drugs (IMiDs) possess antitumor properties revolutionized the treatment of specific types of hematological cancers. Since then, much progress has been made in understanding why the IMiDs are so efficient in targeting the malignant clones in difficult-to-treat diseases. Despite their efficacy, IMiD resistance arises eventually. Herein we summarize the mechanisms of sensitivity and resistance to lenalidomide in del(5q) myelodysplastic syndrome and multiple myeloma, two diseases in which these drugs are at the therapeutic frontline. Understanding the molecular and cellular mechanisms underlying IMiD efficacy and resistance may allow development of specific strategies to eliminate the malignant clone in otherwise incurable diseases.
Collapse
Affiliation(s)
- Sergio Martinez-Høyer
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada; Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
130
|
Lian Q, Gao Y, Li Q, He X, Jiang X, Pu Z, Xu G. Cereblon Promotes the Ubiquitination and Proteasomal Degradation of Interleukin Enhancer-Binding Factor 2. Protein J 2020; 39:411-421. [PMID: 33009960 DOI: 10.1007/s10930-020-09918-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2020] [Indexed: 11/25/2022]
Abstract
Interleukin enhancer-binding factor 2 (ILF2) forms a heterodimer with interleukin enhancer-binding factor 3 (ILF3) via double-stranded RNA-binding motif and zinc finger associated domain and thus regulates gene expression and cancer cell growth. However, how ILF2 is degraded in cells remains elusive. In this work, using stable isotope labeling by amino acids in cell culture (SILAC) quantitative proteomics, we find that ILF2 is downregulated in cells expressing cereblon (CRBN). Using affinity purification and immunoblotting analysis, we demonstrate that CRBN interacts with ILF2 and functions as a substrate receptor of the cullin-4 RING E3 ligase complex. Biochemical experiments disclose that CRBN expression reduces ILF2 protein level and this reduction is diminished when the proteasome is inhibited. Upon protein synthesis inhibition, the degradation of ILF2 is enhanced by CRBN. Moreover, CRBN promotes the ubiquitination of ILF2 and thus results in the ubiquitin-mediated proteasomal degradation. Analyses of previously identified post-translational modification sites and the crystal structure of ILF2 discover the potential ubiquitination sites on ILF2. Through mutagenesis and biochemical experiments, we further reveal that the K45R mutation completely abolishes the effect of CRBN on ILF2, suggesting that this is the key residue responsible for its ubiquitination. Taken together, we identify an E3 ligase that regulates ILF2 and uncover a molecular pathway for its degradation. This work might be helpful to elucidate the molecular mechanism by which CRBN regulates diverse cellular functions.
Collapse
Affiliation(s)
- Qihui Lian
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China
| | - Yuan Gao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China
| | - Qian Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China
| | - Xian He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China
| | - Xiaogang Jiang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China
| | - Zhongjian Pu
- Department of Oncology, Haian Hospital of Traditional Chinese Medicine, Haian, 226600, Jiangsu, China.
| | - Guoqiang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
131
|
Yamamoto J, Suwa T, Murase Y, Tateno S, Mizutome H, Asatsuma-Okumura T, Shimizu N, Kishi T, Momose S, Kizaki M, Ito T, Yamaguchi Y, Handa H. ARID2 is a pomalidomide-dependent CRL4 CRBN substrate in multiple myeloma cells. Nat Chem Biol 2020; 16:1208-1217. [PMID: 32958952 DOI: 10.1038/s41589-020-0645-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 08/10/2020] [Indexed: 12/15/2022]
Abstract
The immunomodulatory drug (IMiD) thalidomide and its derivatives lenalidomide and pomalidomide are therapeutic agents used in the treatment of multiple myeloma. Although pomalidomide offers considerable clinical benefits to patients with lenalidomide-resistant multiple myeloma, the molecular mechanisms underlying its superior efficacy remain unclear. Here we show that ARID2, a component of the polybromo-associated BAF (PBAF) chromatin-remodeling complex, is a pomalidomide-induced neosubstrate of CRL4CRBN. BRD7, another subunit of PBAF, is critical for pomalidomide-induced ARID2 degradation. ARID2 is involved in transcriptional regulation of pomalidomide target genes including MYC. Pomalidomide is more effective than lenalidomide in degrading ARID2 and is capable of inhibiting MYC expression and proliferation in lenalidomide-resistant cell lines. Notably, ARID2 expression is associated with a poor prognosis and is higher in chemoresistant minimal residual disease (MRD) populations, and in patients with relapsed/refractory multiple myeloma. These findings suggest that ARID2 is a promising target for overcoming lenalidomide resistance in patients with multiple myeloma.
Collapse
Affiliation(s)
- Junichi Yamamoto
- Department of Chemical Biology, Tokyo Medical University, Shinjuku, Japan.,School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Tetsufumi Suwa
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yuki Murase
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Shumpei Tateno
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Hirotaka Mizutome
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | | | - Nobuyuki Shimizu
- Department of Chemical Biology, Tokyo Medical University, Shinjuku, Japan
| | - Tsutomu Kishi
- Department of Chemical Biology and Applied Chemistry, Nihon University, Koriyama, Japan
| | - Shuji Momose
- Department of Pathology, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Masahiro Kizaki
- Department of Hematology, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Takumi Ito
- Department of Chemical Biology, Tokyo Medical University, Shinjuku, Japan.,PRESTO, JST, Kawaguchi, Japan
| | - Yuki Yamaguchi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.
| | - Hiroshi Handa
- Department of Chemical Biology, Tokyo Medical University, Shinjuku, Japan.
| |
Collapse
|
132
|
Furihata H, Yamanaka S, Honda T, Miyauchi Y, Asano A, Shibata N, Tanokura M, Sawasaki T, Miyakawa T. Structural bases of IMiD selectivity that emerges by 5-hydroxythalidomide. Nat Commun 2020; 11:4578. [PMID: 32929090 PMCID: PMC7490372 DOI: 10.1038/s41467-020-18488-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/25/2020] [Indexed: 01/31/2023] Open
Abstract
Thalidomide and its derivatives exert not only therapeutic effects as immunomodulatory drugs (IMiDs) but also adverse effects such as teratogenicity, which are due in part to different C2H2 zinc-finger (ZF) transcription factors, IKZF1 (or IKZF3) and SALL4, respectively. Here, we report the structural bases for the SALL4-specific proteasomal degradation induced by 5-hydroxythalidomide, a primary thalidomide metabolite generated by the enzymatic activity of cytochrome P450 isozymes, through the interaction with cereblon (CRBN). The crystal structure of the metabolite-mediated human SALL4-CRBN complex and mutagenesis studies elucidate the complex formation enhanced by the interaction between CRBN and an additional hydroxy group of (S)-5-hydroxythalidomide and the variation in the second residue of β-hairpin structure that underlies the C2H2 ZF-type neo-morphic substrate (neosubstrate) selectivity of 5-hydroxythalidomide. These findings deepen our understanding of the pharmaceutical action of IMiDs and provide structural evidence that the glue-type E3 ligase modulators cause altered neosubstrate specificities through their metabolism.
Collapse
Affiliation(s)
- Hirotake Furihata
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Satoshi Yamanaka
- Proteo-Science Center, Ehime University, 3 Bunkyo-cho, Matsuyama, Ehime, 790-8577, Japan
| | - Toshiaki Honda
- Department of Nanopharmaceutical Sciences and Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Aichi, 466-8555, Japan
| | - Yumiko Miyauchi
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Atsuko Asano
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Norio Shibata
- Department of Nanopharmaceutical Sciences and Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Aichi, 466-8555, Japan
| | - Masaru Tanokura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| | - Tatsuya Sawasaki
- Proteo-Science Center, Ehime University, 3 Bunkyo-cho, Matsuyama, Ehime, 790-8577, Japan.
| | - Takuya Miyakawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| |
Collapse
|
133
|
The novel cereblon modulator CC-885 inhibits mitophagy via selective degradation of BNIP3L. Acta Pharmacol Sin 2020; 41:1246-1254. [PMID: 32210356 PMCID: PMC7608331 DOI: 10.1038/s41401-020-0367-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Mitophagy is a degradative pathway that mediates the degradation of the entire mitochondria, and defects in this process are implicated in many diseases including cancer. In mammals, mitophagy is mediated by BNIP3L (also known as NIX) that is a dual regulator of mitochondrial turnover and programmed cell death pathways. Acute myeloid leukemia (AML) cells with deficiency of BNIP3L are more sensitive to mitochondria-targeting drugs. But small molecular inhibitors for BNIP3L are currently not available. Some immunomodulatory drugs (IMiDs) have been proved by FDA for hematologic malignancies, however, the underlining molecular mechanisms are still elusive, which hindered the applications of BNIP3L inhibition for AML treatment. In this study we carried out MS-based quantitative proteomics analysis to identify the potential neosubstrates of a novel thalidomide derivative CC-885 in A549 cells. In total, we quantified 5029 proteins with 36 downregulated in CRBN+/+ cell after CC-885 administration. Bioinformatic analysis showed that macromitophagy pathway was enriched in the negative pathway after CC-885 treatment. We further found that CC-885 caused both dose- and time-dependent degradation of BNIP3L in CRBN+/+, but not CRBN−/− cell. Thus, our data uncover a novel role of CC-885 in the regulation of mitophagy by targeting BNIP3L for CRL4CRBN E3 ligase-dependent ubiquitination and degradation, suggesting that CC-885 could be used as a selective BNIP3L degradator for the further investigation. Furthermore, we demonstrated that CC-885 could enhance AML cell sensitivity to the mitochondria-targeting drug rotenone, suggesting that combining CC-885 and mitochondria-targeting drugs may be a therapeutic strategy for AML patients.
Collapse
|
134
|
[Expression of Cereblon/CRBN in diffuse large B-cell lymphoma and its significance for prognosis]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 41:254-256. [PMID: 32311898 PMCID: PMC7357926 DOI: 10.3760/cma.j.issn.0253-2727.2020.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
135
|
A452, HDAC6-selective inhibitor synergistically enhances the anticancer activity of immunomodulatory drugs in IMiDs-resistant multiple myeloma. Leuk Res 2020; 95:106398. [DOI: 10.1016/j.leukres.2020.106398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/16/2020] [Accepted: 05/30/2020] [Indexed: 12/17/2022]
|
136
|
Beedie SL, Huang PA, Harris EM, Strope JD, Mahony C, Chau CH, Vargesson N, Figg WD. Role of cereblon in angiogenesis and in mediating the antiangiogenic activity of immunomodulatory drugs. FASEB J 2020; 34:11395-11404. [PMID: 32677118 DOI: 10.1096/fj.201903060rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 11/11/2022]
Abstract
Cereblon (CRBN) is a substrate recruiter element of the E3 cullin 4-RING ubiquitin ligase complex, and a binding target of immunomodulatory agents (IMiDs). CRBN is responsible for the pleiotropic effects of IMiDs, yet its function in angiogenesis and in mediating the antiangiogenic effects of IMiDs remains unclear. We investigated the role of CRBN in the angiogenic process and in propagating the antiangiogenic effects of IMiDs in vitro. siRNA-mediated CRBN knock down in human endothelial cells (HUVEC and HMVEC-L), did not affect endothelial cell proliferation, migration, or tube formation. Using CRBN-deficient mice, we further demonstrated that microvessal formation can occur independently of cereblon in the ex vivo mouse aortic ring model. The cereblon E3 ubiquitin ligase complex can recruit endothelial cell-specific factors, AGO2 (associated with angiogenesis), and SALL4 (associated with embryogenesis/angiogenesis), for ubiquitin-mediated degradation. Knockdown of CRBN caused a corresponding increase in AGO2 and SALL4 protein expression and IMiD treatment was able to rescue the siCRBN effect to increase the CRBN expression. These findings suggest one potential mechanism of action that likely involves a tightly coordinated regulation of CRBN with endothelial cell targets and highlight the need to further elucidate the mechanism(s), which could include cereblon-independent pathways, through which IMiDs exert their antiangiogenic effects.
Collapse
Affiliation(s)
- Shaunna L Beedie
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland
| | - Phoebe A Huang
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emily M Harris
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan D Strope
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Mahony
- School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland
| | - Cindy H Chau
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Neil Vargesson
- School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland
| | - William D Figg
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
137
|
Prediction of immunomodulatory drugs (IMiDs) sensitivity in myeloma via determination of baseline anti-oxidative stress capacity. Leukemia 2020; 34:3060-3063. [PMID: 32503975 DOI: 10.1038/s41375-020-0881-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/06/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022]
|
138
|
Yang K, Zhao Y, Nie X, Wu H, Wang B, Almodovar-Rivera CM, Xie H, Tang W. A Cell-Based Target Engagement Assay for the Identification of Cereblon E3 Ubiquitin Ligase Ligands and Their Application in HDAC6 Degraders. Cell Chem Biol 2020; 27:866-876.e8. [PMID: 32413286 DOI: 10.1016/j.chembiol.2020.04.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/04/2020] [Accepted: 04/22/2020] [Indexed: 01/08/2023]
Abstract
Proteolysis-targeting chimeras (PROTACs) is a paradigm shift for small-molecule drug discovery. However, limited E3 ubiquitin ligase ligands with cellular activity are available. In vitro binding assays involve the expression and purification of a large amount of proteins and they often yield ligands that are inactive in cell-based assays due to poor cell permeability, stability, and other reasons. Herein, we report the development of a practical and efficient cell-based target engagement assay to evaluate the binding affinity of a small library of cereblon ligands to its E3 ligase in cells. Selected cell-permeable E3 ligase ligands derived from this assay are then used to construct HDAC6 degraders with cellular protein degradation activity. Because the assay does not involve any genetic engineering, it is relatively easy to transfer from one cell type to a different one.
Collapse
Affiliation(s)
- Ka Yang
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yu Zhao
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Xueqing Nie
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Hao Wu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Bo Wang
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Haibo Xie
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Weiping Tang
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
139
|
Asatsuma-Okumura T, Ito T, Handa H. Molecular Mechanisms of the Teratogenic Effects of Thalidomide. Pharmaceuticals (Basel) 2020; 13:ph13050095. [PMID: 32414180 PMCID: PMC7281272 DOI: 10.3390/ph13050095] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
Thalidomide was sold worldwide as a sedative over 60 years ago, but it was quickly withdrawn from the market due to its teratogenic effects. Thalidomide was later found to have therapeutic effects in several diseases, although the molecular mechanisms remained unclear. The discovery of cereblon (CRBN), the direct target of thalidomide, a decade ago greatly improved our understanding of its mechanism of action. Accumulating evidence has shown that CRBN functions as a substrate of Cullin RING E3 ligase (CRL4CRBN), whose specificity is controlled by ligands such as thalidomide. For example, lenalidomide and pomalidomide, well-known thalidomide derivatives, degrade the neosubstrates Ikaros and Aiolos, resulting in anti-proliferative effects in multiple myeloma. Recently, novel CRBN-binding drugs have been developed. However, for the safe handling of thalidomide and its derivatives, a greater understanding of the mechanisms of its adverse effects is required. The teratogenic effects of thalidomide occur in multiple tissues in the developing fetus and vary in phenotype, making it difficult to clarify this issue. Recently, several CRBN neosubstrates (e.g., SALL4 (Spalt Like Transcription Factor 4) and p63 (Tumor Protein P63)) have been identified as candidate mediators of thalidomide teratogenicity. In this review, we describe the current understanding of molecular mechanisms of thalidomide, particularly in the context of its teratogenicity.
Collapse
Affiliation(s)
| | - Takumi Ito
- Correspondence: ; Tel.: +81-3-9323-3250; Fax: +81-3-9323-3251
| | | |
Collapse
|
140
|
Yang SJ, Jeon SJ, Van Nguyen T, Deshaies RJ, Park CS, Lee KM. Ubiquitin-dependent proteasomal degradation of AMPK gamma subunit by Cereblon inhibits AMPK activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118729. [PMID: 32333926 DOI: 10.1016/j.bbamcr.2020.118729] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 04/12/2020] [Accepted: 04/19/2020] [Indexed: 01/25/2023]
Abstract
Cereblon (CRBN), a substrate receptor for Cullin-ring E3 ubiquitin ligase (CRL), is a major target protein of immunomodulatory drugs. An earlier study demonstrated that CRBN directly interacts with the catalytic α subunit of AMP-activated protein kinase (AMPK), a master regulator of energy homeostasis, down-regulating the enzymatic activity of AMPK. However, it is not clear how CRBN modulates AMPK activity. To investigate the mechanism of CRBN-dependent AMPK inhibition, we measured protein levels of each AMPK subunit in brains, livers, lungs, hearts, spleens, skeletal muscles, testes, kidneys, and embryonic fibroblasts from wild-type and Crbn-/- mice. Protein levels and stability of the regulatory AMPKγ subunit were increased in Crbn-/- mice. Increased stability of AMPKγ in Crbn-/- MEFs was dramatically reduced by exogenous expression of Crbn. In wild-type MEFs, the proteasomal inhibitor MG132 blocked degradation of AMPKγ. We also found that CRL4CRBN directly ubiquitinated AMPKγ. Taken together, these findings suggest that CRL4CRBN regulates AMPK through ubiquitin-dependent proteasomal degradation of AMPKγ.
Collapse
Affiliation(s)
- Seung-Joo Yang
- School of Life Sciences and Aging Research Institute, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Seung-Je Jeon
- School of Life Sciences and Aging Research Institute, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Thang Van Nguyen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Raymond J Deshaies
- Division of Biology and Biological Engineering, and Howard Hughes Medical Institute, California Institute of Technology, Box 114-96, Pasadena, CA 91125, USA
| | - Chul-Seung Park
- School of Life Sciences and Aging Research Institute, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| | - Kwang Min Lee
- Department of Life Science and Environmental Biochemistry, Pusan National University, Miryang 50463, Republic of Korea.
| |
Collapse
|
141
|
Recent advances in the molecular mechanism of thalidomide teratogenicity. Biomed Pharmacother 2020; 127:110114. [PMID: 32304852 DOI: 10.1016/j.biopha.2020.110114] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/15/2022] Open
Abstract
Thalidomide was first marketed in 1957 but soon withdrawn because of its notorious teratogenicity. Studies on the mechanism of action of thalidomide revealed the pleiotropic properties of this class of drugs, including their anti-inflammatory, antiangiogenic and immunomodulatory activities. Based on their notable activities, thalidomide and its analogues, lenalidomide and pomalidomide, have been repurposed to treat erythema nodosum leprosum, multiple myeloma and other haematological malignancies. Thalidomide analogues were recently found to hijack CRL4CRBN ubiquitin ligase to target a number of cellular proteins for ubiquitination and proteasomal degradation. Thalidomide-mediated degradation of SALL4 and p63, transcription factors essential for embryonic development, very likely plays a critical role in thalidomide embryopathy. In this review, we provide a brief retrospective summary of thalidomide-induced teratogenesis, the mechanism of thalidomide activity, and the latest advances in the molecular mechanism of thalidomide-induced birth malformations.
Collapse
|
142
|
Martinez-Høyer S, Deng Y, Parker J, Jiang J, Mo A, Docking TR, Gharaee N, Li J, Umlandt P, Fuller M, Jädersten M, Kulasekararaj A, Malcovati L, List AF, Hellström-Lindberg E, Platzbecker U, Karsan A. Loss of lenalidomide-induced megakaryocytic differentiation leads to therapy resistance in del(5q) myelodysplastic syndrome. Nat Cell Biol 2020; 22:526-533. [PMID: 32251398 DOI: 10.1038/s41556-020-0497-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 02/28/2020] [Indexed: 12/11/2022]
Abstract
Interstitial deletion of the long arm of chromosome 5 (del(5q)) is the most common structural genomic variant in myelodysplastic syndromes (MDS)1. Lenalidomide (LEN) is the treatment of choice for patients with del(5q) MDS, but half of the responding patients become resistant2 within 2 years. TP53 mutations are detected in ~20% of LEN-resistant patients3. Here we show that patients who become resistant to LEN harbour recurrent variants of TP53 or RUNX1. LEN upregulated RUNX1 protein and function in a CRBN- and TP53-dependent manner in del(5q) cells, and mutation or downregulation of RUNX1 rendered cells resistant to LEN. LEN induced megakaryocytic differentiation of del(5q) cells followed by cell death that was dependent on calpain activation and CSNK1A1 degradation4,5. We also identified GATA2 as a LEN-responsive gene that is required for LEN-induced megakaryocyte differentiation. Megakaryocytic gene-promoter analyses suggested that LEN-induced degradation of IKZF1 enables a RUNX1-GATA2 complex to drive megakaryocytic differentiation. Overexpression of GATA2 restored LEN sensitivity in the context of RUNX1 or TP53 mutations by enhancing LEN-induced megakaryocytic differentiation. Screening for mutations that block LEN-induced megakaryocytic differentiation should identify patients who are resistant to LEN.
Collapse
Affiliation(s)
- Sergio Martinez-Høyer
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada.,Department of Hematology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Yu Deng
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jeremy Parker
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Jihong Jiang
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Angela Mo
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - T Roderick Docking
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Nadia Gharaee
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jenny Li
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Patricia Umlandt
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Megan Fuller
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Martin Jädersten
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Austin Kulasekararaj
- Department of Haematological Medicine, King's College Hospital and King's College London, London, UK
| | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia & Department of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alan F List
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia, Canada. .,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
143
|
Ramakrishna R, Diamond TH, Alexander W, Manoharan A, Golombick T. Use of Curcumin in Multiple Myeloma patients intolerant of steroid therapy. Clin Case Rep 2020; 8:739-744. [PMID: 32274049 PMCID: PMC7141712 DOI: 10.1002/ccr3.2735] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/17/2019] [Accepted: 01/27/2020] [Indexed: 12/03/2022] Open
Abstract
Curcumin, when used in a combination regimen in multiple myeloma patients, has comparable progression-free survival without the adverse effects of steroid-based combination therapies that is curcumin may be a viable alternative to corticosteroids in combination with an immunomodulatory drug or proteasome inhibitor.
Collapse
Affiliation(s)
| | | | - William Alexander
- Southern Sydney HaematologyUniversity of WollongongSydneyNSWAustralia
| | | | - Terry Golombick
- Department of EndocrinologySt George HospitalKogarahNSWAustralia
| |
Collapse
|
144
|
Hansen JD, Correa M, Nagy MA, Alexander M, Plantevin V, Grant V, Whitefield B, Huang D, Kercher T, Harris R, Narla RK, Leisten J, Tang Y, Moghaddam M, Ebinger K, Piccotti J, Havens CG, Cathers B, Carmichael J, Daniel T, Vessey R, Hamann LG, Leftheris K, Mendy D, Baculi F, LeBrun LA, Khambatta G, Lopez-Girona A. Discovery of CRBN E3 Ligase Modulator CC-92480 for the Treatment of Relapsed and Refractory Multiple Myeloma. J Med Chem 2020; 63:6648-6676. [PMID: 32130004 DOI: 10.1021/acs.jmedchem.9b01928] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Many patients with multiple myeloma (MM) initially respond to treatment with modern combination regimens including immunomodulatory agents (lenalidomide and pomalidomide) and proteasome inhibitors. However, some patients lack an initial response to therapy (i.e., are refractory), and although the mean survival of MM patients has more than doubled in recent years, most patients will eventually relapse. To address this need, we explored the potential of novel cereblon E3 ligase modulators (CELMoDs) for the treatment of patients with relapsed or refractory multiple myeloma (RRMM). We found that optimization beyond potency of degradation, including degradation efficiency and kinetics, could provide efficacy in a lenalidomide-resistant setting. Guided by both phenotypic and protein degradation data, we describe a series of CELMoDs for the treatment of RRMM, culminating in the discovery of CC-92480, a novel protein degrader and the first CELMoD to enter clinical development that was specifically designed for efficient and rapid protein degradation kinetics.
Collapse
Affiliation(s)
- Joshua D Hansen
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Matthew Correa
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Mark A Nagy
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Matt Alexander
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Veronique Plantevin
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Virginia Grant
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Brandon Whitefield
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Dehua Huang
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Timothy Kercher
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Roy Harris
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Rama Krishna Narla
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Jim Leisten
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Yang Tang
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Mehran Moghaddam
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Katalin Ebinger
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Joseph Piccotti
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Courtney G Havens
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Brian Cathers
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - James Carmichael
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Thomas Daniel
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Rupert Vessey
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Lawrence G Hamann
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Katerina Leftheris
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Derek Mendy
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Frans Baculi
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Laurie A LeBrun
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Gody Khambatta
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Antonia Lopez-Girona
- Celgene Corporation, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| |
Collapse
|
145
|
Li Y, Kassir N, Wang X, Palmisano M, Zhou S. Population Pharmacokinetics and Exposure Response Analysis of Pomalidomide in Subjects With Relapsed or Refractory Multiple Myeloma From the Novel Combination Treatment of Pomalidomide, Bortezomib, and Low-Dose Dexamethasone. J Clin Pharmacol 2020; 60:1061-1075. [PMID: 32168381 DOI: 10.1002/jcph.1602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/06/2020] [Indexed: 11/11/2022]
Abstract
Multiple myeloma is an incurable progressive neoplastic disease that accounts for 10% of all hematologic malignancies. Even though significant progress has been made in the treatment of newly diagnosed multiple myeloma, the disease follows a relapsing course in the majority of patients, and there is a need for more effective therapeutic options for the treatment of relapsed or refractory multiple myeloma. CC-4047-MM-005 and CC-4047-MM-007 were phase 1 and 3 studies to evaluate the novel combination of pomalidomide, bortezomib, and low-dose dexamethasone for the treatment of patients with relapsed or refractory multiple myeloma who have already received lenalidomide-based treatments early. This analysis was performed to characterize the population pharmacokinetics (PK) of pomalidomide from the combination treatment and to examine exposure-response relationships. Our analysis showed that pomalidomide concentration-time profiles from the combination treatment were adequately described with a 1-compartment PK model, with first-order absorption and elimination and pomalidomide exhibiting linear and time-invariant PK with moderate variability from the combination treatment. Except for the body surface area, none of the tested covariates had an effect on pomalidomide PK. Although body surface area was identified as a statistically significant covariate of pomalidomide PK, the impact was not deemed clinically relevant. A flat exposure-response curve was observed, consistent with a near-saturated drug effect at the tested exposure range suggesting an appropriately recommended clinical dose of 4 mg of pomalidomide for the combination treatment. Finally, pomalidomide exposure was not associated with higher probabilities of dose interruption during cycle 1 or dose reduction during the treatment period.
Collapse
Affiliation(s)
- Yan Li
- Translational Development and Clinical Pharmacology, Celgene Corporation, Summit, New Jersey, USA
| | | | - Xiaomin Wang
- Non-Clinical Development, Celgene Corporation, Summit, New Jersey, USA
| | - Maria Palmisano
- Translational Development and Clinical Pharmacology, Celgene Corporation, Summit, New Jersey, USA
| | - Simon Zhou
- Translational Development and Clinical Pharmacology, Celgene Corporation, Summit, New Jersey, USA
| |
Collapse
|
146
|
Abstract
Although therapeutic strategies have been adapted to age and comorbidities of myeloma patients for a long time, all patients currently experiment the same treatment whatever their genomic risk. However, high-risk patients should benefit right now from the most efficient drugs combinations. Herein, we review and discuss how to optimally define risk to adapt treatment and why a modern multiparametric definition of genomic risk is urgently needed. Minimal residual disease status will probably also take a growing place in patient's management, including in treatment adaptation. We also discuss how next-generation sequencing will definitively represent an essential tool to manage risk-based therapeutic strategies. Finally, despite an explosive knowledge of myeloma molecular landscape, targeted therapy perspectives remain poor, with only few exceptions.
Collapse
|
147
|
Tachita T, Kinoshita S, Ri M, Aoki S, Asano A, Kanamori T, Yoshida T, Totani H, Ito A, Kusumoto S, Komatsu H, Yamagata K, Kubo K, Tohkin M, Fukuda S, Iida S. Expression, mutation, and methylation of cereblon-pathway genes at pre- and post-lenalidomide treatment in multiple myeloma. Cancer Sci 2020; 111:1333-1343. [PMID: 32061138 PMCID: PMC7156787 DOI: 10.1111/cas.14352] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/03/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Cereblon (CRBN) is a target for immunomodulatory drugs. This study investigated the prognostic value of the expression of CRBN‐pathway genes on the clinical relevance of lenalidomide (Len) treatment and evaluated the levels of CRBN‐binding proteins and mutations in these genes after Len treatment. Forty‐eight primary multiple myeloma cells were collected prior to treatment with Len and dexamethasone (Ld) and 25 paired samples were obtained post‐Ld therapy. These tumor cells were used to determine the expression and mutated forms of the CRBN‐pathway genes. Following normalization with CRBN levels, there was a significantly reduced IKZF1/CRBN ratio in samples that responded poorly to Ld therapy. Moreover, patients with low ratios of IKZF1/CRBN showed a significantly shorter progression‐free survival (PFS) and overall survival (OS) than those with higher ratios. However, patients with high ratios of KPNA2/CRBN showed a significantly shorter PFS and OS than patients with lower ratios. Of the 25 paired samples analyzed, most samples showed a reduction in the expression of CRBN and an increase in IKZF1 gene expression. No mutations were observed in CRBN, IKZF1, or CUL4A genes in the post‐Ld samples. In conclusion, a decreased expression of IKZF1 and increased expression of KPNA2 compared to that of CRBN mRNA predicts poor outcomes of Ld therapy.
Collapse
Affiliation(s)
- Takuto Tachita
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shiori Kinoshita
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masaki Ri
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Blood Transfusion and Cell Therapy, Nagoya City University Hospital, Nagoya, Japan
| | - Sho Aoki
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Arisa Asano
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Kanamori
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Yoshida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Clinical Oncology, Nagoya Memorial Hospital, Nagoya, Japan
| | - Haruhito Totani
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Asahi Ito
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shigeru Kusumoto
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirokazu Komatsu
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazufumi Yamagata
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kohmei Kubo
- Department of Hematology, Aomori Prefectural Central Hospital, Aomori, Japan
| | - Masahiro Tohkin
- Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Shinsaku Fukuda
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
148
|
Mechanism of Action and Novel IMiD-Based Compounds and Combinations in Multiple Myeloma. ACTA ACUST UNITED AC 2020; 25:19-31. [PMID: 30694856 DOI: 10.1097/ppo.0000000000000354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Over the last 2 decades, thalidomide analogs have induced significant antimyeloma effects via immune-modulation, antiangiogenesis and antiproliferative effects. While the exact molecular mechanism of the targets or the mediators of thalidomide activity were not known, a seminal discovery of cereblon as a thalidomide-binding protein led to explaining the mechanistic basis of antimyeloma activity for this class of agents. Identification of the mechanisms of resistance for immunomodulatory agents (IMiDs), which will have significant clinical implications, remains poorly understood. Newer cereblon modulators with differential effects and improved increased efficacy in cell lines resistant to the current IMiDs are in development with encouraging preclinical data. In this review, we have summarized the mechanisms of action of IMiDs, clinical development, and potential mechanisms of resistance. We also describe novel IMiD-based combinations and the newer cereblon modulators as well.
Collapse
|
149
|
Hu L, Xu J, Wu T, Fan Z, Sun L, Liu Y, Li Y, Zhang C, Wang J, Ding Y, Wang S. Depletion of ID3 enhances mesenchymal stem cells therapy by targeting BMP4 in Sjögren's syndrome. Cell Death Dis 2020; 11:172. [PMID: 32139667 PMCID: PMC7058624 DOI: 10.1038/s41419-020-2359-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cell (MSCs) transplantation has been used to treat Sjögren’s syndrome (SS) based on the immunoregulatory properties of MSCs. However, the effectiveness need improving and its underlying intrinsic mechanisms remain largely unknown. Here, we show that Id3 is upregulated in bone marrow-derived MSCs (BMMSCs) isolated from NOD/ShiLtJ mice, a widely used SS model, compared with ICR mice as control, suggesting that it functions in SS development and therapy. Transplantation of Id3-deficient BMMSCs rescues salivary gland function more effective than wild-type BMMSCs in NOD/ShiLtJ mice. Mechanistically, we show that ID3 negatively regulated BMP4 expression by preventing binding of basic helix–loop–helix protein E2A to the promoter of the Bmp4 gene. BMP4 in turn promoted PGE2 production in MSCs, and exhibited enhanced suppressive activities of T-cell proliferation and Th1 differentiation. Importantly, BMMSCs from SS patients showed significantly lower BMP4 and PGE2 expression than those from healthy individuals. Taken together, our findings revealed the targeting Id3 may be therapeutically useful for improving MSC immunoregulation and effectiveness of MSCs therapy for SS.
Collapse
Affiliation(s)
- Lei Hu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.
| | - Junji Xu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tingting Wu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Zhipeng Fan
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yi Liu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yan Li
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China.,Fortune Link Triones (Beijing) Scitech Co., Ltd., Beijing, China
| | - Chunmei Zhang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Jingsong Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China
| | - Yaozhong Ding
- Department of Immunology, Capital Medical University, Beijing, 100069, China
| | - Songlin Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China. .,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China.
| |
Collapse
|
150
|
Genome-wide screening reveals a role for subcellular localization of CRBN in the anti-myeloma activity of pomalidomide. Sci Rep 2020; 10:4012. [PMID: 32132601 PMCID: PMC7055313 DOI: 10.1038/s41598-020-61027-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/12/2020] [Indexed: 12/25/2022] Open
Abstract
Pomalidomide, a derivative of thalidomide, is an effective treatment for multiple myeloma. The drug exerts its effects through CRBN, a component of the E3 ubiquitin ligase complex CRL4CRBN. To search for novel factors involved in the anti-cancer activity of pomalidomide, we performed a genome-wide shRNA library screen and identified 445 genes as those affecting pomalidomide sensitivity. Genes encoding components of the ubiquitin-proteasome pathway, such as subunits of the CRL4CRBN complex, the COP9 signalosome, and the 26S proteasome, were among the pomalidomide-affecting genes. Karyopherin beta 1 (KPNB1) was identified as a novel pomalidomide-affecting gene. KPNB1 was required for the nuclear import of CRBN and for the CRBN-directed, pomalidomide-dependent degradation of a clinically relevant substrate, the transcription factor Aiolos. By contrast, the cytoplasmic translation factor GSPT1 was degraded following treatment with the thalidomide derivative CC-885 only when CRBN was present in the cytoplasm, indicating that subcellular distribution of CRBN is critical for the efficacy of thalidomide-based medications.
Collapse
|