1
|
AlZahrani A, Alsuhebany N, Tailor IK, Alrajhi AM. Advancing the understanding of venetoclax in t(11;14)-positive multiple myeloma: a comprehensive review of clinical evidence and future prospects. Hematology 2024; 29:2296809. [PMID: 38149670 DOI: 10.1080/16078454.2023.2296809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/12/2023] [Indexed: 12/28/2023] Open
Abstract
Venetoclax is a selective inhibitor of the anti-apoptotic protein B-cell lymphoma 2 (BCL2), as a targeted therapy for multiple myeloma (MM) patients. It was initially approved by the United States Food and Drug Administration for the treatment of chronic lymphocytic leukemia in April 2016 and later for acute myeloid leukemia in October 2020. However, venetoclax is used as an off-label in a subset group of relapsed and refractory multiple myeloma (RRMM) patients with the presence of translocation t(11;14). Preclinical and clinical studies have highlighted the potential of venetoclax in the management of MM patients, with a specific focus on t(11;14) as a predictive biomarker for initiating venetoclax-based treatment. Later, several studies in RRMM patients that used venetoclax in combination with dexamethasone or/and proteasome inhibitors have shown promising results, in which management guidelines have included venetoclax as one of the options to treat MM patients. Hence, this review focuses on the use of venetoclax in RRMM, clinical efficacy, safety, dosing strategies, and predictive biomarkers for initiating venetoclax. Additionally, we discuss ongoing studies that are investigating different combination of venetoclax regimens in MM patients.
Collapse
Key Words
- AML, acute myeloid leukemia; BCL2, B-cell lymphoma 2; CAR T-cell, chimeric antigen receptor T-cell; CLL, chronic lymphocytic leukemia; FDA, Food and Drug Administration; IC50, half maximal inhibitory concentration; IL-6: interleukin 6; IMiD, immune-modulators; MM, multiple myeloma; ORR, overall response rate; PFS, progression-free survival; PI, proteasome inhibitor; RRMM, relapsed/refractory multiple myeloma
- Venetoclax
- multiple myeloma
- t(11;14)
Collapse
Affiliation(s)
- Abdullah AlZahrani
- Pharmaceutical Care Division, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nada Alsuhebany
- Department of Pharmacy Practice, College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Pharmaceutical Care Services, King Abdulaziz Medical City, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Imran K Tailor
- Department of Adult Hematology and Bone Marrow Transplant, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Abdullah M Alrajhi
- Clinical Pharmacy Department, King Fahad Medical City, Riyadh, Saudi Arabia
- Department of Pharmacy Practice, College of Pharmacy, AlFaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Deng S, Derebail S, Weiler VJ, Fong Ng J, Maroto-Martin E, Chatterjee M, Giorgetti G, Chakraborty C, Kalhotra P, Du T, Yao Y, Prabhala R, Shammas M, Gulla A, Aktas Samur A, Samur MK, Qiu L, Anderson KC, Fulciniti M, Munshi NC. Venetoclax resistance leads to broad resistance to standard-of-care anti-MM agents, but not to immunotherapies. Blood Adv 2024; 8:4025-4034. [PMID: 38861273 PMCID: PMC11339023 DOI: 10.1182/bloodadvances.2023012298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/26/2024] [Accepted: 05/18/2024] [Indexed: 06/12/2024] Open
Abstract
ABSTACT To our knowledge, venetoclax is the first example of personalized medicine for multiple myeloma (MM), with meaningful clinical activity as a monotherapy and in combination in patients with myeloma harboring the t(11:14) translocation. However, despite the high response rates and prolonged progression-free survival, a significant proportion of patients eventually relapse. Here, we aim to study adaptive molecular responses after the acquisition of venetoclax resistance in sensitive t(11:14) MM cell models. We therefore generated single-cell venetoclax-resistant t(11:14) MM cell lines and investigated the mechanisms contributing to resistance as well as the cells' sensitivity to other treatments. Our data suggest that acquired resistance to venetoclax is characterized by reduced mitochondrial priming and changes in B-cell lymphoma-2 (BCL-2) family proteins' expression in MM cells, conferring broad resistance to standard-of-care antimyeloma drugs. However, our results show that the resistant cells are still sensitive to immunotherapeutic treatments, highlighting the need to consider appropriate sequencing of these treatments after venetoclax-based regimens.
Collapse
Affiliation(s)
- Shuhui Deng
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Sanika Derebail
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Vera Joy Weiler
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Jessica Fong Ng
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Elena Maroto-Martin
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Madhumouli Chatterjee
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Giulia Giorgetti
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Chandraditya Chakraborty
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Poonam Kalhotra
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Ting Du
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Yao Yao
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Blood Disease Institute, Key Laboratory of Bone Marrow Stem Cell, Xuzhou Medical University, Xuzhou, China
| | - Rao Prabhala
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Masood Shammas
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Annamaria Gulla
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Anil Aktas Samur
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Mehmet Kemal Samur
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Kenneth C. Anderson
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Mariateresa Fulciniti
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Nikhil C. Munshi
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| |
Collapse
|
3
|
Lu Q, Yang D, Li H, Niu T, Tong A. Multiple myeloma: signaling pathways and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:25. [PMID: 38961036 PMCID: PMC11222366 DOI: 10.1186/s43556-024-00188-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy of plasma cells, characterized by osteolytic bone lesions, anemia, hypercalcemia, renal failure, and the accumulation of malignant plasma cells. The pathogenesis of MM involves the interaction between MM cells and the bone marrow microenvironment through soluble cytokines and cell adhesion molecules, which activate various signaling pathways such as PI3K/AKT/mTOR, RAS/MAPK, JAK/STAT, Wnt/β-catenin, and NF-κB pathways. Aberrant activation of these pathways contributes to the proliferation, survival, migration, and drug resistance of myeloma cells, making them attractive targets for therapeutic intervention. Currently, approved drugs targeting these signaling pathways in MM are limited, with many inhibitors and inducers still in preclinical or clinical research stages. Therapeutic options for MM include non-targeted drugs like alkylating agents, corticosteroids, immunomodulatory drugs, proteasome inhibitors, and histone deacetylase inhibitors. Additionally, targeted drugs such as monoclonal antibodies, chimeric antigen receptor T cells, bispecific T-cell engagers, and bispecific antibodies are being used in MM treatment. Despite significant advancements in MM treatment, the disease remains incurable, emphasizing the need for the development of novel or combined targeted therapies based on emerging theoretical knowledge, technologies, and platforms. In this review, we highlight the key role of signaling pathways in the malignant progression and treatment of MM, exploring advances in targeted therapy and potential treatments to offer further insights for improving MM management and outcomes.
Collapse
Affiliation(s)
- Qizhong Lu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Donghui Yang
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, 712100, China
| | - Hexian Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
4
|
Bhowmick K, von Suskil M, Al-Odat OS, Elbezanti WO, Jonnalagadda SC, Budak-Alpdogan T, Pandey MK. Pathways to therapy resistance: The sheltering effect of the bone marrow microenvironment to multiple myeloma cells. Heliyon 2024; 10:e33091. [PMID: 39021902 PMCID: PMC11252793 DOI: 10.1016/j.heliyon.2024.e33091] [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: 02/16/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Multiple Myeloma (MM) is a malignant expansion of plasma cells in the bone marrow (BM), resulting in a disease characterized by symptoms of end organ damage from light chain secretion, crowding of the BM, and bone lesions. Although the past two decades have been characterized by numerous novel therapies emerging, the disease remains incurable due to intrinsic or acquired drug resistance. A major player in MM's drug resistance arises from its intimate relationship with the BM microenvironment (BMME). Through stress-inducing conditions, soluble messengers, and physical adhesion to BM elements, the BMME activates numerous pathways in the myeloma cell. This not only propagates myeloma progression through survival and growth signals, but also specific mechanisms to circumvent therapeutic actions. In this review, we provide an overview of the BMME, the role of individual components in MM survival, and various therapy-specific resistance mechanisms reported in the literature.
Collapse
Affiliation(s)
- Kuntal Bhowmick
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Max von Suskil
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Omar S. Al-Odat
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Weam Othman Elbezanti
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Hematology, MD Anderson Cancer Center at Cooper, Cooper University Health Care, Camden, NJ, USA
| | - Subash C. Jonnalagadda
- Department of Chemistry and Biochemistry, College of Science and Mathematics, Rowan University, Glassboro, NJ, USA
| | - Tulin Budak-Alpdogan
- Department of Hematology, MD Anderson Cancer Center at Cooper, Cooper University Health Care, Camden, NJ, USA
| | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| |
Collapse
|
5
|
Galas-Filipowicz D, Chavda SJ, Gong JN, Huang DCS, Khwaja A, Yong K. Co-operation of MCL-1 and BCL-X L anti-apoptotic proteins in stromal protection of MM cells from carfilzomib mediated cytotoxicity. Front Oncol 2024; 14:1394393. [PMID: 38651147 PMCID: PMC11033393 DOI: 10.3389/fonc.2024.1394393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction BCL-2 family proteins are important for tumour cell survival and drug resistance in multiple myeloma (MM). Although proteasome inhibitors are effective anti-myeloma drugs, some patients are resistant and almost all eventually relapse. We examined the function of BCL-2 family proteins in stromal-mediated resistance to carfilzomib-induced cytotoxicity in MM cells. Methods Co-cultures employing HS5 stromal cells were used to model the interaction with stroma. MM cells were exposed to CFZ in a 1-hour pulse method. The expression of BCL-2 family proteins was assessed by flow cytometry and WB. Pro-survival proteins: MCL-1, BCL-2 and BCL-XL were inhibited using S63845, ABT-199 and A-1331852 respectively. Changes in BIM binding partners were examined by immunoprecipitation and WB. Results CFZ induced dose-dependent cell death of MM cells, primarily mediated by apoptosis. Culture of MM cells on HS-5 stromal cells resulted in reduced cytotoxicity to CFZ in a cell contact-dependent manner, upregulated expression of MCL-1 and increased dependency on BCL-XL. Inhibiting BCL-XL or MCL-1 with BH-3 mimetics abrogated stromal-mediated protection only at high doses, which may not be achievable in vivo. However, combining BH-3 mimetics at sub-therapeutic doses, which alone were without effect, significantly enhanced CFZ-mediated cytotoxicity even in the presence of stroma. Furthermore, MCL-1 inhibition led to enhanced binding between BCL-XL and BIM, while blocking BCL-XL increased MCL-1/BIM complex formation, indicating the cooperative role of these proteins. Conclusion Stromal interactions alter the dependence on BCL-2 family members, providing a rationale for dual inhibition to abrogate the protective effect of stroma and restore sensitivity to CFZ.
Collapse
Affiliation(s)
| | - Selina J. Chavda
- Cancer Institute, University College London, London, United Kingdom
| | - Jia-Nan Gong
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - David C. S. Huang
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Asim Khwaja
- Cancer Institute, University College London, London, United Kingdom
| | - Kwee Yong
- Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
6
|
Chen X, Varma G, Davies F, Morgan G. Approach to High-Risk Multiple Myeloma. Hematol Oncol Clin North Am 2024; 38:497-510. [PMID: 38195306 DOI: 10.1016/j.hoc.2023.12.008] [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] [Indexed: 01/11/2024]
Abstract
Improving the outcome of high-risk myeloma (HRMM) is a key therapeutic aim for the next decade. To achieve this aim, it is necessary to understand in detail the genetic drivers underlying this clinical behavior and to target its biology therapeutically. Advances have already been made, with a focus on consensus guidance and the application of novel immunotherapeutic approaches. Cases of HRMM are likely to have impaired prognosis even with novel strategies. However, if disease eradication and minimal disease states are achieved, then cure may be possible.
Collapse
Affiliation(s)
- Xiaoyi Chen
- Center Blood Cancer, Perlmutter Cancer Center, New York University, NYCLangone, Room# 496, Medical Science Building 4th Floor, 540 1st Avenue, New York, NY 10016, USA
| | - Gaurav Varma
- Center Blood Cancer, Perlmutter Cancer Center, New York University, NYCLangone, Room# 496, Medical Science Building 4th Floor, 540 1st Avenue, New York, NY 10016, USA
| | - Faith Davies
- Center Blood Cancer, Perlmutter Cancer Center, New York University, NYCLangone, Room# 496, Medical Science Building 4th Floor, 540 1st Avenue, New York, NY 10016, USA
| | - Gareth Morgan
- Center Blood Cancer, Perlmutter Cancer Center, New York University, NYCLangone, Room# 496, Medical Science Building 4th Floor, 540 1st Avenue, New York, NY 10016, USA.
| |
Collapse
|
7
|
Liu Z, Shen H, Han M, Zhao X, Liu H, Ding K, Song J, Fu R. Immune profiles to predict bortezomib-based treatment response for multiple myeloma patients. Int Immunopharmacol 2024; 130:111640. [PMID: 38377849 DOI: 10.1016/j.intimp.2024.111640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND To evaluate the distribution of bone marrow immune cell subsets and their correlation with treatment efficacy in patients with multiple myeloma (MM). METHODS We analyzed the bone marrow lymphocyte subsets of 186 newly diagnosed MM patients at diagnosis and their correlation with clinical characteristics. In our study, eight-color flow cytometry, a method commonly used to detect plasma cell phenotypes, was used to analyze seven bone marrow immune cell groups by change gate-strategy. RESULTS First, for all the 7 immune cell groups, the percentage of immature B cells was significantly lower in stage III patients than in stage I patients, while the trend was reversed in memory B cells in both the International Staging System(p = 0.004) and Revised International Staging System(p = 0.018). Second, the percentage of naïve B cells were significantly lower in patients with severe anemia, while the percentage of memory B cells had reversed trend. The percentage of immature B cells were lower in patients with Cr ≥ 2 mg/dL than in patients with Cr < 2 mg/dL. Then we followed the treatment efficacy of 152 patients who received four cycles of induction therapy (bortezomib + dexamethasone or bortezomib + lenalidomide + dexamethasone) and analyzed the relationship between bone marrow lymphocyte subsets at the initial stage and treatment response datasets. We found that both the percentage of B cells(p<0.001) and immature B(p = 0.002) were increased in patients who achieved very good partial remission(VGPR) after four cycles of induction therapy. The ROC results indicated the combination of the multiple immune subgroups had predictive values (AUC = 0.802, p<0.001) in the treatment effect after four cycles of induction therapy. CONCLUSIONS Overall, these results suggest that the analysis of lymphocyte subsets along with plasma cell immunophenotyping could be a potential index for determining the prognosis of MM patients.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin 300052 P. R. China.
| | - Hongli Shen
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin 300052 P. R. China
| | - Mei Han
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin 300052 P. R. China
| | - Xianghong Zhao
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin 300052 P. R. China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin 300052 P. R. China
| | - Kai Ding
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin 300052 P. R. China
| | - Jia Song
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin 300052 P. R. China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin 300052 P. R. China.
| |
Collapse
|
8
|
Yin C, Li Y, Zhang C, Zang S, Wang Z, Yan X, Ma T, Li X, Li W. Sequential gene expression analysis of myelodysplastic syndrome transformation identifies HOXB3 and HOXB7 as the novel targets for mesenchymal cells in disease. BMC Cancer 2024; 24:111. [PMID: 38254070 PMCID: PMC10802074 DOI: 10.1186/s12885-024-11859-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Myelodysplastic syndrome (MDS) is known to arise through the pathogenic bone marrow mesenchymal stem cells (MSC) by interacting with hematopoietic stem cells (HSC). However, due to the strong heterogeneity of MDS patients, it is difficult to find common targets in studies with limited sample sizes. This study aimed to describe sequential molecular changes and identify biomarkers in MSC of MDS transformation. METHODS Multidimensional data from three publicly available microarray and TCGA datasets were analyzed. MDS-MSC was further isolated and cultured in vitro to determine the potential diagnostic and prognostic value of the identified biomarkers. RESULTS We demonstrated that normal MSCs presented greater molecular homogeneity than MDS-MSC. Biological process (embryonic skeletal system morphogenesis and angiogenesis) and pathways (p53 and MAPK) were enriched according to the differential gene expression. Furthermore, we identified HOXB3 and HOXB7 as potential causative genes gradually upregulated during the normal-MDS-AML transition. Blocking the HOXB3 and HOXB7 in MSCs could enhance the cell proliferation and differentiation, inhibit cell apoptosis and restore the function that supports hematopoietic differentiation in HSCs. CONCLUSION Our comprehensive study of gene expression profiling has identified dysregulated genes and biological processes in MSCs during MDS. HOXB3 and HOXB7 are proposed as novel surrogate targets for therapeutic and diagnostic applications in MDS.
Collapse
Affiliation(s)
- Chunlai Yin
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Yanqi Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Cheng Zhang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Shizhu Zang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Zilong Wang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Xue Yan
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Tonghui Ma
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Xia Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, 116044, China.
| | - Weiping Li
- Department of Hematology, the Second Hospital of Dalian Medical University, Dalian, Liaoning, 116027, China.
| |
Collapse
|
9
|
Mielnik M, Szudy-Szczyrek A, Homa-Mlak I, Mlak R, Podgajna-Mielnik M, Gorący A, Małecka-Massalska T, Hus M. The Clinical Relevance of Selected Cytokines in Newly Diagnosed Multiple Myeloma Patients. Biomedicines 2023; 11:3012. [PMID: 38002012 PMCID: PMC10669681 DOI: 10.3390/biomedicines11113012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/29/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematological neoplasm. Cytokines, chemokines, and their receptors, induced by the microenvironment of MM, participate in tumor growth, the attraction of leukocytes, cell homing, and bone destruction. This study aimed to assess the correlation between the pretreatment serum concentrations of interleukin-6 (IL-6), interleukin-8 (IL-8), angiogenic chemokine monocyte chemoattractant protein-1 (MCP-1), and vascular endothelial growth factor (VEGF) and the clinical outcomes and survival of patients newly diagnosed with MM. The study group consisted of 82 individuals. The IL-8 concentration was significantly positively correlated with the age of onset (p = 0.007), the International Staging System (ISS) stage (p = 0.03), the Eastern Cooperative Oncology Group (ECOG) performance status (p < 0.001), the degree of anemia before treatment (p < 0.0001), the degree of kidney disease (p < 0.001), and VEGF (p = 0.0364). Chemotherapy responders had significantly lower concentrations of IL-8 (p < 0.001), IL-6 (p < 0.001), and VEGF (p = 0.04) compared with non-responders. Patients with treatment-induced polyneuropathy had significantly higher levels of IL-8 (p = 0.033). Patients with a high level of IL-6 had a 2-fold higher risk of progression-free survival (PFS) reduction (17 vs. 35 months; HR = 1.89; p = 0.0078), and a more than 2.5-fold higher risk of overall survival (OS) reduction (28 vs. 78 months; HR = 2.62; p < 0.001). High levels of IL-6, IL-8, and VEGF demonstrated significant predictive values for some clinical conditions or outcomes of newly diagnosed MM patients. Patients with an early response to chemotherapy had a significantly lower concentration of these cytokines. A high pretreatment IL-6 concentration was an independent negative prognostic marker for newly diagnosed MM patients.
Collapse
Affiliation(s)
- Michał Mielnik
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
| | - Aneta Szudy-Szczyrek
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
| | - Iwona Homa-Mlak
- Department of Human Physiology, Medical University of Lublin, 20-080 Lublin, Poland; (I.H.-M.)
| | - Radosław Mlak
- Department of Laboratory Diagnostics, Medical University of Lublin, Doktora Witolda Chodźki 1 Str., 20-093 Lublin, Poland;
| | - Martyna Podgajna-Mielnik
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
| | - Aneta Gorący
- Department of Hematology and Bone Marrow Transplantation, Saint Jan of Dukla Oncology Centre of the Lublin Region, Doktora Kazimierza Jaczewskiego 7 Str., 20-090 Lublin, Poland
| | | | - Marek Hus
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
| |
Collapse
|
10
|
Sklavenitis-Pistofidis R, Lightbody ED, Reidy M, Tsuji J, Aranha MP, Heilpern-Mallory D, Huynh D, Chong SJF, Hackett L, Haradhvala NJ, Wu T, Su NK, Berrios B, Alberge JB, Dutta A, Davids MS, Papaioannou M, Getz G, Ghobrial IM, Manier S. Systematic characterization of therapeutic vulnerabilities in Multiple Myeloma with Amp1q reveals increased sensitivity to the combination of MCL1 and PI3K inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551480. [PMID: 37577538 PMCID: PMC10418223 DOI: 10.1101/2023.08.01.551480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The development of targeted therapy for patients with Multiple Myeloma (MM) is hampered by the low frequency of actionable genetic abnormalities. Gain or amplification of chr1q (Amp1q) is the most frequent arm-level copy number gain in patients with MM, and it is associated with higher risk of progression and death despite recent advances in therapeutics. Thus, developing targeted therapy for patients with MM and Amp1q stands to benefit a large portion of patients in need of more effective management. Here, we employed large-scale dependency screens and drug screens to systematically characterize the therapeutic vulnerabilities of MM with Amp1q and showed increased sensitivity to the combination of MCL1 and PI3K inhibitors. Using single-cell RNA sequencing, we compared subclones with and without Amp1q within the same patient tumors and showed that Amp1q is associated with higher levels of MCL1 and the PI3K pathway. Furthermore, by isolating isogenic clones with different copy number for part of the chr1q arm, we showed increased sensitivity to MCL1 and PI3K inhibitors with arm-level gain. Lastly, we demonstrated synergy between MCL1 and PI3K inhibitors and dissected their mechanism of action in MM with Amp1q.
Collapse
Affiliation(s)
- Romanos Sklavenitis-Pistofidis
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elizabeth D. Lightbody
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Mairead Reidy
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Junko Tsuji
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Michelle P. Aranha
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Daniel Heilpern-Mallory
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Daisy Huynh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen J. F. Chong
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Liam Hackett
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicholas J. Haradhvala
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Ting Wu
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Nang K. Su
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brianna Berrios
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jean-Baptiste Alberge
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Ankit Dutta
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Matthew S. Davids
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria Papaioannou
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Hematology Unit, 1st Internal Medicine Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Gad Getz
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Irene M. Ghobrial
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Salomon Manier
- INSERM UMRS1277, CNRS UMR9020, Lille University, 59000, France
- Department of Hematology, CHU Lille, Lille University, 59000, France
| |
Collapse
|
11
|
Tantawy SI, Timofeeva N, Sarkar A, Gandhi V. Targeting MCL-1 protein to treat cancer: opportunities and challenges. Front Oncol 2023; 13:1226289. [PMID: 37601693 PMCID: PMC10436212 DOI: 10.3389/fonc.2023.1226289] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/03/2023] [Indexed: 08/22/2023] Open
Abstract
Evading apoptosis has been linked to tumor development and chemoresistance. One mechanism for this evasion is the overexpression of prosurvival B-cell lymphoma-2 (BCL-2) family proteins, which gives cancer cells a survival advantage. Mcl-1, a member of the BCL-2 family, is among the most frequently amplified genes in cancer. Targeting myeloid cell leukemia-1 (MCL-1) protein is a successful strategy to induce apoptosis and overcome tumor resistance to chemotherapy and targeted therapy. Various strategies to inhibit the antiapoptotic activity of MCL-1 protein, including transcription, translation, and the degradation of MCL-1 protein, have been tested. Neutralizing MCL-1's function by targeting its interactions with other proteins via BCL-2 interacting mediator (BIM)S2A has been shown to be an equally effective approach. Encouraged by the design of venetoclax and its efficacy in chronic lymphocytic leukemia, scientists have developed other BCL-2 homology (BH3) mimetics-particularly MCL-1 inhibitors (MCL-1i)-that are currently in clinical trials for various cancers. While extensive reviews of MCL-1i are available, critical analyses focusing on the challenges of MCL-1i and their optimization are lacking. In this review, we discuss the current knowledge regarding clinically relevant MCL-1i and focus on predictive biomarkers of response, mechanisms of resistance, major issues associated with use of MCL-1i, and the future use of and maximization of the benefits from these agents.
Collapse
Affiliation(s)
- Shady I. Tantawy
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalia Timofeeva
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aloke Sarkar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
12
|
Yao H, Cheng L, Chen D, Zhang Q, Qiu L, Ren SH, Dou BT, Wang H, Huang J, Fan FY. Role of the bone marrow microenvironment in multiple myeloma treatment using CAR-T therapy. Expert Rev Anticancer Ther 2023; 23:807-815. [PMID: 37343305 DOI: 10.1080/14737140.2023.2229029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/20/2023] [Indexed: 06/23/2023]
Abstract
INTRODUCTION Multiple myeloma (MM) is a malignant tumor caused by abnormal proliferation of bone marrow (BM) plasma cells and is the second most common hematologic malignancy. A variety of CAR-T cells targeting multiple myeloma-specific markers have shown good efficacy in clinical trials. However, CAR-T therapy still limits the insufficient duration of efficacy and recurrence of the disease. AREAS COVERED This article reviews the cell populations in the bone marrow of MM, and discusses the potential way to improve the efficiency of CAR-T cells in the treatment of MM by targeting the bone marrow microenvironment. EXPERT OPINION The limits of CAR-T therapy in MM may related to the impairment of T cell activity in the bone marrow microenvironment. This article reviews the cell populations of the immune microenvironment and nonimmune microenvironment in the bone marrow of multiple myeloma, and discusses the potential way to improve the efficiency of CAR-T cells in the treatment of MM by targeting the bone marrow. This may provides a new idea for the CAR-T therapy of multiple myeloma.
Collapse
Affiliation(s)
- Hao Yao
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Lei Cheng
- Department of Pharmacy, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Dan Chen
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Qian Zhang
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Ling Qiu
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Shi-Hui Ren
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Bai-Tao Dou
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Huan Wang
- Department of Hematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, SiChuan, China
- University of Electronic Science and Technology of China, Chengdu, SiChuan, China
| | - Juan Huang
- Department of Hematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, SiChuan, China
| | - Fang-Yi Fan
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| |
Collapse
|
13
|
Suzuki K, Yano S. Treatment Strategy for Ultra-High-Risk Multiple Myelomas with Chromosomal Aberrations Considering Minimal Residual Disease Status and Bone Marrow Microenvironment. Cancers (Basel) 2023; 15:cancers15092418. [PMID: 37173885 PMCID: PMC10177433 DOI: 10.3390/cancers15092418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Despite the development of anti-myeloma therapeutics, such as proteasome inhibitors, immunomodulatory drugs, anti-CD38 monoclonal antibodies, and autologous stem cell transplantation (ASCT), multiple myeloma remains incurable. A trial treatment combining four drugs-daratumumab, carfilzomib, lenalidomide, and dexamethasone-followed by ASCT frequently results in minimal residual disease (MRD) negativity and prevents progressive disease in patients with standard- and high-risk cytogenetics; however, it is insufficient to overcome the poor outcomes in patients with ultra-high-risk chromosomal aberration (UHRCA). In fact, MRD status in autografts can predict clinical outcomes after ASCT. Therefore, the current treatment strategy might be insufficient to overcome the negative impact of UHRCA in patients with MRD positivity after the four-drug induction therapy. High-risk myeloma cells lead to poor clinical outcomes not only by aggressive myeloma behavior but also via the generation of a poor bone marrow microenvironment. Meanwhile, the immune microenvironment effectively suppresses myeloma cells with a low frequency of high-risk cytogenetic abnormalities in early-stage myeloma compared to late-stage myeloma. Therefore, early intervention might be key to improving clinical outcomes in myeloma patients. The purpose of this review is to improve clinical outcomes in patients with UHRCA by considering MRD assessment results and improvement of the microenvironment.
Collapse
Affiliation(s)
- Kazuhito Suzuki
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| |
Collapse
|
14
|
Aksoy O, Lind J, Sunder-Plaßmann V, Vallet S, Podar K. Bone marrow microenvironment- induced regulation of Bcl-2 family members in multiple myeloma (MM): Therapeutic implications. Cytokine 2023; 161:156062. [PMID: 36332463 DOI: 10.1016/j.cyto.2022.156062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/19/2022] [Accepted: 09/30/2022] [Indexed: 11/23/2022]
Abstract
In Multiple Myeloma (MM) the finely tuned homeostasis of the bone marrow (BM) microenvironment is disrupted. Evasion of programmed cell death (apoptosis) represents a hallmark of cancer. Besides genetic aberrations, the supportive and protective MM BM milieu, which is constituted by cytokines and growth factors, intercellular and cell: extracellular matrix (ECM) interactions and exosomes, in particular, plays a key role in the abundance of pro-survival members of the Bcl-2 family (i.e., Mcl-1, Bcl-2, and Bcl-xL) in tumor cells. Moreover, microenvironmental cues have also an impact on stability- regulating post-translational modifications of anti-apoptotic proteins including de/phosphorylation, polyubiquitination; on their intracellular binding affinities, and localization. Advances of our molecular knowledge on the escape of cancer cells from apoptosis have informed the development of a new class of small molecules that mimic the action of BH3-only proteins. Indeed, approaches to directly target anti-apoptotic Bcl-2 family members are among today's most promising therapeutic strategies and BH3-mimetics (i.e., venetoclax) are currently revolutionizing not only the treatment of CLL and AML, but also hold great therapeutic promise in MM. Furthermore, approaches that activate apoptotic pathways indirectly via modification of the tumor microenvironment have already entered clinical practice. The present review article will summarize our up-to-date knowledge on molecular mechanisms by which the MM BM microenvironment, cytokines, and growth factors in particular, mediates tumor cell evasion from apoptosis. Moreover, it will discuss some of the most promising science- derived therapeutic strategies to overcome Bcl-2- mediated tumor cell survival in order to further improve MM patient outcome.
Collapse
Affiliation(s)
- Osman Aksoy
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Judith Lind
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Vincent Sunder-Plaßmann
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Sonia Vallet
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria; Department of Internal Medicine 2, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
| | - Klaus Podar
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria; Department of Internal Medicine 2, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria.
| |
Collapse
|
15
|
Shi D, Gao L, Wan XC, Li J, Tian T, Hu J, Zhang QL, Su YF, Zeng YP, Hu ZJ, Yu BH, Li XQ, Wei P, Li JW, Zhou XY. Clinicopathologic features and abnormal signaling pathways in plasmablastic lymphoma: a multicenter study in China. BMC Med 2022; 20:483. [PMID: 36522654 PMCID: PMC9753322 DOI: 10.1186/s12916-022-02683-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Plasmablastic lymphoma (PBL) is a rare but aggressive B-cell lymphoma subtype with poor prognosis. Knowledge about the etiology, clinicopathologic and molecular features, and outcomes of PBL is limited. This study aimed to examine the clinicopathologic characteristics, therapeutic approaches, and clinical outcomes of PBL patients in a Chinese population. METHODS A total of 102 PBL patients were recruited from three cancer centers. The pathologic features and clinical outcomes of 56 patients with available treatment details and follow-up data were reviewed and analyzed. RNA sequencing was performed in 6 PBL and 11 diffuse large B-cell lymphoma (DLBCL) patients. RESULTS Most patients in our cohort were male (n = 36, 64.3%), and 35 patients presented with Ann Arbor stage I/II disease at diagnosis. All these patients showed negative findings for human immunodeficiency virus, and the vast majority of patients in our cohort were immunocompetent. Lymph nodes (n = 13, 23.2%) and gastrointestinal tract (n = 10, 17.9%) were the most commonly involved site at presentation. Post-treatment complete remission (CR) was the only prognostic factor affecting overall survival (OS) and progression-free survival (PFS) in the multivariate analysis. RNA-seq demonstrated that B-cell receptor (BCR), T-cell receptor (TCR), P53, calcium signaling, and Wnt signaling pathways were significantly downregulated in PBLs compared with GCB (or non-GCB) DLBCLs. CONCLUSIONS In this multicenter study in the Chinese population, PBL mainly occurred in immunocompetent individuals and most patients present with early-stage disease at diagnosis. Post-treatment CR was an important prognostic factor affecting OS and PFS. RNA-seq showed that the B-cell receptor (BCR), P53, calcium signaling, cell adhesion molecules, and Wnt signaling pathways significantly differed between PBL and GCB (or non-GCB) DLBCL, which provided theoretical basis for its pathogenesis and future treatment.
Collapse
Affiliation(s)
- Di Shi
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Lin Gao
- GenePlus-Shenzhen, Shenzhen, 518000, People's Republic of China.,Institute of Microbiology, Chinese Academy of Sciences, Beijing, 102199, China
| | - Xiao-Chun Wan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Jin Li
- Department of Oncology, Hunan Cancer Hospital, Changsha, 410000, China
| | - Tian Tian
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Jue Hu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Qun-Ling Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Yi-Fan Su
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Yu-Peng Zeng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Zi-Juan Hu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Bao-Hua Yu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Xiao-Qiu Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institute of Pathology, Fudan University, Shanghai, 200032, China
| | - Ji-Wei Li
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Xiao-Yan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Institute of Pathology, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
16
|
Ovejero S, Viziteu E, Dutrieux L, Devin J, Lin YL, Alaterre E, Jourdan M, Basbous J, Requirand G, Robert N, de Boussac H, Seckinger A, Hose D, Vincent L, Herbaux C, Constantinou A, Pasero P, Moreaux J. The BLM helicase is a new therapeutic target in multiple myeloma involved in replication stress survival and drug resistance. Front Immunol 2022; 13:983181. [PMID: 36569948 PMCID: PMC9780552 DOI: 10.3389/fimmu.2022.983181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is a hematologic cancer characterized by accumulation of malignant plasma cells in the bone marrow. To date, no definitive cure exists for MM and resistance to current treatments is one of the major challenges of this disease. The DNA helicase BLM, whose depletion or mutation causes the cancer-prone Bloom's syndrome (BS), is a central factor of DNA damage repair by homologous recombination (HR) and genomic stability maintenance. Using independent cohorts of MM patients, we identified that high expression of BLM is associated with a poor outcome with a significant enrichment in replication stress signature. We provide evidence that chemical inhibition of BLM by the small molecule ML216 in HMCLs (human myeloma cell lines) leads to cell cycle arrest and increases apoptosis, likely by accumulation of DNA damage. BLM inhibition synergizes with the alkylating agent melphalan to efficiently inhibit growth and promote cell death in HMCLs. Moreover, ML216 treatment re-sensitizes melphalan-resistant cell lines to this conventional therapeutic agent. Altogether, these data suggest that inhibition of BLM in combination with DNA damaging agents could be of therapeutic interest in the treatment of MM, especially in those patients with high BLM expression and/or resistance to melphalan.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Elena Viziteu
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Laure Dutrieux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Julie Devin
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Yea-Lih Lin
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Elina Alaterre
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Michel Jourdan
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Jihane Basbous
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Guilhem Requirand
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Nicolas Robert
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | | | | | - Dirk Hose
- Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Laure Vincent
- Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Charles Herbaux
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France,Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | - Philippe Pasero
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Jérôme Moreaux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France,Department of Clinical Hematology, CHU Montpellier, Montpellier, France,*Correspondence: Jérôme Moreaux,
| |
Collapse
|
17
|
Sullivan GP, Flanagan L, Rodrigues DA, Ní Chonghaile T. The path to venetoclax resistance is paved with mutations, metabolism, and more. Sci Transl Med 2022; 14:eabo6891. [PMID: 36475901 DOI: 10.1126/scitranslmed.abo6891] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Venetoclax is a B cell lymphoma 2 (BCL-2)-selective antagonist used to treat chronic lymphocytic leukemia (CLL) and acute myelogenous leukemia (AML). Although this has been a promising therapeutic option for these patients, many of these patients develop resistance and relapsed disease. Here, we summarize the emerging mechanisms of resistance to venetoclax treatment, discuss the promising combination strategies, and highlight the combinations that are currently in clinical trials. Efforts to understand mechanisms of resistance are critical to advance the development of new targeted therapeutic strategies and further our understanding of the biological functions of BCL-2 in tumor cells.
Collapse
Affiliation(s)
- Graeme P Sullivan
- Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Lyndsey Flanagan
- Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Daniel Alencar Rodrigues
- Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Tríona Ní Chonghaile
- Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland.,Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
18
|
Zuo X, Liu D. Mechanism of immunomodulatory drug resistance and novel therapeutic strategies in multiple myeloma. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2022; 27:1110-1121. [PMID: 36121114 DOI: 10.1080/16078454.2022.2124694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The mechanism of immunomodulatory drugs (IMiDs) resistance to multiple myeloma (MM) cells has been gradually demonstrated by recently studies, and some potential novel strategies have been confirmed to have antimyeloma activity and be associated with IMiD activity in MM. METHODS This article searched the Pubmed library, reviewed some recently studies related to IMiD resistance to MM cells and summarized some potent agents to improve IMiD resistance to MM cells. RESULTS Studies have confirmed that cereblon is a primary direct protein target of IMiDs. IRF4 not only is affected by the IKZF protein but also can directly inhibit the expression of BMF and BIM, thereby promoting the survival of MM cells. Additionally, the expression of IRF4 and MYC also plays an important role in three important signaling pathways (Wnt, STAT3 and MAPK/ERK) related to IMiD resistance. Notably, MYC, a downstream factor of IRF4, may be upregulated by BRD4, and upregulation of MYC promotes cell proliferation in MM and disease progression. Recently, some novel therapeutic agents targeting BRD4, a histone modification-related 'reader' of epigenetic marks, or other important factors (e.g. TAK1) in relevant signaling pathways have been developed and they may provide new options for relapse/refractory MM therapy, such as BET inhibitors, CBP/EP300 inhibitors, dual-target BET-CBP/EP300 inhibitors, TAK1 inhibitors, and they may provide new options for relapsed/refractory MM therapy. CONCLUSIONS Accumulated studies have revealed that some key factors associated with the mechanism of IMiD resistance to MM cells. Some agents represent promising new therapeutics of MM to regulate the IRF4/MYC axis by inhibiting BRD4 expression or signaling pathway activation.
Collapse
Affiliation(s)
- Xiaojia Zuo
- Department of Hematology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, People's Republic of China.,Department of Oncology and Hematology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, People's Republic of China.,Guizhou Medical University, Guiyang, People's Republic of China
| | - Dingsheng Liu
- Department of Hematology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| |
Collapse
|
19
|
Liu J, Chen Y, Yu L, Yang L. Mechanisms of venetoclax resistance and solutions. Front Oncol 2022; 12:1005659. [PMID: 36313732 PMCID: PMC9597307 DOI: 10.3389/fonc.2022.1005659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/09/2022] [Indexed: 11/25/2022] Open
Abstract
The BCL-2 inhibitor venetoclax is currently approved for treatment of hematologic diseases and is widely used either as monotherapy or in combination strategies. It has produced promising results in the treatment of refractory or relapsed (R/R) and aged malignant hematologic diseases. However, with clinical use, resistance to venetoclax has emerged. We review the mechanism of reduced dependence on BCL-2 mediated by the upregulation of antiapoptotic proteins other than BCL-2, such as MCL-1 and BCL-XL, which is the primary mechanism of venetoclax resistance, and find that this mechanism is achieved through different pathways in different hematologic diseases. Additionally, this paper also summarizes the current investigations of the mechanisms of venetoclax resistance in terms of altered cellular metabolism, changes in the mitochondrial structure, altered or modified BCL-2 binding domains, and some other aspects; this article also reviews relevant strategies to address these resistance mechanisms.
Collapse
Affiliation(s)
- Jiachen Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yidong Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lihua Yu
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lihua Yang
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Sharma A, Nair R, Achreja A, Mittal A, Gupta P, Balakrishnan K, Edgar CL, Animasahun O, Dwivedi B, Barwick BG, Gupta VA, Matulis SM, Bhasin M, Lonial S, Nooka AK, Wiita AP, Boise LH, Nagrath D, Shanmugam M. Therapeutic implications of mitochondrial stress-induced proteasome inhibitor resistance in multiple myeloma. SCIENCE ADVANCES 2022; 8:eabq5575. [PMID: 36170375 PMCID: PMC9519052 DOI: 10.1126/sciadv.abq5575] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The connections between metabolic state and therapy resistance in multiple myeloma (MM) are poorly understood. We previously reported that electron transport chain (ETC) suppression promotes sensitivity to the BCL-2 antagonist venetoclax. Here, we show that ETC suppression promotes resistance to proteasome inhibitors (PIs). Interrogation of ETC-suppressed MM reveals integrated stress response-dependent suppression of protein translation and ubiquitination, leading to PI resistance. ETC and protein translation gene expression signatures from the CoMMpass trial are down-regulated in patients with poor outcome and relapse, corroborating our in vitro findings. ETC-suppressed MM exhibits up-regulation of the cystine-glutamate antiporter SLC7A11, and analysis of patient single-cell RNA-seq shows that clusters with low ETC gene expression correlate with higher SLC7A11 expression. Furthermore, erastin or venetoclax treatment diminishes mitochondrial stress-induced PI resistance. In sum, our work demonstrates that mitochondrial stress promotes PI resistance and underscores the need for implementing combinatorial regimens in MM cognizant of mitochondrial metabolic state.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Remya Nair
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Abhinav Achreja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Anjali Mittal
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Pulkit Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Kamakshi Balakrishnan
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Claudia L. Edgar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Olamide Animasahun
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Bhakti Dwivedi
- Department of Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Vikas A. Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Shannon M. Matulis
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Manoj Bhasin
- Department of Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Ajay K. Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Arun P. Wiita
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Deepak Nagrath
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
- Corresponding author.
| |
Collapse
|
21
|
Solimando AG, Malerba E, Leone P, Prete M, Terragna C, Cavo M, Racanelli V. Drug resistance in multiple myeloma: Soldiers and weapons in the bone marrow niche. Front Oncol 2022; 12:973836. [PMID: 36212502 PMCID: PMC9533079 DOI: 10.3389/fonc.2022.973836] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is still an incurable disease, despite considerable improvements in treatment strategies, as resistance to most currently available agents is not uncommon. In this study, data on drug resistance in MM were analyzed and led to the following conclusions: resistance occurs via intrinsic and extrinsic mechanisms, including intraclonal heterogeneity, drug efflux pumps, alterations of drug targets, the inhibition of apoptosis, increased DNA repair and interactions with the bone marrow (BM) microenvironment, cell adhesion, and the release of soluble factors. Since MM involves the BM, interactions in the MM-BM microenvironment were examined as well, with a focus on the cross-talk between BM stromal cells (BMSCs), adipocytes, osteoclasts, osteoblasts, endothelial cells, and immune cells. Given the complex mechanisms that drive MM, next-generation treatment strategies that avoid drug resistance must target both the neoplastic clone and its non-malignant environment. Possible approaches based on recent evidence include: (i) proteasome and histone deacetylases inhibitors that not only target MM but also act on BMSCs and osteoclasts; (ii) novel peptide drug conjugates that target both the MM malignant clone and angiogenesis to unleash an effective anti-MM immune response. Finally, the role of cancer stem cells in MM is unknown but given their roles in the development of solid and hematological malignancies, cancer relapse, and drug resistance, their identification and description are of paramount importance for MM management.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
- Istituto di ricovero e cura a carattere scientifico (IRCCS) Istituto Tumori ‘Giovanni Paolo II’ of Bari, Bari, Italy
| | - Eleonora Malerba
- Department of Biomedical Sciences and Human Oncology, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
| | - Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
| | - Marcella Prete
- Department of Interdisciplinary Medicine, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
| | - Carolina Terragna
- ’Seràgnoli’ Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Michele Cavo
- ’Seràgnoli’ Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Vito Racanelli
- Department of Interdisciplinary Medicine, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
- *Correspondence: Vito Racanelli,
| |
Collapse
|
22
|
Mark C, Warrick J, Callander NS, Hematti P, Miyamoto S. A Hyaluronan and Proteoglycan Link Protein 1 Matrikine: Role of Matrix Metalloproteinase 2 in Multiple Myeloma NF-κB Activation and Drug Resistance. Mol Cancer Res 2022; 20:1456-1466. [PMID: 35604822 PMCID: PMC9444915 DOI: 10.1158/1541-7786.mcr-21-0941] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/18/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022]
Abstract
The NF-κB signaling pathway plays key roles in inflammation and the pathogenesis of many solid and hematologic malignancies, including multiple myeloma, a malignancy of the plasma cells. While proteasome inhibitors, such as bortezomib, employed in multiple myeloma treatments may inhibit NF-κB signaling pathways, multiple myeloma cells often become drug resistant in part due to non-cell autonomous mechanism(s) from the multiple myeloma tumor microenvironment. We previously found that fragments of, but not full-length, hyaluronan and proteoglycan link protein 1 (HAPLN1), produced by multiple myeloma bone marrow stromal cells (BMSC), activate an atypical bortezomib-resistant NF-κB pathway in multiple myeloma cells. In our current study, we found that multiple myeloma cells promote HAPLN1 expression and matrix metalloproteinase 2 (MMP2) activity in cocultured BMSCs and MMP2 activity is higher in BMSCs established from multiple myeloma patients' BM aspirates relative to normal equivalents. Moreover, MMP2 cleaves HAPLN1 into forms similar in size to those previously observed in patients with multiple myeloma with progressive disease. Both HAPLN1 and MMP2 in BMSCs were required to enhance NF-κB activation and resistance to bortezomib-induced cell death in cocultured multiple myeloma cells. We propose that MMP2-processing of HAPLN1 produces a matrikine that induces NF-κB activation and promotes bortezomib resistance in multiple myeloma cells. IMPLICATIONS HAPLN1 and MMP2 produced by BMSCs obtained from patients with multiple myeloma promote NF-κB activity and resistance to bortezomib toxicity in multiple myeloma cells, uncovering their potential as biomarkers or therapeutic targets to address bortezomib resistance in patients with multiple myeloma.
Collapse
Affiliation(s)
- Christina Mark
- Cancer Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705
| | - Jay Warrick
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705
| | - Natalie S. Callander
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705,Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Peiman Hematti
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705,Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Shigeki Miyamoto
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705,McArdle Laboratory of Cancer Research, University of Wisconsin-Madison, Madison, WI 53705,Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
23
|
Venetoclax ex vivo functional profiling predicts improved progression-free survival. Blood Cancer J 2022; 12:115. [PMID: 35927247 PMCID: PMC9352682 DOI: 10.1038/s41408-022-00710-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/25/2022] Open
|
24
|
Sklavenitis-Pistofidis R, Getz G, Ghobrial I, Papaioannou M. Multiple Myeloma With Amplification of Chr1q: Therapeutic Opportunity and Challenges. Front Oncol 2022; 12:961421. [PMID: 35912171 PMCID: PMC9331166 DOI: 10.3389/fonc.2022.961421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy with a heterogeneous genetic background. Each MM subtype may have its own therapeutic vulnerabilities, and tailored therapy could improve outcomes. However, the cumulative frequency of druggable targets across patients is very low, which has precluded the widespread adoption of precision therapy for patients with MM. Amplification of the long arm of chromosome 1 (Amp1q) is one of the most frequent genetic alterations observed in patients with MM, and its presence predicts inferior outcomes in the era of proteasome inhibitors and immunomodulatory agents. Therefore, establishing precision medicine for MM patients with Amp1q stands to benefit a large portion of patients who are otherwise at higher risk of relapse. In this article, we review the prevalence and clinical significance of Amp1q in patients with MM, its pathogenesis and therapeutic vulnerabilities, and discuss the opportunities and challenges for Amp1q-targeted therapy.
Collapse
Affiliation(s)
- Romanos Sklavenitis-Pistofidis
- Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gad Getz
- Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - Irene Ghobrial
- Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Maria Papaioannou
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Hematology Unit, 1st Internal Medicine Department, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
25
|
Moser-Katz T, Gavile CM, Barwick BG, Lee KP, Boise LH. PDZ Proteins SCRIB and DLG1 Regulate Myeloma Cell Surface CD86 Expression, Growth, and Survival. Mol Cancer Res 2022; 20:1122-1136. [PMID: 35380688 PMCID: PMC9262820 DOI: 10.1158/1541-7786.mcr-21-0681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/28/2022] [Accepted: 04/01/2022] [Indexed: 01/09/2023]
Abstract
Despite advances in the treatment of multiple myeloma in the past decades, the disease remains incurable, and understanding signals and molecules that can control myeloma growth and survival are important for the development of novel therapeutic strategies. One such molecule, CD86, regulates multiple myeloma cell survival via its interaction with CD28 and signaling through its cytoplasmic tail. Although the CD86 cytoplasmic tail has been shown to be involved in drug resistance and can induce molecular changes in multiple myeloma cells, its function has been largely unexplored. Here, we show that CD86 cytoplasmic tail has a role in trafficking CD86 to the cell surface. This is due in part to a PDZ-binding motif at its C-terminus which is important for proper trafficking from the Golgi apparatus. BioID analysis revealed 10 PDZ domain-containing proteins proximal to CD86 cytoplasmic tail in myeloma cells. Among them, we found the planar cell polarity proteins, SCRIB and DLG1, are important for proper CD86 surface expression and the growth and survival of myeloma cells. These findings indicate a mechanism by which myeloma cells confer cellular survival and drug resistance and indicate a possible motif to target for therapeutic gain. IMPLICATIONS These findings demonstrate the importance of proper trafficking of CD86 to the cell surface in myeloma cell survival and may provide a new therapeutic target in this disease.
Collapse
Affiliation(s)
- Tyler Moser-Katz
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Catherine M. Gavile
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Kelvin P. Lee
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| |
Collapse
|
26
|
Schwestermann J, Besse A, Driessen C, Besse L. Contribution of the Tumor Microenvironment to Metabolic Changes Triggering Resistance of Multiple Myeloma to Proteasome Inhibitors. Front Oncol 2022; 12:899272. [PMID: 35692781 PMCID: PMC9178120 DOI: 10.3389/fonc.2022.899272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Virtually all patients with multiple myeloma become unresponsive to treatment with proteasome inhibitors over time. Relapsed/refractory multiple myeloma is accompanied by the clonal evolution of myeloma cells with heterogeneous genomic aberrations, diverse proteomic and metabolic alterations, and profound changes of the bone marrow microenvironment. However, the molecular mechanisms that drive resistance to proteasome inhibitors within the context of the bone marrow microenvironment remain elusive. In this review article, we summarize the latest knowledge about the complex interaction of malignant plasma cells with its surrounding microenvironment. We discuss the pivotal role of metabolic reprograming of malignant plasma cells within the tumor microenvironment with a subsequent focus on metabolic rewiring in plasma cells upon treatment with proteasome inhibitors, driving multiple ways of adaptation to the treatment. At the same time, mutual interaction of plasma cells with the surrounding tumor microenvironment drives multiple metabolic alterations in the bone marrow. This provides a tumor-promoting environment, but at the same time may offer novel therapeutic options for the treatment of relapsed/refractory myeloma patients.
Collapse
Affiliation(s)
| | | | | | - Lenka Besse
- Laboratory of Experimental Oncology, Clinics for Medical Hematology and Oncology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
27
|
Ferrarini I, Rigo A, Visco C. The mitochondrial anti-apoptotic dependencies of hematologic malignancies: from disease biology to advances in precision medicine. Haematologica 2022; 107:790-802. [PMID: 35045693 PMCID: PMC8968907 DOI: 10.3324/haematol.2021.280201] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondria are critical organelles in the regulation of intrinsic apoptosis. As a general feature of blood cancers, different antiapoptotic members of the BCL-2 protein family localize at the outer mitochondrial membrane to sequester variable amounts of proapoptotic activators, and hence protect cancer cells from death induction. However, the impact of distinct anti-apoptotic members on apoptosis prevention, a concept termed anti-apoptotic dependence, differs remarkably across disease entities. Over the last two decades, several genetic and functional methodologies have been established to uncover the anti-apoptotic dependencies of the majority of blood cancers, inspiring the development of a new class of small molecules called BH3 mimetics. In this review, we highlight the rationale of targeting mitochondrial apoptosis in hematology, and provide a comprehensive map of the anti-apoptotic dependencies that are currently guiding novel therapeutic strategies. Cell-extrinsic and -intrinsic mechanisms conferring resistance to BH3 mimetics are also examined, with insights on potential strategies to overcome them. Finally, we discuss how the field of mitochondrial apoptosis might be complemented with other dimensions of precision medicine for more successful treatment of 'highly complex' hematologic malignancies.
Collapse
Affiliation(s)
- Isacco Ferrarini
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy.
| | - Antonella Rigo
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Carlo Visco
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy.
| |
Collapse
|
28
|
Biological Hallmarks and Emerging Strategies to Target STAT3 Signaling in Multiple Myeloma. Cells 2022; 11:cells11060941. [PMID: 35326392 PMCID: PMC8946161 DOI: 10.3390/cells11060941] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy, characterized by an abnormal accumulation of plasma cells in the bone marrow. Signal transducer and activator of transcription 3 (STAT3) is a cytoplasmic transcription factor that modulates the transcription of multiple genes to regulate various principal biological functions, for example, cell proliferation and survival, stemness, inflammation and immune responses. Aberrant STAT3 activation has been identified as a key driver of tumorigenesis in many types of cancers, including MM. Herein, we summarize the current evidence for the role of STAT3 in affecting cancer hallmark traits by: (1) sustaining MM cell survival and proliferation, (2) regulating tumor microenvironment, (3) inducing immunosuppression. We also provide an update of different strategies for targeting STAT3 in MM with special emphasis on JAK inhibitors that are currently undergoing clinical trials. Finally, we discuss the challenges and future direction of understanding STAT3 signaling in MM biology and the clinical development of STAT3 inhibitors.
Collapse
|
29
|
Gozzetti A, Ciofini S, Sicuranza A, Pacelli P, Raspadori D, Cencini E, Tocci D, Bocchia M. Drug resistance and minimal residual disease in multiple myeloma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:171-183. [PMID: 35582527 PMCID: PMC8992600 DOI: 10.20517/cdr.2021.116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/17/2022] [Accepted: 01/29/2022] [Indexed: 11/12/2022]
Abstract
Great progress has been made in improving survival in multiple myeloma (MM) patients over the last 30 years. New drugs have been introduced and complete responses are frequently seen. However, the majority of MM patients do experience a relapse at a variable time after treatment, and ultimately the disease becomes drug-resistant following therapies. Recently, minimal residual disease (MRD) detection has been introduced in clinical trials utilizing novel therapeutic agents to measure the depth of response. MRD can be considered as a surrogate for both progression-free and overall survival. In this perspective, the persistence of a residual therapy-resistant myeloma plasma cell clone can be associated with inferior survivals. The present review gives an overview of drug resistance in MM, i.e., mutation of β5 subunit of the proteasome; upregulation of pumps of efflux; heat shock protein induction for proteasome inhibitors; downregulation of CRBN expression; deregulation of IRF4 expression; mutation of CRBN, IKZF1, and IKZF3 for immunomodulatory drugs and decreased target expression; complement protein increase; sBCMA increase; and BCMA down expression for monoclonal antibodies. Multicolor flow cytometry, or next-generation flow, and next-generation sequencing are currently the techniques available to measure MRD with sensitivity at 10-5. Sustained MRD negativity is related to prolonged survival, and it is evaluated in all recent clinical trials as a surrogate of drug efficacy.
Collapse
Affiliation(s)
- Alessandro Gozzetti
- Hematology, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena 53100, Italy
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Harmon KA, Roman S, Lancaster HD, Chowhury S, Cull E, Goodwin RL, Arce S, Fanning S. Structural and Ultrastructural Analysis of the Multiple Myeloma Cell Niche and a Patient-Specific Model of Plasma Cell Dysfunction. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2022; 28:254-264. [PMID: 34881690 DOI: 10.1017/s1431927621013805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Multiple myeloma (MM) is a deadly, incurable malignancy in which antibody-secreting plasma cells (PCs) become neoplastic. Previous studies have shown that the PC niche plays a role cancer progression. Bone marrow (BM) cores from MM and a premalignant condition known as monoclonal gammopathy of unknown significance (MGUS) patients were analyzed with confocal and transmission electron microscopy. The BM aspirates from these patients were used to generate 3D PC cultures. These in vitro cultures were then assayed for the molecular, cellular, and ultrastructural hallmarks of dysfunctional PC at days 1 and 5. In vivo, evidence of PC endoplasmic reticulum stress was found in both MM and MGUS BM; however, evidence of PC autophagy was found only in MM BM. Analysis of in vitro cultures found that MM PC can survive and maintain a differentiated phenotype over an unprecedented 5 days, had higher levels of paraprotein production when compared to MGUS-derived cultures, and showed evidence of PC autophagy as well. Increased fibronectin deposition around PC associated with disease severity and autophagy dysregulation was also observed. 3D cultures constructed from BM aspirates from MGUS and MM patients allow for long-term culture of functional PC while maintaining their distinct morphological phenotypes.
Collapse
Affiliation(s)
| | | | - Harrison D Lancaster
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, USA
| | - Saeeda Chowhury
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, USA
- Department of Internal Medicine, Prisma Health System Upstate, Greenville, SC29605, USA
- Prisma Health Cancer Institute, Greenville, SC29605, USA
| | - Elizabeth Cull
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, USA
- Department of Internal Medicine, Prisma Health System Upstate, Greenville, SC29605, USA
- Prisma Health Cancer Institute, Greenville, SC29605, USA
| | - Richard L Goodwin
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, USA
| | - Sergio Arce
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, USA
- Prisma Health Cancer Institute, Greenville, SC29605, USA
| | - Suzanne Fanning
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, USA
- Department of Internal Medicine, Prisma Health System Upstate, Greenville, SC29605, USA
- Prisma Health Cancer Institute, Greenville, SC29605, USA
| |
Collapse
|
31
|
Abstract
Multiple myeloma is a common hematological malignancy of plasma cells, the terminally differentiated B cells that secrete antibodies as part of the adaptive immune response. Significant progress has been made in treating multiple myeloma, but this disease remains largely incurable, and most patients will eventually suffer a relapse of disease that becomes refractory to further therapies. Moreover, a portion of patients with multiple myeloma present with disease that is refractory to all treatments from the initial diagnosis, and no current therapeutic approaches can help. Therefore, the task remains to advance new therapeutic strategies to help these vulnerable patients. One strategy to meet this challenge is to unravel the complex web of pathogenic signaling pathways in malignant plasma cells and use this information to design novel precision medicine strategies to assist these patients most at risk.
Collapse
Affiliation(s)
- Arnold Bolomsky
- Wilhelminen Cancer Research Institute, Dept. of Medicine I, Wilhelminenspital, Vienna Austria
| | - Ryan M. Young
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Lymphoid Malignancies Branch, Bethesda MD 20892,Lymphoid Malignancies Branch, Center for Cancer Research, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD. 20892, , 240-858-3513
| |
Collapse
|
32
|
Wang C, Zhang Y, Wu Y, Xing D. Developments of CRBN-based PROTACs as potential therapeutic agents. Eur J Med Chem 2021; 225:113749. [PMID: 34411892 DOI: 10.1016/j.ejmech.2021.113749] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022]
Abstract
Protease-targeted chimeras (PROTACs) are a new technology that is receiving much attention in the treatment of diseases. The mechanism is to inhibit protein function by hijacking the ubiquitin E3 ligase for protein degradation. Heterogeneous bifunctional PROTACs contain a ligand for recruiting E3 ligase, a linker, and another ligand to bind to the target protein for degradation. A variety of small-molecule PROTACs (CRBN, VHL, IAPs, MDM2, DCAF15, DCAF16, and RNF114-based PROTACs) have been identified so far. In particular, CRBN-based PROTACs (e.g., ARV-110 and ARV-471) have received more attention for their promising therapeutic intervention. To date, CRBN-based PRTOACs have been extensively explored worldwide and have excelled not only in cancer diseases but also in cardiovascular diseases, immune diseases, neurodegenerative diseases, and viral infections. In this review, we will provide a comprehensive update on the latest research progress in CRBN-based PRTOACs area. Following the criteria, such as disease area and drug target class, we will present the degradants in alphabetical order by target. We also provide our own perspective on the future prospects and potential challenges facing PROTACs.
Collapse
Affiliation(s)
- Chao Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong, China.
| | - Yudong Wu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
| | - Dongming Xing
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
33
|
Heterogeneous modulation of Bcl-2 family members and drug efflux mediate MCL-1 inhibitor resistance in multiple myeloma. Blood Adv 2021; 5:4125-4139. [PMID: 34478517 PMCID: PMC8945627 DOI: 10.1182/bloodadvances.2020003826] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/07/2021] [Indexed: 01/19/2023] Open
Abstract
Antiapoptotic Bcl-2 family members have recently (re)emerged as key drug targets in cancer, with a tissue- and tumor-specific activity profile of available BH3 mimetics. In multiple myeloma, MCL-1 has been described as a major gatekeeper of apoptosis. This discovery has led to the rapid establishment of clinical trials evaluating the impact of various MCL-1 inhibitors. However, our understanding about the clinical impact and optimal use of MCL-1 inhibitors is still limited. We therefore explored mechanisms of acquired MCL-1 inhibitor resistance and optimization strategies in myeloma. Our findings indicated heterogeneous paths to resistance involving baseline Bcl-2 family alterations of proapoptotic (BAK, BAX, and BIM) and antiapoptotic (Bcl-2 and MCL-1) proteins. These manifestations depend on the BH3 profile of parental cells that guide the enhanced formation of Bcl-2:BIM and/or the dynamic (ie, treatment-induced) formation of Bcl-xL:BIM and Bcl-xL:BAK complexes. Accordingly, an unbiased high-throughput drug-screening approach (n = 528) indicated alternative BH3 mimetics as top combination partners for MCL-1 inhibitors in sensitive and resistant cells (Bcl-xL>Bcl-2 inhibition), whereas established drug classes were mainly antagonistic (eg, antimitotic agents). We also revealed reduced activity of MCL-1 inhibitors in the presence of stromal support as a drug-class effect that was overcome by concurrent Bcl-xL or Bcl-2 inhibition. Finally, we demonstrated heterogeneous Bcl-2 family deregulation and MCL-1 inhibitor cross-resistance in carfilzomib-resistant cells, a phenomenon linked to the MDR1-driven drug efflux of MCL-1 inhibitors. The implications of our findings for clinical practice emphasize the need for patient-adapted treatment protocols, with the tracking of tumor- and/or clone-specific adaptations in response to MCL-1 inhibition.
Collapse
|
34
|
DKK1 activates noncanonical NF-κB signaling via IL-6-induced CKAP4 receptor in multiple myeloma. Blood Adv 2021; 5:3656-3667. [PMID: 34470047 DOI: 10.1182/bloodadvances.2021004315] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/12/2021] [Indexed: 01/16/2023] Open
Abstract
Proteasome inhibitors, such as bortezomib (BTZ), represent the key elements in chemotherapy regimens for multiple myeloma (MM), whereas acquired chemoresistance and ultimately relapse remain a major obstacle. In the current study, we screened differently expressed cytokines in bortezomib-resistant MM cells and found that Dickkopf-1 (DKK1) level was remarkably augmented, whereas CD138 level was significantly suppressed. DKK1 in vitro specifically enhanced the resistance of myeloma cells to bortezomib treatment, and excessive DKK1 drove CD138 downregulation via inhibition of canonical Wnt signaling. Notably, DKK1 mainly induced drug resistance in MM cells via the receptor of CKAP4. Mechanistically, CKAP4 transduced DKK1 signal and evoked NF-κB pathway through recruiting and preventing cullin associated and neddylation dissociated 1 from hampering the assembly of E3 ligase-mediated ubiquitination of IκBα. In addition, we found that interleukin-6 (IL-6) stimulated CKAP4 expression to generate drug resistance, and disturbance of DKK1-CKAP4 axis improved sensitivity to BTZ treatment of MM and attenuated bone destruction in a mouse model. Collectively, our study revealed the previously unidentified role of DKK1 in myeloma drug resistance via Wnt signaling dependent and independent manners, and clarified the importance of antagonism of DKK1-IL-6 loop in bone marrow microenvironment.
Collapse
|
35
|
Burger R, Otte A, Brdon J, Peipp M, Gramatzki M. Dual intracellular targeting by ruxolitinib and the Mcl-1 inhibitor S63845 in interleukin-6-dependent myeloma cells blocks. Haematologica 2021; 106:2507-2510. [PMID: 33882638 PMCID: PMC8409037 DOI: 10.3324/haematol.2020.276865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 11/28/2022] Open
Affiliation(s)
- Renate Burger
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel.
| | - Anna Otte
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel
| | - Jan Brdon
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel
| |
Collapse
|
36
|
Nahi H, Kashif M, Klimkowska M, Karvouni M, Wallblom A, Gran C, Hauenstein J, Frengen N, Gustafsson C, Afram G, Uttervall K, Lund J, Månsson R, Wagner AK, Alici E. Low dose venetoclax as a single agent treatment of plasma cell malignancies harboring t(11;14). Am J Hematol 2021; 96:925-933. [PMID: 33901326 DOI: 10.1002/ajh.26207] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Approximately 20% of newly diagnosed multiple myeloma (NDMM) patients harbor t(11;14), a marker of inferior prognosis, resulting in up-regulation of CCND1. These patients respond to BCL2 inhibitor experimental drug venetoclax. Furthermore, t(11;14) is reported to be associated with increased BCL2/MCL1 ratio. We investigated the use of venetoclax (400 mg daily) in a cohort of 25 multiple myeloma (MM) and AL-amyloidosis patients harboring t(11;14) and assessed safety and efficacy. Efficacy was assessed by response rate (RR) and time on treatment. Furthermore, immunohistochemistry (IHC), for BCL2 family member expression was assessed at diagnosis and relapse in the venetoclax-treated group and analyzed for correlation with clinical RR. Additionally, patient material from venetoclax non-treated group including non-t(11;14) diagnosis (n = 27), t(11;14) diagnosis (n = 17), t(11;14) relapse (n = 7), hyperdiploidy (n = 6) and hyperdiploidy + t(11;14) (n = 6) was used for RNA sequencing (RNASeq) and validation by qPCR. Venetoclax treatment in t(11;14) patients demonstrated manageable safety and promising efficacy. Partial responses or better were observed in eleven patients (44%). Responding patients had significantly higher BCL2/MCL1 (p = 0.031) as well as BCL2/BCL-XL (p = 0.021) ratio, regardless of time of measurement before venetoclax treatment. Furthermore, an IRF5 motif was enriched (p < .001) in the downregulated genes in t(11;14) relapses vs diagnoses. The RR with single agent venetoclax was 71% in AL-amyloidosis and 33% in MM, and IHC proved useful in prediction of treatment outcome. We could also demonstrate possible resistance mechanisms of t(11;14), downregulation of IRF5 targeted genes, which can be exploited for therapeutic advantages.
Collapse
Affiliation(s)
- Hareth Nahi
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Muhammad Kashif
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
| | - Monika Klimkowska
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
- Department of Clinical Pathology and Cytology Karolinska University Hospital Stockholm Sweden
| | - Maria Karvouni
- Department of Medicine Karolinska Institutet Stockholm Sweden
| | - Ann Wallblom
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
| | - Charlotte Gran
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Clinical Chemistry Karolinska University Hospital Stockholm Sweden
| | - Julia Hauenstein
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Nicolai Frengen
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Charlotte Gustafsson
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Gabriel Afram
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Katarina Uttervall
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Johan Lund
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Robert Månsson
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | | | - Evren Alici
- Department of Medicine Karolinska Institutet Stockholm Sweden
| |
Collapse
|
37
|
Gupta VA, Barwick BG, Matulis SM, Shirasaki R, Jaye DL, Keats JJ, Oberlton B, Joseph NS, Hofmeister CC, Heffner LT, Dhodapkar MV, Nooka AK, Lonial S, Mitsiades CS, Kaufman JL, Boise LH. Venetoclax sensitivity in multiple myeloma is associated with B-cell gene expression. Blood 2021; 137:3604-3615. [PMID: 33649772 PMCID: PMC8462405 DOI: 10.1182/blood.2020007899] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 01/29/2021] [Indexed: 01/31/2023] Open
Abstract
Venetoclax is a highly potent, selective BCL2 inhibitor capable of inducing apoptosis in cells dependent on BCL2 for survival. Most myeloma is MCL1-dependent; however, a subset of myeloma enriched for translocation t(11;14) is codependent on BCL2 and thus sensitive to venetoclax. The biology underlying this heterogeneity remains poorly understood. We show that knockdown of cyclin D1 does not induce resistance to venetoclax, arguing against a direct role for cyclin D1 in venetoclax sensitivity. To identify other factors contributing to venetoclax response, we studied a panel of 31 myeloma cell lines and 25 patient samples tested for venetoclax sensitivity. In cell lines, we corroborated our previous observation that BIM binding to BCL2 correlates with venetoclax response and further showed that knockout of BIM results in decreased venetoclax sensitivity. RNA-sequencing analysis identified expression of B-cell genes as enriched in venetoclax-sensitive myeloma, although no single gene consistently delineated sensitive and resistant cells. However, a panel of cell surface makers correlated well with ex vivo prediction of venetoclax response in 21 patient samples and may serve as a biomarker independent of t(11;14). Assay for transposase-accessible chromatin sequencing of myeloma cell lines also identified an epigenetic program in venetoclax-sensitive cells that was more similar to B cells than that of venetoclax-resistant cells, as well as enrichment for basic leucine zipper domain-binding motifs such as BATF. Together, these data indicate that remnants of B-cell biology are associated with BCL2 dependency and point to novel biomarkers of venetoclax-sensitive myeloma independent of t(11;14).
Collapse
MESH Headings
- B-Lymphocytes/metabolism
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Line, Tumor
- Chromosomes, Human, Pair 11/genetics
- Chromosomes, Human, Pair 11/metabolism
- Chromosomes, Human, Pair 14/genetics
- Chromosomes, Human, Pair 14/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Epigenesis, Genetic/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Knockdown Techniques
- Humans
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Sulfonamides/pharmacology
- Translocation, Genetic/drug effects
Collapse
Affiliation(s)
- Vikas A Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Benjamin G Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Shannon M Matulis
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Ryosuke Shirasaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - David L Jaye
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute of Emory University, Atlanta, GA; and
| | - Jonathan J Keats
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Benjamin Oberlton
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Nisha S Joseph
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Leonard T Heffner
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | | | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| |
Collapse
|
38
|
Cytokine-like protein 1-induced survival of monocytes suggests a combined strategy targeting MCL1 and MAPK in CMML. Blood 2021; 137:3390-3402. [PMID: 33690800 DOI: 10.1182/blood.2020008729] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/11/2021] [Indexed: 12/20/2022] Open
Abstract
Mouse models of chronic myeloid malignancies suggest that targeting mature cells of the malignant clone disrupts feedback loops that promote disease expansion. Here, we show that in chronic myelomonocytic leukemia (CMML), monocytes that accumulate in the peripheral blood show a decreased propensity to die by apoptosis. BH3 profiling demonstrates their addiction to myeloid cell leukemia-1 (MCL1), which can be targeted with the small molecule inhibitor S63845. RNA sequencing and DNA methylation pattern analysis both point to the implication of the mitogen-activated protein kinase (MAPK) pathway in the resistance of CMML monocytes to death and reveal an autocrine pathway in which the secreted cytokine-like protein 1 (CYTL1) promotes extracellular signal-regulated kinase (ERK) activation through C-C chemokine receptor type 2 (CCR2). Combined MAPK and MCL1 inhibition restores apoptosis of monocytes from patients with CMML and reduces the expansion of patient-derived xenografts in mice. These results show that the combined inhibition of MCL1 and MAPK is a promising approach to slow down CMML progression by inducing leukemic monocyte apoptosis.
Collapse
|
39
|
The spleen as a sanctuary site for residual leukemic cells following ABT-199 monotherapy in ETP-ALL. Blood Adv 2021; 5:1963-1976. [PMID: 33830207 PMCID: PMC8045507 DOI: 10.1182/bloodadvances.2021004177] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
B-cell lymphoma 2 (BCL-2) has recently emerged as a therapeutic target for early T-cell progenitor acute lymphoblastic leukemia (ETP-ALL), a high-risk subtype of human T-cell ALL. The major clinical challenge with targeted therapeutics, such as the BCL-2 inhibitor ABT-199, is the development of acquired resistance. We assessed the in vivo response of luciferase-positive LOUCY cells to ABT-199 monotherapy and observed specific residual disease in the splenic microenvironment. Of note, these results were confirmed by using a primary ETP-ALL patient-derived xenograft. Splenomegaly has previously been associated with poor prognosis in diverse types of leukemia. However, the exact mechanism by which the splenic microenvironment alters responses to specific targeted therapies remains largely unexplored. We show that residual LOUCY cells isolated from the spleen microenvironment displayed reduced BCL-2 dependence, which was accompanied by decreased BCL-2 expression levels. Notably, this phenotype of reduced BCL-2 dependence could be recapitulated by using human splenic fibroblast coculture experiments and was confirmed in an in vitro chronic ABT-199 resistance model of LOUCY. Finally, single-cell RNA-sequencing was used to show that ABT-199 triggers transcriptional changes in T-cell differentiation genes in leukemic cells obtained from the spleen microenvironment. Of note, increased expression of CD1a and sCD3 was also observed in ABT199-resistant LOUCY clones, further reinforcing the idea that a more differentiated leukemic population might display decreased sensitivity toward BCL-2 inhibition. Overall, our data reveal the spleen as a site of residual disease for ABT-199 treatment in ETP-ALL and provide evidence for plasticity in T-cell differentiation as a mechanism of therapy resistance.
Collapse
|
40
|
Zhu P, Qian T, Si C, Liu Y, Cui L, Huang W, Fu L, Deng C, Zeng T. High expression of CPNE5 and CPNE9 predicts positive prognosis in multiple myeloma. Cancer Biomark 2021; 31:77-85. [PMID: 33780365 DOI: 10.3233/cbm-203108] [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] [Indexed: 12/14/2022]
Abstract
BACKGROUND CPNEs are significant biomarkers which can affect the progression and prognosis of various tumor diseases. However, the prognosis role of CPNEs in multiple myeloma (MM) is still unclear. OBJECTIVES To investigate the prognosis role of CPNEs in MM. METHODS Seven hundred and thirty-five samples from two independent data sets were involved to analyze the clinical and molecular characteristics, and prognosis role of the expression of CPNE1-9 in MM. RESULTS MM patients with higher expressions of CPNE5 and CPNE9 had longer event-free survival (EFS) and overall survival (OS) compared with CPNE5low and CPNE9low expression groups (EFS: P= 0.0054, 0.0065; OS: P= 0.015, 0.016, respectively). Multivariate regression analysis showed that CPNE5 was an independent favorable predictor for EFS and OS (EFS: P= 0.005; OS: P= 0.006), and CPNE9 was an independent positive indicator for EFS (P= 0.002). Moreover, the survival probability and the cumulative event of EFS and OS in CPNE5highCPNE9high group were significantly longer than other groups. CONCLUSIONS High expressions of CPNE5 and CPNE9 might be used as positive indicators for MM, and their combination was a better predictor for the survival of MM patients.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tingting Qian
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chaozeng Si
- Information Center, China-Japan Friendship Hospital, Beijing, China.,Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yan Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, Henan, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Longzhen Cui
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, Henan, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Wenhui Huang
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lin Fu
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, Henan, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Cong Deng
- Department of Clinical Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tiansheng Zeng
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
41
|
Schmidt TM, Fonseca R, Usmani SZ. Chromosome 1q21 abnormalities in multiple myeloma. Blood Cancer J 2021; 11:83. [PMID: 33927196 PMCID: PMC8085148 DOI: 10.1038/s41408-021-00474-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Gain of chromosome 1q (+1q) is one of the most common recurrent cytogenetic abnormalities in multiple myeloma (MM), occurring in approximately 40% of newly diagnosed cases. Although it is often considered a poor prognostic marker in MM, +1q has not been uniformly adopted as a high-risk cytogenetic abnormality in guidelines. Controversy exists regarding the importance of copy number, as well as whether +1q is itself a driver of poor outcomes or merely a common passenger genetic abnormality in biologically unstable disease. Although the identification of a clear pathogenic mechanism from +1q remains elusive, many genes at the 1q21 locus have been proposed to cause early progression and resistance to anti-myeloma therapy. The plethora of potential drivers suggests that +1q is not only a causative factor or poor outcomes in MM but may be targetable and/or predictive of response to novel therapies. This review will summarize our current understanding of the pathogenesis of +1q in plasma cell neoplasms, the impact of 1q copy number, identify potential genetic drivers of poor outcomes within this subset, and attempt to clarify its clinical significance and implications for the management of patients with multiple myeloma.
Collapse
Affiliation(s)
| | - Rafael Fonseca
- Department of Hematology, Mayo Clinic, Scottsdale, AZ, USA
| | - Saad Z Usmani
- Plasma Cell Disorders Division, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA.
| |
Collapse
|
42
|
Lernoux M, Schnekenburger M, Dicato M, Diederich M. Susceptibility of multiple myeloma to B-cell lymphoma 2 family inhibitors. Biochem Pharmacol 2021; 188:114526. [PMID: 33741332 DOI: 10.1016/j.bcp.2021.114526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 01/18/2023]
Abstract
Multiple myeloma (MM) is a biologically complex hematological disorder defined by the clonal proliferation of malignant plasma cells producing excessive monoclonal immunoglobulin that interacts with components of the bone marrow microenvironment, resulting in the major clinical features of MM. Despite the development of numerous protocols to treat MM patients, this cancer remains currently incurable; due in part to the emergence of resistant clones, highlighting the unmet need for innovative therapeutic approaches. Accumulating evidence suggests that the survival of MM molecular subgroups depends on the expression profiles of specific subsets of anti-apoptotic B-cell lymphoma (BCL)-2 family members. This review summarizes the mechanisms underlying the anti-myeloma activities of the potent BCL-2 family protein inhibitors, individually or in combination with conventional therapeutic options, and provides an overview of the strong rationale to clinically investigate such interventions for MM therapy.
Collapse
Affiliation(s)
- Manon Lernoux
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
43
|
Gupta VA, Ackley J, Kaufman JL, Boise LH. BCL2 Family Inhibitors in the Biology and Treatment of Multiple Myeloma. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2021; 11:11-24. [PMID: 33737856 PMCID: PMC7965688 DOI: 10.2147/blctt.s245191] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022]
Abstract
Although much progress has been made in the treatment of multiple myeloma, the majority of patients fail to be cured and require numerous lines of therapy. Inhibitors of the BCL2 family represent an exciting new class of drugs with a novel mechanism of action that are likely to have activity as single agents and in combination with existing myeloma therapies. The BCL2 proteins are oncogenes that promote cell survival and are frequently upregulated in multiple myeloma, making them attractive targets. Venetoclax, a BCL2 specific inhibitor, is furthest along in development and has shown promising results in a subset of myeloma characterized by the t(11;14) translocation. Combining venetoclax with proteasome inhibitors and monoclonal antibodies has improved responses in a broader group of patients, but has come at the expense of a toxicity safety signal that requires additional follow-up. MCL1 inhibitors are likely to be effective in a broader range of patients and are currently in early clinical trials. This review will cover much of what is known about the biology of these drugs, biomarkers that predict response, mechanisms of resistance, and unanswered questions as they pertain to multiple myeloma.
Collapse
Affiliation(s)
- Vikas A Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - James Ackley
- Cancer Biology Graduate Program, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
44
|
Algarín EM, Quwaider D, Campos-Laborie FJ, Díaz-Tejedor A, Mogollón P, Vuelta E, Martín-Sánchez M, San-Segundo L, González-Méndez L, Gutiérrez NC, García-Sanz R, Paíno T, De Las Rivas J, Ocio EM, Garayoa M. Stroma-Mediated Resistance to S63845 and Venetoclax through MCL-1 and BCL-2 Expression Changes Induced by miR-193b-3p and miR-21-5p Dysregulation in Multiple Myeloma. Cells 2021; 10:cells10030559. [PMID: 33806619 PMCID: PMC8001939 DOI: 10.3390/cells10030559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/20/2021] [Accepted: 02/27/2021] [Indexed: 12/18/2022] Open
Abstract
BH3-mimetics targeting anti-apoptotic proteins such as MCL-1 (S63845) or BCL-2 (venetoclax) are currently being evaluated as effective therapies for the treatment of multiple myeloma (MM). Interleukin 6, produced by mesenchymal stromal cells (MSCs), has been shown to modify the expression of anti-apoptotic proteins and their interaction with the pro-apoptotic BIM protein in MM cells. In this study, we assess the efficacy of S63845 and venetoclax in MM cells in direct co-culture with MSCs derived from MM patients (pMSCs) to identify additional mechanisms involved in the stroma-induced resistance to these agents. MicroRNAs miR-193b-3p and miR-21-5p emerged among the top deregulated miRNAs in myeloma cells when directly co-cultured with pMSCs, and we show their contribution to changes in MCL-1 and BCL-2 protein expression and in the activity of S63845 and venetoclax. Additionally, direct contact with pMSCs under S63845 and/or venetoclax treatment modifies myeloma cell dependence on different BCL-2 family anti-apoptotic proteins in relation to BIM, making myeloma cells more dependent on the non-targeted anti-apoptotic protein or BCL-XL. Finally, we show a potent effect of the combination of S63845 and venetoclax even in the presence of pMSCs, which supports this combinatorial approach for the treatment of MM.
Collapse
Affiliation(s)
- Esperanza M. Algarín
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Dalia Quwaider
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Francisco J. Campos-Laborie
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca (USAL) and Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (F.J.C.-L.); (J.D.L.R.)
- The Gurdon Institute (Wellcome Trust/Cancer Research UK), University of Cambridge, Cambridge CB2 1QN, UK
| | - Andrea Díaz-Tejedor
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Pedro Mogollón
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Elena Vuelta
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Montserrat Martín-Sánchez
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Laura San-Segundo
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Lorena González-Méndez
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Norma C. Gutiérrez
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Center for Biomedical Research in Network of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Ramón García-Sanz
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Center for Biomedical Research in Network of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Teresa Paíno
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Center for Biomedical Research in Network of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca (USAL) and Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (F.J.C.-L.); (J.D.L.R.)
| | - Enrique M. Ocio
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39011 Santander, Spain;
| | - Mercedes Garayoa
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Correspondence: ; Tel.: +34-923-295812
| |
Collapse
|
45
|
Phosphatase PP2A enhances MCL-1 protein half-life in multiple myeloma cells. Cell Death Dis 2021; 12:229. [PMID: 33658484 PMCID: PMC7930201 DOI: 10.1038/s41419-020-03351-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 01/31/2023]
Abstract
Multiple myeloma (MM), a treatable but incurable malignancy, is characterized by the growth of clonal plasma cells in protective niches in the bone marrow. MM cells depend on expression of BCL-2 family proteins, in particular MCL-1, for survival. The regulation of MCL-1 is complex and cell type-dependent. Unraveling the exact mechanism by which MCL-1 is overexpressed in MM may provide new therapeutic strategies for inhibition in malignant cells, preferably limiting side effects in healthy cells. In this study, we reveal that one cause of overexpression could be stabilization of the MCL-1 protein. We demonstrate this in a subset of MM and diffuse large B cell lymphoma (DLBCL) cell lines and MM patient samples. We applied a phosphatase siRNA screen to identify phosphatases responsible for MCL-1 stabilization in MM, and revealed PP2A as the MCL-1 stabilizing phosphatase. Using the PP2A inhibitor okadaic acid, we validated that PP2A dephosphorylates MCL-1 at Ser159 and/or Thr163, and thereby stabilizes MCL-1 in MM cells with long MCL-1 half-life, but not in DLBCL cells. Combined kinase and phosphatase inhibition experiments suggest that the MCL-1 half-life in MM is regulated by the counteracting functions of JNK and PP2A. These findings increase the understanding of the mechanisms by which MCL-1 is post-translationally regulated, which may provide novel strategies to inhibit MCL-1 in MM cells.
Collapse
|
46
|
Moser-Katz T, Joseph NS, Dhodapkar MV, Lee KP, Boise LH. Game of Bones: How Myeloma Manipulates Its Microenvironment. Front Oncol 2021; 10:625199. [PMID: 33634031 PMCID: PMC7900622 DOI: 10.3389/fonc.2020.625199] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma is a clonal disease of long-lived plasma cells and is the second most common hematological cancer behind Non-Hodgkin's Lymphoma. Malignant transformation of plasma cells imparts the ability to proliferate, causing harmful lesions in patients. In advanced stages myeloma cells become independent of their bone marrow microenvironment and form extramedullary disease. Plasma cells depend on a rich array of signals from neighboring cells within the bone marrow for survival which myeloma cells exploit for growth and proliferation. Recent evidence suggests, however, that both the myeloma cells and the microenvironment have undergone alterations as early as during precursor stages of the disease. There are no current therapies routinely used for treating myeloma in early stages, and while recent therapeutic efforts have improved patients' median survival, most will eventually relapse. This is due to mutations in myeloma cells that not only allow them to utilize its bone marrow niche but also facilitate autocrine pro-survival signaling loops for further progression. This review will discuss the stages of myeloma cell progression and how myeloma cells progress within and outside of the bone marrow microenvironment.
Collapse
Affiliation(s)
- Tyler Moser-Katz
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Nisha S. Joseph
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Madhav V. Dhodapkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Kelvin P. Lee
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, United States
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| |
Collapse
|
47
|
Grundy M, Al‐Kaisi F, Cull J, Williams C, Smith D, Seedhouse CH. A same-day assay predicts apoptotic response to combined BCL-2 and MCL-1 BH3-mimetic targeting in multiple myeloma cells. EJHAEM 2021; 2:40-47. [PMID: 35846088 PMCID: PMC9175957 DOI: 10.1002/jha2.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 12/01/2022]
Abstract
Recent advances in treatment options for multiple myeloma (MM) have positive impact on patient survival. However, there is a short fall of rapid and reliable assays that can predict patient response to novel agents. The anti-apoptotic proteins B-cell lymphoma-2 (BCL-2) and myeloid cell leukaemia-1 (MCL-1), are necessary for MM survival, although most myelomas are more dependent on MCL-1. BCL-2 inhibition alone yields significant cytotoxicity in only a minority of cases, therefore targeting both proteins simultaneously, is a therapeutic option. Venetoclax and S63845 are BCL-2 and MCL-1 targeting BH3-mimetics which have demonstrated apoptotic synergy in MM. We investigated whether a novel short-term flow cytometric cytochrome c release assay could predict response to dual BH3-mimetic targeting in MM cells. Six human myeloma cell lines (HMCL) and seven primary samples were treated with venetoclax and S63845 alone or in combination. The 4-hour assay confirmed the drug combination was synergistic in all HMCL tested. Annexin-V data at 48 hours corresponded with 4-hour response verifying the assay as a predictor of drug sensitivity. All primary samples responded to the drug combination, including samples with 1q gain and t(4;14) translocation. Normal stem cells were unaffected by the drug combination. We have developed a novel assay with the potential to predict response to therapy in MM cells.
Collapse
Affiliation(s)
- Martin Grundy
- Blood Cancer and Stem Cells, Division of Cancer and Stem CellsSchool of MedicineUniversity of Nottingham Biodiscovery InstituteNottinghamUK
- Clinical HaematologyNottingham University HospitalsNottinghamUK
| | - Firas Al‐Kaisi
- Clinical HaematologyNottingham University HospitalsNottinghamUK
| | - Joanna Cull
- Clinical HaematologyNottingham University HospitalsNottinghamUK
| | - Cathy Williams
- Clinical HaematologyNottingham University HospitalsNottinghamUK
| | - Dean Smith
- Clinical HaematologyNottingham University HospitalsNottinghamUK
| | - Claire H. Seedhouse
- Blood Cancer and Stem Cells, Division of Cancer and Stem CellsSchool of MedicineUniversity of Nottingham Biodiscovery InstituteNottinghamUK
| |
Collapse
|
48
|
Bolomsky A, Vogler M, Köse MC, Heckman CA, Ehx G, Ludwig H, Caers J. MCL-1 inhibitors, fast-lane development of a new class of anti-cancer agents. J Hematol Oncol 2020; 13:173. [PMID: 33308268 PMCID: PMC7731749 DOI: 10.1186/s13045-020-01007-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022] Open
Abstract
Cell death escape is one of the most prominent features of tumor cells and closely linked to the dysregulation of members of the Bcl-2 family of proteins. Among those, the anti-apoptotic family member myeloid cell leukemia-1 (MCL-1) acts as a master regulator of apoptosis in various human malignancies. Irrespective of its unfavorable structure profile, independent research efforts recently led to the generation of highly potent MCL-1 inhibitors that are currently evaluated in clinical trials. This offers new perspectives to target a so far undruggable cancer cell dependency. However, a detailed understanding about the tumor and tissue type specific implications of MCL-1 are a prerequisite for the optimal (i.e., precision medicine guided) use of this novel drug class. In this review, we summarize the major functions of MCL-1 with a special focus on cancer, provide insights into its different roles in solid vs. hematological tumors and give an update about the (pre)clinical development program of state-of-the-art MCL-1 targeting compounds. We aim to raise the awareness about the heterogeneous role of MCL-1 as drug target between, but also within tumor entities and to highlight the importance of rationale treatment decisions on a case by case basis.
Collapse
Affiliation(s)
- Arnold Bolomsky
- Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Meike Vogler
- Department of Clinical Hematology, GIGA-I3, University of Liège, CHU De Liège, 35, Dom Univ Sart Tilman B, 4000, Liège, Belgium
| | - Murat Cem Köse
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland-FIMM, HiLIFE-Helsinki Institute of Life Science, iCAN Digital Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Grégory Ehx
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Jo Caers
- Department of Clinical Hematology, GIGA-I3, University of Liège, CHU De Liège, 35, Dom Univ Sart Tilman B, 4000, Liège, Belgium.
| |
Collapse
|
49
|
Lin VS, Xu ZF, Huang DCS, Thijssen R. BH3 Mimetics for the Treatment of B-Cell Malignancies-Insights and Lessons from the Clinic. Cancers (Basel) 2020; 12:cancers12113353. [PMID: 33198338 PMCID: PMC7696913 DOI: 10.3390/cancers12113353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
Simple Summary B-cell malignancies, including chronic lymphocytic leukemia (CLL), non-Hodgkin lymphoma (NHL), and plasma cell dyscrasias, are significant contributors to cancer morbidity and mortality worldwide. The pathogenesis of many B-cell malignancies involves perturbations in the intrinsic pathway of apoptosis that allow cells to evade cell death. BH3 mimetics represent a class of anti-cancer agents that can restore the ability of cancer cells to undergo apoptosis. Venetoclax, a recently approved BH3 mimetic, has transformed the therapeutic landscape for CLL. Other BH3 mimetics are currently under development. This review summarizes the available data on existing BH3 mimetics and highlights both the rapidly expanding role of BH3 mimetics in the treatment of B-cell malignancies and the clinical challenges of their use. Abstract The discovery of the link between defective apoptotic regulation and cancer cell survival engendered the idea of targeting aberrant components of the apoptotic machinery for cancer therapy. The intrinsic pathway of apoptosis is tightly controlled by interactions amongst members of three distinct subgroups of the B-cell lymphoma 2 (BCL2) family of proteins. The pro-survival BCL2 proteins prevent apoptosis by keeping the pro-apoptotic effector proteins BCL2-associated X protein (BAX) and BCL2 homologous antagonist/killer (BAK) in check, while the BH3-only proteins initiate apoptosis by either neutralizing the pro-survival BCL2 proteins or directly activating the pro-apoptotic effector proteins. This tripartite regulatory mechanism is commonly perturbed in B-cell malignancies facilitating cell death evasion. Over the past two decades, structure-based drug discovery has resulted in the development of a series of small molecules that mimic the function of BH3-only proteins called the BH3 mimetics. The most clinically advanced of these is venetoclax, which is a highly selective inhibitor of BCL2 that has transformed the treatment landscape for chronic lymphocytic leukemia (CLL). Other BH3 mimetics, which selectively target myeloid cell leukemia 1 (MCL1) and B-cell lymphoma extra large (BCLxL), are currently under investigation for use in diverse malignancies. Here, we review the current role of BH3 mimetics in the treatment of CLL and other B-cell malignancies and address open questions in this rapidly evolving field.
Collapse
Affiliation(s)
- Victor S. Lin
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, 3052 Parkville, Australia; (V.S.L.); (Z.-F.X.); (D.C.S.H.)
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, 3000 Melbourne, Australia
| | - Zhuo-Fan Xu
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, 3052 Parkville, Australia; (V.S.L.); (Z.-F.X.); (D.C.S.H.)
- School of Medicine, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing 100084, China
| | - David C. S. Huang
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, 3052 Parkville, Australia; (V.S.L.); (Z.-F.X.); (D.C.S.H.)
- Department of Medical Biology, University of Melbourne, 3000 Melbourne, Australia
| | - Rachel Thijssen
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, 3052 Parkville, Australia; (V.S.L.); (Z.-F.X.); (D.C.S.H.)
- Department of Medical Biology, University of Melbourne, 3000 Melbourne, Australia
- Correspondence:
| |
Collapse
|
50
|
We need CD38 STAT-JAK. Blood 2020; 136:2246-2248. [PMID: 33180919 DOI: 10.1182/blood.2020007467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|