1
|
Chen KY, Hsueh SC, Parekh P, Batsaikhan B, Tweedie D, Luo W, Patel C, Chiang YH, Bambakidis N, Hoffer BJ, Huang CZ, Yu SJ, Wu KJ, Wang Y, Hong E, Kim DS, Greig NH. 3-Monothiopomalidomide, a new immunomodulatory imide drug (IMiD), blunts inflammation and mitigates ischemic stroke in the rat. GeroScience 2025:10.1007/s11357-025-01573-1. [PMID: 40095189 DOI: 10.1007/s11357-025-01573-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
An overactive neuroinflammatory response is often evident in the elderly and is a significant contributor to brain tissue damage following acute ischemic stroke. Such an inflammatory response is largely mediated by microglial cells and peripheral blood mononuclear cells (PBMCs). Classical anti-inflammatory agents have not proved clinically effective in mitigating the impact of ischemic stroke but have highlighted targets for new drug development, in particular excessive proinflammatory cytokine release. The immunomodulatory imide drug (IMiD) class has shown potential in reducing neuroinflammation and switching microglial phenotypic expression away from a proinflammatory to a regenerative anti-inflammatory one. 3-Monothiopomalidomide (3-MP), a new IMiD, has a brain/plasma concentration ratio of 0.5 to 0.6, an oral bioavailability of 38.5%, and a monophasic disappearance of half-life 3.2 h following oral administration. 3-MP pretreatment mitigates lipopolysaccharide (LPS)-induced inflammation in cellular human PBMCs and, in rat studies, 3-MP pretreatment lowers proinflammatory cytokine levels in the conditioned media and in plasma and the brain, respectively. Administered systemically to rats challenged with middle cerebral artery occlusion (MCAo) and reperfusion, 3-MP post-MCAo treatment reduced infarction volume; improved body asymmetry, a behavioral measure of stroke impact; and lowered inflammation. In summary, 3-MP exerted neuroprotective effects via anti-inflammatory actions against MCAo-induced ischemic injury and represents a therapeutic that warrants further investigation as a treatment for brain damage and related disorders associated with excessive inflammation.
Collapse
Affiliation(s)
- Kai-Yun Chen
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
- Neuroscience Research Center, Taipei Medical University, Taipei, 110, Taiwan
| | - Shih-Chang Hsueh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Pathik Parekh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Buyandelger Batsaikhan
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Weiming Luo
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Chirag Patel
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Yung-Hsiao Chiang
- Neuroscience Research Center, Taipei Medical University, Taipei, 110, Taiwan
- Department of Neurosurgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Nicholas Bambakidis
- Department of Neurosurgery, University Hospitals of Cleveland, Cleveland, OH, 44106, USA
| | - Barry J Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Cleveland, OH, 44106, USA
| | - Chi-Zong Huang
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
| | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Kuo-Jen Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Eunji Hong
- Aevis Bio Inc, Daejeon, 34141, Republic of Korea
| | - Dong Seok Kim
- Aevis Bio Inc, Daejeon, 34141, Republic of Korea.
- Aevisbio Inc, Gaithersburg, MD, 20878, USA.
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD, 21224, USA.
| |
Collapse
|
2
|
Moreno Rueda LY, Wang H, Akagi K, Dang M, Vora A, Qin L, Lee HC, Patel KK, Lin P, Mery DE, Zhan F, Shaughnessy JD, Yi Q, Song Y, Jiang B, Gillison ML, Thomas SK, Weber DM, Diao L, Wang J, Kuiatse I, Manasanch EE, Symer DE, Orlowski RZ. Single-cell analysis of neoplastic plasma cells identifies myeloma pathobiology mediators and potential targets. Cell Rep Med 2025; 6:101925. [PMID: 39855192 DOI: 10.1016/j.xcrm.2024.101925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/26/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025]
Abstract
Multiple myeloma is a clonal plasma cell (PC) dyscrasia that arises from precursors and has been studied utilizing approaches focused on CD138+ cells. By combining single-cell RNA sequencing (scRNA-seq) with scB-cell receptor sequencing (scBCR-seq), we differentiate monoclonal/neoplastic from polyclonal/normal PCs and find more dysregulated genes, especially in precursor patients, than we would have by analyzing bulk PCs. To determine whether this approach can identify oncogenes that contribute to disease pathobiology, mitotic arrest deficient-2 like-1 (MAD2L1) and S-adenosylmethionine synthase isoform type-2 (MAT2A) are validated as targets with drug-like molecules that suppress myeloma growth in preclinical models. Moreover, functional studies show a role of lysosomal-associated membrane protein family member-5 (LAMP5), which is uniquely expressed in neoplastic PCs, in tumor progression and aggressiveness via interactions with c-MYC. Finally, a monoclonal antibody recognizing cell-surface LAMP5 shows efficacy as an antibody-drug conjugate and in a chimeric antigen receptor-guided T-cell format. These studies provide additional insights into myeloma biology and identify potential targeted therapeutic approaches that can be applied to reverse myeloma progression.
Collapse
Affiliation(s)
- Luz Yurany Moreno Rueda
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hua Wang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keiko Akagi
- Department of Thoracic-Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Minghao Dang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amishi Vora
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Qin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hans C Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Krina K Patel
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pei Lin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David E Mery
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Fenghuang Zhan
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - John D Shaughnessy
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Qing Yi
- Department of Cancer Biology in Medicine, Houston Methodist Dr. Mary and Ron Neal Cancer Center, Houston, TX, USA
| | - Yang Song
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bo Jiang
- Department of Thoracic-Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maura L Gillison
- Department of Thoracic-Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sheeba K Thomas
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Donna M Weber
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Isere Kuiatse
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elisabet E Manasanch
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David E Symer
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Medicine, VA Boston Healthcare System, Boston, MA, USA
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
3
|
Bergsagel PL, Chesi M. Immunocompetent mouse models of multiple myeloma. Semin Hematol 2025; 62:50-57. [PMID: 39674742 PMCID: PMC11911088 DOI: 10.1053/j.seminhematol.2024.11.003] [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: 09/16/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 12/16/2024]
Abstract
Immunocompetent murine models of multiple myeloma are critical for understanding the pathogenesis of multiple myeloma and for the development of novel immunotherapeutics. Different models are available in Balb/c and C57Bl strains, each with different advantages and disadvantages. The availability of many transplantable cell lines allows for the conduct of experiments with large cohorts of mice bearing identical tumors, while cell lines that grow in vitro can be used for genetic manipulations. The introduction of human CRBN into these models allows for the study of IMiDs and cereblon based PROTACs in mice. New genetically engineered models based on germinal center cell activation of Nsd2 or Ccnd1 together with constitutive NFkB are being developed to model some of the important genetic subtypes of human multiple myeloma.
Collapse
Affiliation(s)
- Peter Leif Bergsagel
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, AZ 85259.
| | - Marta Chesi
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, AZ 85259
| |
Collapse
|
4
|
Xiong W, Xiao L, Duan R, Wang Q, Xian M, Zhang C, Su P, Li Y, Zhong L, Qian J, Zheng C, Yi Q. Depletion of myeloid-derived suppressor cells sensitizes murine multiple myeloma to PD-1 checkpoint inhibitors. J Immunother Cancer 2025; 13:e008979. [PMID: 39755583 PMCID: PMC11749690 DOI: 10.1136/jitc-2024-008979] [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: 01/31/2024] [Accepted: 11/13/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Cancer immunotherapy using immune checkpoint blockade (ICB) has revolutionized cancer treatment. However, patients with multiple myeloma (MM) rarely respond to ICB. Accumulating evidence indicates that the complicated tumor microenvironment (TME) significantly impacts the efficacy of ICB therapy. Therefore, investigating how TME components in MM influence ICB treatment is urgent. METHODS We employed two well-established murine myeloma models, 5TGM1 and Vk*MYC, by intravenously injecting 5TGM1 or Vk*MYC cells into mice, respectively, to determine ICB therapeutic efficacy in MM. Total mouse IgG or Ig2b ELISA or QuickGel split beta SPE kits and in vivo bioluminescent imaging were used to monitor MM tumor burden. Cytometry by time of flight (CyTOF) was used to quantify MM TME components. T cell proliferation and function were detected using flow cytometry. Peptide-Fc fusion proteins were used to deplete myeloid-derived suppressor cells (MDSCs). MMDTR, Foxp3DTR, CD4 KO and CD8 KO mice were used to elucidate the underlying mechanisms. Gene expression levels in human MM were analyzed using Gene Expression Omnibus public datasets. RESULTS We found that programmed cell death protein 1 (PD-1) antibody treatment had a therapeutic effect in 5TGM1 mice; it was ineffective in Vk*MYC mice. CyTOF indicated that the bone marrow (BM) of both models was inflamed, suggesting that immune suppressive cells might be inhibiting the reactivation of T cells in the BM. We observed higher numbers of MDSCs, regulatory T (Treg) cells, and tumor-associated macrophage (TAMs) in myeloma BM compared with that of tumor-free mice. Specifically, depleting MDSCs, but not Treg cells or TAMs, sensitized Vk*MYC mice and enhanced the response of 5TGM1 mice to PD-1 ICB, which was dependent on CD8+ but not CD4+ T cells. MDSCs, especially M-MDSCs and CD84+ MDSCs, significantly inhibited the activation and cytotoxic cytokine production of CD8+ T cells in vitro. Moreover, database profiling of patient BM revealed a negative correlation between MDSCs signature genes and cytotoxic CD8+ T cell signature genes, with post-maintenance patients with myeloma displaying a higher ratio of cytotoxic CD8+ T cell to MDSCs signature genes compared with pretreated patients. CONCLUSION Our study highlights the potential of MDSCs depletion in enhancing the sensitivity of patients with myeloma to PD-1 ICB therapy.
Collapse
Affiliation(s)
- Wei Xiong
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Liuling Xiao
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
- First Affiliated Hospital, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Rui Duan
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Qiang Wang
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Miao Xian
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Chuanchao Zhang
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Pan Su
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Yabo Li
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Ling Zhong
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Jianfei Qian
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Chengyun Zheng
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| | - Qing Yi
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
5
|
Platt JL, Zhao C, Chicca J, Pianko MJ, Han J, The S, Rao A, Keller ET, Garcia de Mattos Barbosa M, Naing L, Pasieka-Axenov T, Axenov L, Schaefer S, Farkash E, Cascalho M. Complement C3d enables cell-mediated immunity capable of distinguishing spontaneously transformed from nontransformed cells. Proc Natl Acad Sci U S A 2024; 121:e2405824121. [PMID: 39693340 DOI: 10.1073/pnas.2405824121] [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: 03/26/2024] [Accepted: 10/19/2024] [Indexed: 12/20/2024] Open
Abstract
Immune surveillance depends in part on the recognition of peptide variants by T cell antigen receptors. Given that both normal B cells and malignant B cells accumulate mutations we chose a murine model of multiple myeloma to test conditions to induce cell-mediated immunity targeting malignant plasma cell (PC) clones but sparing of normal PCs. Revealing a previously unknown function for intracellular C3d, we found that C3d engaged T cell responses against malignant PC in the bone marrow of mice that had developed multiple myeloma spontaneously. Our results show that C3d internalized by cells augments immune surveillance by several mechanisms. In one, C3d induces a master transcription regulator, E2f1, to increase the expression of long noncoding (lnc) RNAs, to generate peptides for MHC-I presentation, and increase MHC-I expression. In another, C3d increases expression of RNAs encoding ribosomal proteins linked to processing of defective ribosomal products that arise from noncanonical translation and known to promote immunosurveillance. Cancer cells are uniquely susceptible to increased expression and presentation of mutant peptides given the extent of protein misfolding and accumulation of somatic mutations. Accordingly, although C3d can be internalized by any cell, C3d preferentially targets malignant clones by evoking specific T cell-mediated immunity and sparing most nontransformed polyclonal B cells and PC with lower mutation loads. Malignant PC deletion was blocked by cyclosporin or by CD8 depletion confirming that endogenous T cells mediated malignant clone clearance. Besides the potential for therapeutic application our results highlight how intracellular C3d modifies cellular metabolism to augment immune surveillance.
Collapse
Affiliation(s)
- Jeffrey L Platt
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Chong Zhao
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Jeffrey Chicca
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Matthew J Pianko
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Joshua Han
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Stephanie The
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
- Cancer Data Science Shared Research Core, University of Michigan Rogel Cancer Center, Ann Arbor, MI 48109
| | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
- Cancer Data Science Shared Research Core, University of Michigan Rogel Cancer Center, Ann Arbor, MI 48109
| | - Evan T Keller
- Department of Urology and the Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | | | - Lwar Naing
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
- Department of Microbiology and Immunology, Graduate Program in Molecular and Cellular Biology, Ann Arbor, MI 48109
| | | | - Lev Axenov
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Simon Schaefer
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| | - Evan Farkash
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Marilia Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
6
|
Jiang H, Li Q, Yang X, Jia L, Cheng H, Wang J, Wang S, Li X, Xie Y, Wang J, Wang Y, Hu M, Guo J, Peng Z, Wang M, Li T, Zhao H, Wang L, Liu Z. Bone marrow stromal cells protect myeloma cells from ferroptosis through GPX4 deSUMOylation. Cancer Lett 2024; 611:217388. [PMID: 39653239 DOI: 10.1016/j.canlet.2024.217388] [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: 08/31/2024] [Revised: 11/18/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Bone marrow stromal cells (BMSCs) are vital for preventing chemotherapy induced apoptosis of multiple myeloma (MM), but roles and machinery in other forms of cell death have not been well elucidated. Here, using an in vitro BMSC-MM interacting model, we observed BMSCs protected MM cells from labile iron pool (LIP) and reactive oxygen species (ROS) triggered ferroptosis by elevating glutathione peroxidase 4 (GPX4). Mechanistically, direct interaction with BMSCs upregulated the expression of SUMO-specific protease 3 (SENP3) in MM cells through CD40/CD40L signaling pathway, and SENP3 de-conjugated SUMO2 at lysine 75 residue to stabilize GPX4 protein, thereby consuming ROS to obviate ferroptosis in MM cells from the Vk∗MYC mouse model, as well as in CD138+B220- cells separated from the Cd40lfl/fl;Prx1Cre/+ mice (CD40-CKO) and Sumo2 knock out (SUMO2-KO) mice. Using the NOD-scid IL2Rgammanull (NSG) mouse based xenograft model and intra-bone MM growth model, we validated that target SENP3 enhanced the killing effect of GPX4 inhibitor RSL3, thereby reduced tumor burden, prolonged survival of mice, and alleviated bone disruption of mice bearing MM tumors. Our study deciphers the mechanism of BMSCs preventing MM cells from spontaneous ferroptosis, and clarifies the therapeutic potential of non-apoptosis strategies in managing refractory or relapsed MM patients.
Collapse
Affiliation(s)
- Hongmei Jiang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China; Department of Pathology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Qian Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xudan Yang
- Department of Pathology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Linchuang Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Hao Cheng
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Jingya Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Sheng Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Xin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Ying Xie
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jingjing Wang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yixuan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Meilin Hu
- Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Jing Guo
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Ziyi Peng
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Mengqi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Tiantian Li
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Haifeng Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Lijuan Wang
- Central Laboratory, Linyi People's Hospital, Linyi, Shandong Province, 276037, China.
| | - Zhiqiang Liu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
7
|
Platt JL, Zhao C, Chicca J, Pianko MJ, Han J, The S, Rao A, Keller E, de Mattos Barbosa MG, Naing L, Pasieka-Axenov T, Axenov L, Schaefer S, Farkash E, Cascalho M. Complement C3d enables protective immunity capable of distinguishing spontaneously transformed from non-transformed cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606044. [PMID: 39211250 PMCID: PMC11360951 DOI: 10.1101/2024.07.31.606044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Immune-surveillance depends in part on the recognition of peptide variants by T cell antigen receptors. Given that both normal B cells and malignant B cells accumulate mutations we chose a murine model of multiple myeloma to test conditions to induce cell-mediated immunity targeting malignant plasma cell (PC) clones but sparing of normal PCs. Revealing a novel function for intracellular C3d, we discovered that C3d engaged T cell responses against malignant plasma cells in the bone marrow of mice that had developed multiple myeloma spontaneously. Our results show that C3d internalized by cells augments immune surveillance by several mechanisms. In one, C3d induces a master transcription regulator, E2f1, to increase the expression of long non-coding (lnc) RNAs, to generate peptides for MHC-I presentation and increase MHC-I expression. In another, C3d increases expression of RNAs encoding ribosomal proteins linked to processing of defective ribosomal products (DRiPs) that arise from non-canonical translation and known to promote immunosurveillance. Cancer cells are uniquely susceptible to increased expression and presentation of mutant peptides given the extent of protein misfolding and accumulation of somatic mutations. Accordingly, although C3d can be internalized by any cell, C3d preferentially targets malignant clones by evoking specific T cell mediated immunity (CMI) and sparing most non-transformed polyclonal B cells and plasma cells with lower mutation loads. Malignant plasma cell deletion was blocked by cyclosporin or by CD8 depletion confirming that endogenous T cells mediated malignant clone clearance. Besides the potential for therapeutic application our results highlight how intracellular C3d modifies cellular metabolism to augment immune surveillance. One Sentence Summary We show that intracellular soluble fragment 3d of complement (C3d) induces regression of spontaneous multiple myeloma in mice reducing tumor burden by 10 fold, after 8 weeks. C3d enables cell-mediated immunity to target multiple myeloma clones sparing non-transformed polyclonal B cells and plasma cells with lower mutation loads. We show that C3d increases the expression of ribosomal subunits associated with the translation of defective ribosomal products (DRiPs). C3d also decreases expression of protein arginine methyl transferase (PRMT) 5 which in turn relieves E2f1 repression increasing the expression of Lnc RNAs and derived peptides that evoke anti-tumor cellular immunity. The approach increases MHC-I expression by tumor cells and generates a CMI response that overcomes tumor immune-evasion strategies. Significance Tumors are immunogenic in part because of somatic mutations that originate novel peptides that once presented on MHC engage cell-mediated immunity (CMI). However, in spite of the higher mutation load most tumors evade immunity. We discovered that a component of the complement system (C3d) overcomes tumor immune evasion by augmenting expression of ribosomal proteins and lncRNAs linked to the presentation of novel peptides by tumor cells. C3d induced CMI targets cancer cells sparing non transformed cells uncovering a novel function for complement in immune surveillance.
Collapse
|
8
|
Lu K, Wang W, Liu Y, Xie C, Liu J, Xing L. Advancements in microenvironment-based therapies: transforming the landscape of multiple myeloma treatment. Front Oncol 2024; 14:1413494. [PMID: 39087026 PMCID: PMC11288838 DOI: 10.3389/fonc.2024.1413494] [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: 04/07/2024] [Accepted: 06/20/2024] [Indexed: 08/02/2024] Open
Abstract
Multiple myeloma (MM) is the most prevalent malignant monoclonal disease of plasma cells. There is mounting evidence that interactions with the bone marrow (BM) niche are essential for the differentiation, proliferation, survival, migration, and treatment resistance of myeloma cells. For this reason, gaining a deeper comprehension of how BM microenvironment compartments interact with myeloma cells may inspire new therapeutic ideas that enhance patient outcomes. This review will concentrate on the most recent findings regarding the mechanisms of interaction between microenvironment and MM and highlight research on treatment targeting the BM niche.
Collapse
Affiliation(s)
- Ke Lu
- Department of Lymphoma, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wen Wang
- Department of Lymphoma, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuntong Liu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Chao Xie
- Department of Respiratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jiye Liu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Lijie Xing
- Department of Lymphoma, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Key Laboratory of Biopharmaceuticals, Postdoctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan, Shandong, China
| |
Collapse
|
9
|
Maura F, Coffey DG, Stein CK, Braggio E, Ziccheddu B, Sharik ME, Du MT, Tafoya Alvarado Y, Shi CX, Zhu YX, Meermeier EW, Morgan GJ, Landgren O, Bergsagel PL, Chesi M. The genomic landscape of Vk*MYC myeloma highlights shared pathways of transformation between mice and humans. Nat Commun 2024; 15:3844. [PMID: 38714690 PMCID: PMC11076575 DOI: 10.1038/s41467-024-48091-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: 07/12/2023] [Accepted: 04/15/2024] [Indexed: 05/10/2024] Open
Abstract
Multiple myeloma (MM) is a heterogeneous disease characterized by frequent MYC translocations. Sporadic MYC activation in the germinal center of genetically engineered Vk*MYC mice is sufficient to induce plasma cell tumors in which a variety of secondary mutations are spontaneously acquired and selected over time. Analysis of 119 Vk*MYC myeloma reveals recurrent copy number alterations, structural variations, chromothripsis, driver mutations, apolipoprotein B mRNA-editing enzyme, catalytic polypeptide (APOBEC) mutational activity, and a progressive decrease in immunoglobulin transcription that inversely correlates with proliferation. Moreover, we identify frequent insertional mutagenesis by endogenous retro-elements as a murine specific mechanism to activate NF-kB and IL6 signaling pathways shared with human MM. Despite the increased genomic complexity associated with progression, advanced tumors remain dependent on MYC. In summary, here we credential the Vk*MYC mouse as a unique resource to explore MM genomic evolution and describe a fully annotated collection of diverse and immortalized murine MM tumors.
Collapse
Affiliation(s)
| | - David G Coffey
- Division of Myeloma, University of Miami, Miami, FL, USA
| | - Caleb K Stein
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Esteban Braggio
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Meaghen E Sharik
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Megan T Du
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Yuliza Tafoya Alvarado
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Chang-Xin Shi
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Yuan Xiao Zhu
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Erin W Meermeier
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Gareth J Morgan
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - Ola Landgren
- Division of Myeloma, University of Miami, Miami, FL, USA
| | - P Leif Bergsagel
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Marta Chesi
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA.
| |
Collapse
|
10
|
Jiang H, Wang L, Zhang Q, Wang S, Jia L, Cheng H, Wang J, Li X, Xie Y, Wang Y, Hu M, Guo J, Li Q, Peng Z, Wang M, Xie Y, Li T, Wang Y, Geng BD, Swaminathan S, Bergsagel PL, Liu Z. Bone marrow stromal cells dictate lanosterol biosynthesis and ferroptosis of multiple myeloma. Oncogene 2024; 43:1644-1653. [PMID: 38594504 PMCID: PMC11108777 DOI: 10.1038/s41388-024-03020-5] [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: 01/07/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Ferroptosis has been demonstrated a promising way to counteract chemoresistance of multiple myeloma (MM), however, roles and mechanism of bone marrow stromal cells (BMSCs) in regulating ferroptosis of MM cells remain elusive. Here, we uncovered that MM cells were more susceptible to ferroptotic induction under the interaction of BMSCs using in vitro and in vivo models. Mechanistically, BMSCs elevated the iron level in MM cells, thereby activating the steroid biosynthesis pathway, especially the production of lanosterol, a major source of reactive oxygen species (ROS) in MM cells. We discovered that direct coupling of CD40 ligand and CD40 receptor constituted the key signaling pathway governing lanosterol biosynthesis, and disruption of CD40/CD40L interaction using an anti-CD40 neutralizing antibody or conditional depletion of Cd40l in BMSCs successfully eliminated the iron level and lanosterol production of MM cells localized in the Vk*MYC Vk12653 or NSG mouse models. Our study deciphers the mechanism of BMSCs dictating ferroptosis of MM cells and highlights the therapeutic potential of non-apoptosis strategies for managing refractory or relapsed MM patients.
Collapse
Affiliation(s)
- Hongmei Jiang
- Department of Pathology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Lijuan Wang
- Central Laboratory, Linyi People's Hospital, Linyi, Shandong Province, 276037, China
| | - Qiguo Zhang
- Department of Hematology, The First People's Hospital of Chuzhou, Chuzhou Hospital Affiliated to Anhui Medical University, Chuzhou, 239000, China
- Department of Hematology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Sheng Wang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Linchuang Jia
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Hao Cheng
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Jingya Wang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Xin Li
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Ying Xie
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Yixuan Wang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Meilin Hu
- School of Stomatology, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Jing Guo
- Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Tianjin Key Laboratory of Cancer Prevention and Therapy; Tianjin's Clinical Research Center for Cancer, Tianjin, 300192, China
| | - Qian Li
- Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Tianjin Key Laboratory of Cancer Prevention and Therapy; Tianjin's Clinical Research Center for Cancer, Tianjin, 300192, China
| | - Ziyi Peng
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Mengqi Wang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Yangyang Xie
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Tiantian Li
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, 300070, China
| | - Yafei Wang
- Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Tianjin Key Laboratory of Cancer Prevention and Therapy; Tianjin's Clinical Research Center for Cancer, Tianjin, 300192, China
| | - Bill D Geng
- School of Natual Science, University of Texas at Austin, Austin, TX, 78712, USA
| | | | - P Leif Bergsagel
- Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA.
| | - Zhiqiang Liu
- The Proton Center of Shandong Cancer Institute and Hospital, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, 250117, China.
| |
Collapse
|
11
|
Frebel K, Albring JC, Wohlgemuth A, Schwöppe C, Hailfinger S, Lenz G, Stelljes M. Comparison of antibody-based immunotherapeutics for malignant hematological disease in an experimental murine model. Blood Adv 2024; 8:1934-1945. [PMID: 38197968 PMCID: PMC11021910 DOI: 10.1182/bloodadvances.2023011647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
ABSTRACT Antibody-based immunotherapies have revolutionized leukemia and lymphoma treatment, with animal studies being crucial in evaluating effectiveness and side effects. By targeting the evolutionary conserved Slamf7 immune receptor, which is naturally expressed by the murine multiple myeloma cell line MPC-11, we have developed a syngeneic mouse model for direct comparison of 3 immunotherapies: monoclonal antibodies (mAb), bispecific T-cell engagers (BiTE), and chimeric antigen receptor (CAR) T cells (CART), all targeting Slamf7. Slamf7-BiTE is a bispecific single-chain antibody consisting of α-Slamf7 and α-CD3 Fv fragments joined through a Gly-Ser linker, and Slamf7-CART comprises the α-Slamf7 Fv fragment fused to the msCD8α transmembrane and msCD28, 4-1BB, and CD3ζ intracellular signaling domains. Slamf7-BiTE and Slamf7-CART effectively killed MPC-11 cells in vitro, independently of Slamf7-mediated inhibitory signaling by self-ligation. After chimerizing the constant region of the rat-anti-mouse Slamf7 antibody to mouse Fc-immunoglobulin G2a for enhanced effector functions, Slamf7-mAb triggered antigen-specific antibody-dependent cellular cytotoxicity by binding to Fcγ receptor IV. In vivo, all 3 immunotherapies showed antitumor effects against Slamf7-expressing targets. Unlike Slamf7-mAb, Slamf7-BiTE led to considerable side effects in test animals, including weight loss and general malaise, which were also observed to a lesser extent after Slamf7-CART infusion. In allogeneic transplant, Slamf7-BiTE and Slamf7-CART maintained activity compared with the nontransplant setting, whereas Slamf7-mAb displayed enhanced antimyeloma activity. In summary, our model faithfully replicates treatment efficacy and side effects detected after human immunotherapy. It aids in developing and improving immunotherapies and may help devise novel approaches to mitigate undesired effects in steady state and allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Karin Frebel
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Jörn C. Albring
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Anika Wohlgemuth
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Christian Schwöppe
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Stephan Hailfinger
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Matthias Stelljes
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| |
Collapse
|
12
|
Minnie SA, Waltner OG, Zhang P, Takahashi S, Nemychenkov NS, Ensbey KS, Schmidt CR, Legg SRW, Comstock M, Boiko JR, Nelson E, Bhise SS, Wilkens AB, Koyama M, Dhodapkar MV, Chesi M, Riddell SR, Green DJ, Spencer A, Furlan SN, Hill GR. TIM-3 + CD8 T cells with a terminally exhausted phenotype retain functional capacity in hematological malignancies. Sci Immunol 2024; 9:eadg1094. [PMID: 38640253 PMCID: PMC11093588 DOI: 10.1126/sciimmunol.adg1094] [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: 12/02/2022] [Accepted: 03/27/2024] [Indexed: 04/21/2024]
Abstract
Chronic antigen stimulation is thought to generate dysfunctional CD8 T cells. Here, we identify a CD8 T cell subset in the bone marrow tumor microenvironment that, despite an apparent terminally exhausted phenotype (TPHEX), expressed granzymes, perforin, and IFN-γ. Concurrent gene expression and DNA accessibility revealed that genes encoding these functional proteins correlated with BATF expression and motif accessibility. IFN-γ+ TPHEX effectively killed myeloma with comparable efficacy to transitory effectors, and disease progression correlated with numerical deficits in IFN-γ+ TPHEX. We also observed IFN-γ+ TPHEX within CD19-targeted chimeric antigen receptor T cells, which killed CD19+ leukemia cells. An IFN-γ+ TPHEX gene signature was recapitulated in TEX cells from human cancers, including myeloma and lymphoma. Here, we characterize a TEX subset in hematological malignancies that paradoxically retains function and is distinct from dysfunctional TEX found in chronic viral infections. Thus, IFN-γ+ TPHEX represent a potential target for immunotherapy of blood cancers.
Collapse
Affiliation(s)
- Simone A. Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Olivia G. Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Ping Zhang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Shuichiro Takahashi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Nicole S. Nemychenkov
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Kathleen S. Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Christine R. Schmidt
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Samuel RW. Legg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Melissa Comstock
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Julie R. Boiko
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Department of Pediatrics, University of Washington; WA, UNITED STATES
| | - Ethan Nelson
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Shruti S. Bhise
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Alec B. Wilkens
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Madhav V. Dhodapkar
- Department of Hematology/Medical Oncology, Atlanta, GA, UNITED STATES
- Winship Cancer Institute, Emory University, Atlanta, GA, UNITED STATES
| | - Marta Chesi
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, UNITED STATES
| | - Stanley R. Riddell
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Division of Medical Oncology, University of Washington; Seattle, WA, UNITED STATES
| | - Damian J. Green
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Division of Medical Oncology, University of Washington; Seattle, WA, UNITED STATES
| | - Andrew Spencer
- Australian Center for Blood Diseases, Monash University/The Alfred Hospital, Melbourne, VIC, AUSTRALIA
- Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital, Melbourne, VIC, AUSTRALIA
- Department of Clinical Haematology, Monash University, Melbourne, VIC
| | - Scott N. Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Department of Pediatrics, University of Washington; WA, UNITED STATES
| | - Geoffrey R. Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Division of Medical Oncology, University of Washington; Seattle, WA, UNITED STATES
| |
Collapse
|
13
|
Takahashi S, Minnie SA, Ensbey KS, Schmidt CR, Sekiguchi T, Legg SRW, Zhang P, Koyama M, Olver SD, Collinge AD, Keshmiri S, Comstock ML, Varelias A, Green DJ, Hill GR. Regulatory T cells suppress myeloma-specific immunity during autologous stem cell mobilization and transplantation. Blood 2024; 143:1656-1669. [PMID: 38295333 PMCID: PMC11103090 DOI: 10.1182/blood.2023022000] [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: 07/27/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/02/2024] Open
Abstract
ABSTRACT Autologous stem cell transplantation (ASCT) is the standard of care consolidation therapy for eligible patients with myeloma but most patients eventually progress, an event associated with features of immune escape. Novel approaches to enhance antimyeloma immunity after ASCT represent a major unmet need. Here, we demonstrate that patient-mobilized stem cell grafts contain high numbers of effector CD8 T cells and immunosuppressive regulatory T cells (Tregs). We showed that bone marrow (BM)-residing T cells are efficiently mobilized during stem cell mobilization (SCM) and hypothesized that mobilized and highly suppressive BM-derived Tregs might limit antimyeloma immunity during SCM. Thus, we performed ASCT in a preclinical myeloma model with or without stringent Treg depletion during SCM. Treg depletion generated SCM grafts containing polyfunctional CD8 T effector memory cells, which dramatically enhanced myeloma control after ASCT. Thus, we explored clinically tractable translational approaches to mimic this scenario. Antibody-based approaches resulted in only partial Treg depletion and were inadequate to recapitulate this effect. In contrast, a synthetic interleukin-2 (IL-2)/IL-15 mimetic that stimulates the IL-2 receptor on CD8 T cells without binding to the high-affinity IL-2Ra used by Tregs efficiently expanded polyfunctional CD8 T cells in mobilized grafts and protected recipients from myeloma progression after ASCT. We confirmed that Treg depletion during stem cell mobilization can mitigate constraints on tumor immunity and result in profound myeloma control after ASCT. Direct and selective cytokine signaling of CD8 T cells can recapitulate this effect and represent a clinically testable strategy to improve responses after ASCT.
Collapse
Affiliation(s)
- Shuichiro Takahashi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Simone A. Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kathleen S. Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Christine R. Schmidt
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Tomoko Sekiguchi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Samuel R. W. Legg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ping Zhang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Stuart D. Olver
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | | | - Sara Keshmiri
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Melissa L. Comstock
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, St Lucia, QLD, Australia
| | - Damian J. Green
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Geoffrey R. Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
14
|
Mitsiades CS. Proteasome Inhibitors in Multiple Myeloma: Biological Insights on Mechanisms of Action or Resistance Informed by Functional Genomics. Hematol Oncol Clin North Am 2024; 38:321-336. [PMID: 38278626 DOI: 10.1016/j.hoc.2023.12.016] [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] [Indexed: 01/28/2024]
Abstract
During the last 20 years, proteasome inhibitors have been a cornerstone for the therapeutic management of multiple myeloma (MM). This review highlights how MM research has evolved over time in terms of our understanding of the mechanistic basis for the pronounced clinical activity of proteasome inhibitors in MM, compared with the limited clinical applications of this drug class outside the setting of plasma cell dyscrasias.
Collapse
Affiliation(s)
- Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA.
| |
Collapse
|
15
|
Du MT, Bergsagel PL, Chesi M. Immunocompetent Mouse Models of Multiple Myeloma: Therapeutic Implications. Hematol Oncol Clin North Am 2024; 38:533-546. [PMID: 38233233 PMCID: PMC10942746 DOI: 10.1016/j.hoc.2023.12.014] [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] [Indexed: 01/19/2024]
Abstract
Immunocompetent mouse models of multiple myeloma (MM) are particularly needed in the era of T cell redirected therapy to understand drivers of sensitivity and resistance, optimize responses, and prevent toxicities. Three mouse models have been extensively characterized: the Balb/c plasmacytomas, the 5TMM, and the Vk*MYC. In the last year, additional models have been generated, which, for the first time, capture primary MM initiating events, like MMSET/NSD2 or cyclin D1 dysregulation. However, the long latency needed for tumor development and the lack of transplantable lines limit their utilization. Future studies should focus on modeling hyperdiploid MM.
Collapse
Affiliation(s)
- Megan Tien Du
- Department of Medicine, Mayo Clinic, 13400 East Shea Boulevard, MCCRB 3-040, Scottsdale, AZ 85259, USA
| | - Peter Leif Bergsagel
- Department of Medicine, Mayo Clinic, 13400 East Shea Boulevard, MCCRB 3-040, Scottsdale, AZ 85259, USA
| | - Marta Chesi
- Department of Medicine, Mayo Clinic, 13400 East Shea Boulevard, MCCRB 3-040, Scottsdale, AZ 85259, USA.
| |
Collapse
|
16
|
Maclachlan KH, Gitareja K, Kang J, Cuddihy A, Cao Y, Hein N, Cullinane C, Ang CS, Brajanovski N, Pearson RB, Khot A, Sanij E, Hannan RD, Poortinga G, Harrison SJ. Targeting the ribosome to treat multiple myeloma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200771. [PMID: 38596309 PMCID: PMC10905045 DOI: 10.1016/j.omton.2024.200771] [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: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 04/11/2024]
Abstract
The high rates of protein synthesis and processing render multiple myeloma (MM) cells vulnerable to perturbations in protein homeostasis. The induction of proteotoxic stress by targeting protein degradation with proteasome inhibitors (PIs) has revolutionized the treatment of MM. However, resistance to PIs is inevitable and represents an ongoing clinical challenge. Our first-in-human study of the selective inhibitor of RNA polymerase I transcription of ribosomal RNA genes, CX-5461, has demonstrated a potential signal for anti-tumor activity in three of six heavily pre-treated MM patients. Here, we show that CX-5461 has potent anti-myeloma activity in PI-resistant MM preclinical models in vitro and in vivo. In addition to inhibiting ribosome biogenesis, CX-5461 causes topoisomerase II trapping and replication-dependent DNA damage, leading to G2/M cell-cycle arrest and apoptotic cell death. Combining CX-5461 with PI does not further enhance the anti-myeloma activity of CX-5461 in vivo. In contrast, CX-5461 shows synergistic interaction with the histone deacetylase inhibitor panobinostat in both the Vk∗MYC and the 5T33-KaLwRij mouse models of MM by targeting ribosome biogenesis and protein synthesis through distinct mechanisms. Our findings thus provide strong evidence to facilitate the clinical development of targeting the ribosome to treat relapsed and refractory MM.
Collapse
Affiliation(s)
- Kylee H. Maclachlan
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Clinical Hematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Kezia Gitareja
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine- St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Jian Kang
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine- St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Cuddihy
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Yuxi Cao
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Clinical Hematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Nadine Hein
- The ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Carleen Cullinane
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Ching-Seng Ang
- The Bio21 Institute of Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Natalie Brajanovski
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Richard B. Pearson
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Amit Khot
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Clinical Hematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Elaine Sanij
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine- St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Ross D. Hannan
- The ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Gretchen Poortinga
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Simon J. Harrison
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Clinical Hematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
17
|
Sasine JP, Kozlova NY, Valicente L, Dukov J, Tran DH, Himburg HA, Kumar S, Khorsandi S, Chan A, Grohe S, Li M, Kan J, Sehl ME, Schiller GJ, Reinhardt B, Singh BK, Ho R, Yue P, Pasquale EB, Chute JP. Inhibition of Ephrin B2 Reverse Signaling Abolishes Multiple Myeloma Pathogenesis. Cancer Res 2024; 84:919-934. [PMID: 38231476 PMCID: PMC10940855 DOI: 10.1158/0008-5472.can-23-1950] [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: 06/30/2023] [Revised: 11/14/2023] [Accepted: 01/11/2024] [Indexed: 01/18/2024]
Abstract
Bone marrow vascular endothelial cells (BM EC) regulate multiple myeloma pathogenesis. Identification of the mechanisms underlying this interaction could lead to the development of improved strategies for treating multiple myeloma. Here, we performed a transcriptomic analysis of human ECs with high capacity to promote multiple myeloma growth, revealing overexpression of the receptor tyrosine kinases, EPHB1 and EPHB4, in multiple myeloma-supportive ECs. Expression of ephrin B2 (EFNB2), the binding partner for EPHB1 and EPHB4, was significantly increased in multiple myeloma cells. Silencing EPHB1 or EPHB4 in ECs suppressed multiple myeloma growth in coculture. Similarly, loss of EFNB2 in multiple myeloma cells blocked multiple myeloma proliferation and survival in vitro, abrogated multiple myeloma engraftment in immune-deficient mice, and increased multiple myeloma sensitivity to chemotherapy. Administration of an EFNB2-targeted single-chain variable fragment also suppressed multiple myeloma growth in vivo. In contrast, overexpression of EFNB2 in multiple myeloma cells increased STAT5 activation, increased multiple myeloma cell survival and proliferation, and decreased multiple myeloma sensitivity to chemotherapy. Conversely, expression of mutant EFNB2 lacking reverse signaling capacity in multiple myeloma cells increased multiple myeloma cell death and sensitivity to chemotherapy and abolished multiple myeloma growth in vivo. Complementary analysis of multiple myeloma patient data revealed that increased EFNB2 expression is associated with adverse-risk disease and decreased survival. This study suggests that EFNB2 reverse signaling controls multiple myeloma pathogenesis and can be therapeutically targeted to improve multiple myeloma outcomes. SIGNIFICANCE Ephrin B2 reverse signaling mediated by endothelial cells directly regulates multiple myeloma progression and treatment resistance, which can be overcome through targeted inhibition of ephrin B2 to abolish myeloma.
Collapse
Affiliation(s)
- Joshua P. Sasine
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California
| | - Natalia Y. Kozlova
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Lisa Valicente
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Jennifer Dukov
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Dana H. Tran
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Heather A. Himburg
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sanjeev Kumar
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California
| | - Sarah Khorsandi
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Aldi Chan
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Samantha Grohe
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michelle Li
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California
| | - Jenny Kan
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California
| | - Mary E. Sehl
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California
| | - Gary J. Schiller
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California
| | - Bryanna Reinhardt
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Brijesh Kumar Singh
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, California
| | - Ritchie Ho
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, California
| | - Peibin Yue
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Elena B. Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, California
| | - John P. Chute
- Division of Hematology & Cellular Therapy, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California
| |
Collapse
|
18
|
Nedal TMV, Moen SH, Roseth IA, Tryggestad SS, Aass KR, Hov GG, Hella H, Sponaas AM, Standal T. Diet-induced obesity reduces bone marrow T and B cells and promotes tumor progression in a transplantable Vk*MYC model of multiple myeloma. Sci Rep 2024; 14:3643. [PMID: 38351079 PMCID: PMC10864380 DOI: 10.1038/s41598-024-54193-8] [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: 11/20/2023] [Accepted: 02/09/2024] [Indexed: 02/16/2024] Open
Abstract
Obesity is associated with an increased risk of developing multiple myeloma (MM). The molecular mechanisms causing this association is complex and incompletely understood. Whether obesity affects bone marrow immune cell composition in multiple myeloma is not characterized. Here, we examined the effect of diet-induced obesity on bone marrow immune cell composition and tumor growth in a Vk*MYC (Vk12653) transplant model of multiple myeloma. We find that diet-induced obesity promoted tumor growth in the bone marrow and spleen and reduced the relative number of T and B cells in the bone marrow. Our results suggest that obesity may reduce MM immune surveillance and thus may contribute to increased risk of developing MM.
Collapse
Affiliation(s)
- Tonje Marie Vikene Nedal
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Siv Helen Moen
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Research, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Ingrid Aass Roseth
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Synne Stokke Tryggestad
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kristin Roseth Aass
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Gunhild Garmo Hov
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Clinical Chemistry, St. Olavs Hospital, Trondheim, Norway
| | - Hanne Hella
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Anne-Marit Sponaas
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Therese Standal
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Hematology, St. Olavs Hospital, Trondheim, Norway.
| |
Collapse
|
19
|
Li M, Bennett MK, Toubia J, Pope VS, Tea MN, Tamang S, Samuel MS, Anderson PH, Gliddon BL, Powell JA, Pitson SM. An orthotopic syngeneic mouse model of bortezomib-resistant multiple myeloma. Br J Haematol 2024; 204:566-570. [PMID: 38053270 DOI: 10.1111/bjh.19240] [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: 08/17/2023] [Revised: 11/11/2023] [Accepted: 11/21/2023] [Indexed: 12/07/2023]
Abstract
While bortezomib has significant benefits in multiple myeloma (MM) therapy, the disease remains incurable due to the invariable development of bortezomib resistance. This emphasises the need for advanced models for preclinical evaluation of new therapeutic approaches for bortezomib-resistant MM. Here, we describe the development of an orthotopic syngeneic bortezomib-resistant MM mouse model based on the most well-characterised syngeneic MM mouse model derived from spontaneous MM-forming C57BL/KaLwRij mice. Using bortezomib-resistant 5TGM1 cells, we report and characterise a robust syngeneic mouse model of bortezomib-resistant MM that is well suited to the evaluation of new therapeutic approaches for proteasome inhibitor-resistant MM.
Collapse
Affiliation(s)
- Manjun Li
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Melissa K Bennett
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - John Toubia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Victoria S Pope
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Melinda N Tea
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Sarah Tamang
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Michael S Samuel
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Paul H Anderson
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Briony L Gliddon
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Jason A Powell
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
20
|
Welsh SJ, Barwick BG, Meermeier EW, Riggs DL, Shi CX, Zhu YX, Sharik ME, Du MT, Abrego Rocha LD, Garbitt VM, Stein CK, Petit JL, Meurice N, Tafoya Alvarado Y, Fonseca R, Todd KT, Brown S, Hammond ZJ, Cuc NH, Wittenberg C, Herzog C, Roschke AV, Demchenko YN, Chen WDD, Li P, Liao W, Leonard WJ, Lonial S, Bahlis NJ, Neri P, Boise LH, Chesi M, Bergsagel PL. Transcriptional Heterogeneity Overcomes Super-Enhancer Disrupting Drug Combinations in Multiple Myeloma. Blood Cancer Discov 2024; 5:34-55. [PMID: 37767768 PMCID: PMC10772542 DOI: 10.1158/2643-3230.bcd-23-0062] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/05/2023] [Accepted: 09/27/2023] [Indexed: 09/29/2023] Open
Abstract
Multiple myeloma (MM) is a malignancy that is often driven by MYC and that is sustained by IRF4, which are upregulated by super-enhancers. IKZF1 and IKZF3 bind to super-enhancers and can be degraded using immunomodulatory imide drugs (IMiD). Successful IMiD responses downregulate MYC and IRF4; however, this fails in IMiD-resistant cells. MYC and IRF4 downregulation can also be achieved in IMiD-resistant tumors using inhibitors of BET and EP300 transcriptional coactivator proteins; however, in vivo these drugs have a narrow therapeutic window. By combining IMiDs with EP300 inhibition, we demonstrate greater downregulation of MYC and IRF4, synergistic killing of myeloma in vitro and in vivo, and an increased therapeutic window. Interestingly, this potent combination failed where MYC and IRF4 expression was maintained by high levels of the AP-1 factor BATF. Our results identify an effective drug combination and a previously unrecognized mechanism of IMiD resistance. SIGNIFICANCE These results highlight the dependence of MM on IKZF1-bound super-enhancers, which can be effectively targeted by a potent therapeutic combination pairing IMiD-mediated degradation of IKZF1 and IKZF3 with EP300 inhibition. They also identify AP-1 factors as an unrecognized mechanism of IMiD resistance in MM. See related article by Neri, Barwick, et al., p. 56. See related commentary by Yun and Cleveland, p. 5. This article is featured in Selected Articles from This Issue, p. 4.
Collapse
Affiliation(s)
- Seth J. Welsh
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Erin W. Meermeier
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Daniel L. Riggs
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Chang-Xin Shi
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Yuan Xiao Zhu
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Meaghen E. Sharik
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Megan T. Du
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Leslie D. Abrego Rocha
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Victoria M. Garbitt
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Caleb K. Stein
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Joachim L. Petit
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Nathalie Meurice
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Yuliza Tafoya Alvarado
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Rodrigo Fonseca
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Kennedi T. Todd
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Sochilt Brown
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Zachery J. Hammond
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Nicklus H. Cuc
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Courtney Wittenberg
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Camille Herzog
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Anna V. Roschke
- Genetics Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | | | - Wei-dong D. Chen
- Genetics Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Peng Li
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - Wei Liao
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - Warren J. Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Nizar J. Bahlis
- Department of Medical Oncology and Hematology, Tom Baker Cancer Center, Calgary, Canada
- Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada
| | - Paola Neri
- Department of Medical Oncology and Hematology, Tom Baker Cancer Center, Calgary, Canada
- Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Marta Chesi
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - P. Leif Bergsagel
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| |
Collapse
|
21
|
Cheng Y, Sun F, Alapat DV, Wanchai V, Mery D, Guo W, Cao H, Zhu Y, Ashby C, Bauer MA, Nookaew I, Siegel ER, Ying J, Chen JR, Gai D, Peng B, Xu H, Bailey C, Al Hadidi S, Schinke C, Thanendrarajan S, Zangari M, Chesi M, Bergsagel PL, van Rhee F, Janz S, Tricot G, Shaughnessy JD, Zhan F. High NEK2 expression in myeloid progenitors suppresses T cell immunity in multiple myeloma. Cell Rep Med 2023; 4:101214. [PMID: 37794587 PMCID: PMC10591052 DOI: 10.1016/j.xcrm.2023.101214] [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: 04/17/2023] [Revised: 07/21/2023] [Accepted: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Multiple myeloma (MM) growth is supported by an immune-tolerant bone marrow microenvironment. Here, we find that loss of Never in mitosis gene A (NIMA)-related kinase 2 (NEK2) in tumor microenvironmental cells is associated with MM growth suppression. The absence of NEK2 leads to both fewer tumor-associated macrophages (TAMs) and inhibitory T cells. NEK2 expression in myeloid progenitor cells promotes the generation of functional TAMs when stimulated with MM conditional medium. Clinically, high NEK2 expression in MM cells is associated with increased CD8+ T effector memory cells, while low NEK2 is associated with an IFN-γ gene signature and activated T cell response. Inhibition of NEK2 upregulates PD-L1 expression in MM cells and myeloid cells. In a mouse model, the combination of NEK2 inhibitor INH154 with PD-L1 blockade effectively eliminates MM cells and prolongs survival. Our results provide strong evidence that NEK2 inhibition may overcome tumor immune escape and support its further clinical development.
Collapse
Affiliation(s)
- Yan Cheng
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Fumou Sun
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Daisy V Alapat
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Visanu Wanchai
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - David Mery
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Wancheng Guo
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Huojun Cao
- Iowa Institute for Oral Health Research, Division of Biostatistics and Computational Biology, Department of Endodontics, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Yuqi Zhu
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Cody Ashby
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Michael Anton Bauer
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Intawat Nookaew
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Eric R Siegel
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jun Ying
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jin-Ran Chen
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Dongzheng Gai
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Bailu Peng
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Hongwei Xu
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Clyde Bailey
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Samer Al Hadidi
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Carolina Schinke
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sharmilan Thanendrarajan
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Maurizio Zangari
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Marta Chesi
- Department of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - P Leif Bergsagel
- Department of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Frits van Rhee
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Siegfried Janz
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Guido Tricot
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - John D Shaughnessy
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Fenghuang Zhan
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
22
|
Oh DH, Ma X, Hogg SJ, He J, Kearney C, Brasacchio D, Susanto O, Maher B, Jennings IG, Newbold A, Fraser P, Gruber E, Kats LM, Gregory GP, Johnstone RW, Thompson PE, Shortt J. Rationally designed chimeric PI3K-BET bromodomain inhibitors elicit curative responses in MYC-driven lymphoma. Proc Natl Acad Sci U S A 2023; 120:e2306414120. [PMID: 37643213 PMCID: PMC10483632 DOI: 10.1073/pnas.2306414120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/01/2023] [Indexed: 08/31/2023] Open
Abstract
Targeted inhibitors of bromodomain and extraterminal (BET)-bromodomains and phosphatidylinositol-3-kinase (PI3K) signaling demonstrate potent but self-limited antilymphoma activity as single agents in the context of cellular Myelocytomatosis (cMYC) oncogene-dysregulation. However, combined PI3K and BET inhibition imparts synergistic anticancer activity with the potential for more sustained disease responses due to the mutual antagonism of compensatory epigenetic and signaling networks. Here, we describe the mechanistic and therapeutic validation of rationally designed dual PI3K/BET bromodomain inhibitors, built by linkage of established PI3K and BET inhibitor pharmacophores. The lead candidate demonstrates high selectivity, nanomolar range cellular potency, and compelling in vivo efficacy, including curative responses in the aggressive Eµ-Myc lymphoma model. These studies further support the therapeutic strategy of combined PI3K and BET inhibition and provide a potential step-change in approach to orthogonal MYC antagonism using optimized chimeric small-molecule technology.
Collapse
Affiliation(s)
- Danielle H. Oh
- Blood Cancer Therapeutics Laboratory, School of Clinical Sciences at Monash Health, Faculty of Medicine Nursing and Health Sciences, Monash University, MelbourneVIC3168, Australia
- Monash Haematology, Monash Health, MelbourneVIC3168, Australia
- Cancer Biology Therapeutics Program, Peter MacCallum Cancer Centre, MelbourneVIC3000, Australia
| | - Xiao Ma
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, ParkvilleVIC3052, Australia
- Department of Systems Biology, Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA02115
| | - Simon J. Hogg
- Cancer Biology Therapeutics Program, Peter MacCallum Cancer Centre, MelbourneVIC3000, Australia
- Oncology Discovery Research, Abbvie, South San Francisco, CA94080
| | - Jackson He
- Blood Cancer Therapeutics Laboratory, School of Clinical Sciences at Monash Health, Faculty of Medicine Nursing and Health Sciences, Monash University, MelbourneVIC3168, Australia
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, ParkvilleVIC3052, Australia
| | - Conor Kearney
- Olivia Newton-John Cancer Research Institute, HeidelbergVIC3084, Australia
- School of Cancer Medicine, La Trobe University, HeidelbergVIC3084, Australia
| | - Daniella Brasacchio
- Blood Cancer Therapeutics Laboratory, School of Clinical Sciences at Monash Health, Faculty of Medicine Nursing and Health Sciences, Monash University, MelbourneVIC3168, Australia
| | - Olivia Susanto
- Blood Cancer Therapeutics Laboratory, School of Clinical Sciences at Monash Health, Faculty of Medicine Nursing and Health Sciences, Monash University, MelbourneVIC3168, Australia
| | - Belinda Maher
- Blood Cancer Therapeutics Laboratory, School of Clinical Sciences at Monash Health, Faculty of Medicine Nursing and Health Sciences, Monash University, MelbourneVIC3168, Australia
| | - Ian G. Jennings
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, ParkvilleVIC3052, Australia
| | - Andrea Newbold
- Cancer Biology Therapeutics Program, Peter MacCallum Cancer Centre, MelbourneVIC3000, Australia
- Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, MelbourneVIC3000, Australia
| | - Peter Fraser
- Cancer Biology Therapeutics Program, Peter MacCallum Cancer Centre, MelbourneVIC3000, Australia
- Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, MelbourneVIC3000, Australia
| | - Emily Gruber
- Cancer Biology Therapeutics Program, Peter MacCallum Cancer Centre, MelbourneVIC3000, Australia
- Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, MelbourneVIC3000, Australia
| | - Lev M. Kats
- Cancer Biology Therapeutics Program, Peter MacCallum Cancer Centre, MelbourneVIC3000, Australia
- Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, MelbourneVIC3000, Australia
| | - Gareth P. Gregory
- Blood Cancer Therapeutics Laboratory, School of Clinical Sciences at Monash Health, Faculty of Medicine Nursing and Health Sciences, Monash University, MelbourneVIC3168, Australia
- Monash Haematology, Monash Health, MelbourneVIC3168, Australia
- Cancer Biology Therapeutics Program, Peter MacCallum Cancer Centre, MelbourneVIC3000, Australia
| | - Ricky W. Johnstone
- Cancer Biology Therapeutics Program, Peter MacCallum Cancer Centre, MelbourneVIC3000, Australia
- Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, MelbourneVIC3000, Australia
| | - Philip E. Thompson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, ParkvilleVIC3052, Australia
| | - Jake Shortt
- Blood Cancer Therapeutics Laboratory, School of Clinical Sciences at Monash Health, Faculty of Medicine Nursing and Health Sciences, Monash University, MelbourneVIC3168, Australia
- Monash Haematology, Monash Health, MelbourneVIC3168, Australia
- Cancer Biology Therapeutics Program, Peter MacCallum Cancer Centre, MelbourneVIC3000, Australia
- Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, MelbourneVIC3000, Australia
| |
Collapse
|
23
|
Ishibashi M, Takahashi M, Yamaya T, Imai Y. Current and Future PET Imaging for Multiple Myeloma. Life (Basel) 2023; 13:1701. [PMID: 37629558 PMCID: PMC10455506 DOI: 10.3390/life13081701] [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: 06/07/2023] [Revised: 07/26/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Positron emission tomography (PET) is an imaging modality used for the noninvasive assessment of tumor staging and response to therapy. PET with 18F labeled fluorodeoxyglucose (18F-FDG PET) is widely used to assess the active and inactive lesions in patients with multiple myeloma (MM). Despite the availability of 18F-FDG PET for the management of MM, PET imaging is less sensitive than next-generation flow cytometry and sequencing. Therefore, the novel PET radiotracers 64Cu-LLP2A, 68Ga-pentixafor, and 89Zr-daratumumab have been developed to target the cell surface antigens of MM cells. Furthermore, recent studies attempted to visualize the tumor-infiltrating lymphocytes using PET imaging in patients with cancer to investigate their prognostic effect; however, these studies have not yet been performed in MM patients. This review summarizes the recent studies on PET with 18F-FDG and novel radiotracers for the detection of MM and the resulting preclinical research using MM mouse models and clinical studies. Novel PET technologies may be useful for developing therapeutic strategies for MM in the future.
Collapse
Affiliation(s)
- Mariko Ishibashi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan;
| | - Miwako Takahashi
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (M.T.); (T.Y.)
| | - Taiga Yamaya
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (M.T.); (T.Y.)
| | - Yoichi Imai
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi 321-0293, Japan
| |
Collapse
|
24
|
Maura F, Coffey DG, Stein CK, Braggio E, Ziccheddu B, Sharik ME, Du M, Alvarado YT, Shi CX, Zhu YX, Meermeier EW, Morgan GJ, Landgren O, Leif Bergsagel P, Chesi M. The Vk*MYC Mouse Model recapitulates human multiple myeloma evolution and genomic diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550482. [PMID: 37546905 PMCID: PMC10402028 DOI: 10.1101/2023.07.25.550482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Despite advancements in profiling multiple myeloma (MM) and its precursor conditions, there is limited information on mechanisms underlying disease progression. Clincal efforts designed to deconvolute such mechanisms are challenged by the long lead time between monoclonal gammopathy and its transformation to MM. MM mouse models represent an opportunity to overcome this temporal limitation. Here, we profile the genomic landscape of 118 genetically engineered Vk*MYC MM and reveal that it recapitulates the genomic heterogenenity and life history of human MM. We observed recurrent copy number alterations, structural variations, chromothripsis, driver mutations, APOBEC mutational activity, and a progressive decrease in immunoglobulin transcription that inversely correlates with proliferation. Moreover, we identified frequent insertional mutagenesis by endogenous retro-elements as a murine specific mechanism to activate NF-kB and IL6 signaling pathways shared with human MM. Despite the increased genomic complexity associated with progression, advanced tumors remain dependent on MYC expression, that drives the progression of monoclonal gammopathy to MM.
Collapse
|
25
|
Pichler AC, Carrié N, Cuisinier M, Ghazali S, Voisin A, Axisa PP, Tosolini M, Mazzotti C, Golec DP, Maheo S, do Souto L, Ekren R, Blanquart E, Lemaitre L, Feliu V, Joubert MV, Cannons JL, Guillerey C, Avet-Loiseau H, Watts TH, Salomon BL, Joffre O, Grinberg-Bleyer Y, Schwartzberg PL, Lucca LE, Martinet L. TCR-independent CD137 (4-1BB) signaling promotes CD8 +-exhausted T cell proliferation and terminal differentiation. Immunity 2023; 56:1631-1648.e10. [PMID: 37392737 PMCID: PMC10649891 DOI: 10.1016/j.immuni.2023.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 03/29/2023] [Accepted: 06/08/2023] [Indexed: 07/03/2023]
Abstract
CD137 (4-1BB)-activating receptor represents a promising cancer immunotherapeutic target. Yet, the cellular program driven by CD137 and its role in cancer immune surveillance remain unresolved. Using T cell-specific deletion and agonist antibodies, we found that CD137 modulates tumor infiltration of CD8+-exhausted T (Tex) cells expressing PD1, Lag-3, and Tim-3 inhibitory receptors. T cell-intrinsic, TCR-independent CD137 signaling stimulated the proliferation and the terminal differentiation of Tex precursor cells through a mechanism involving the RelA and cRel canonical NF-κB subunits and Tox-dependent chromatin remodeling. While Tex cell accumulation induced by prophylactic CD137 agonists favored tumor growth, anti-PD1 efficacy was improved with subsequent CD137 stimulation in pre-clinical mouse models. Better understanding of T cell exhaustion has crucial implications for the treatment of cancer and infectious diseases. Our results identify CD137 as a critical regulator of Tex cell expansion and differentiation that holds potential for broad therapeutic applications.
Collapse
Affiliation(s)
- Andrea C Pichler
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Cell Signaling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nadège Carrié
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Marine Cuisinier
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Institut Universitaire du Cancer, CHU Toulouse, Toulouse, France
| | - Samira Ghazali
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), UPS, INSERM, CNRS, Toulouse, France
| | - Allison Voisin
- Centre de Recherche en Cancérologie de Lyon, Labex DEVweCAN, INSERM, CNRS, Université Claude Bernard Lyon 1, Centre Léon Bérard, Lyon, France
| | - Pierre-Paul Axisa
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Marie Tosolini
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Céline Mazzotti
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Institut Universitaire du Cancer, CHU Toulouse, Toulouse, France
| | - Dominic P Golec
- Cell Signaling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sabrina Maheo
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Institut Universitaire du Cancer, CHU Toulouse, Toulouse, France
| | - Laura do Souto
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Institut Universitaire du Cancer, CHU Toulouse, Toulouse, France
| | - Rüçhan Ekren
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Eve Blanquart
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Lea Lemaitre
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Virginie Feliu
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Marie-Véronique Joubert
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Institut Universitaire du Cancer, CHU Toulouse, Toulouse, France
| | - Jennifer L Cannons
- Cell Signaling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Camille Guillerey
- Cancer Immunotherapies Group, The University of Queensland, Brisbane, QLD, Australia
| | - Hervé Avet-Loiseau
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Institut Universitaire du Cancer, CHU Toulouse, Toulouse, France
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Benoit L Salomon
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), UPS, INSERM, CNRS, Toulouse, France; Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Olivier Joffre
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), UPS, INSERM, CNRS, Toulouse, France
| | - Yenkel Grinberg-Bleyer
- Centre de Recherche en Cancérologie de Lyon, Labex DEVweCAN, INSERM, CNRS, Université Claude Bernard Lyon 1, Centre Léon Bérard, Lyon, France
| | - Pamela L Schwartzberg
- Cell Signaling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Liliana E Lucca
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France.
| | - Ludovic Martinet
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Institut Universitaire du Cancer, CHU Toulouse, Toulouse, France.
| |
Collapse
|
26
|
Flietner E, Yu M, Poudel G, Veltri AJ, Zhou Y, Rajagopalan A, Feng Y, Lasho T, Wen Z, Sun Y, Patnaik MM, Callander NS, Asimakopoulos F, Wang D, Zhang J. Molecular characterization stratifies VQ myeloma cells into two clusters with distinct risk signatures and drug responses. Oncogene 2023; 42:1751-1762. [PMID: 37031341 PMCID: PMC10367583 DOI: 10.1038/s41388-023-02684-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/10/2023]
Abstract
Multiple myeloma (MM) is a cancer of malignant plasma cells in the bone marrow and extramedullary sites. We previously characterized a VQ model for human high-risk MM. The various VQ lines display different disease phenotypes and survival rates, suggesting significant intra-model variation. Here, we use whole-exome sequencing and copy number variation (CNV) analysis coupled with RNA-Seq to stratify the VQ lines into corresponding clusters: Group A cells had monosomy chromosome (chr) 5 and overexpressed genes and pathways associated with sensitivity to bortezomib (Btz) treatment in human MM patients. By contrast, Group B VQ cells carried recurrent amplification (Amp) of chr3 and displayed high-risk MM features, including downregulation of Fam46c, upregulation of cancer growth pathways associated with functional high-risk MM, and expression of Amp1q and high-risk UAMS-70 and EMC-92 gene signatures. Consistently, in sharp contrast to Group A VQ cells that showed short-term response to Btz, Group B VQ cells were de novo resistant to Btz in vivo. Our study highlights Group B VQ lines as highly representative of the human MM subset with ultrahigh risk.
Collapse
Affiliation(s)
- Evan Flietner
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Mei Yu
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
| | - Govinda Poudel
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | | | - Yun Zhou
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Adhithi Rajagopalan
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Yubin Feng
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Terra Lasho
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55902, USA
| | - Zhi Wen
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI, 54449, USA
| | - Yuqian Sun
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55902, USA
| | - Natalie S Callander
- Division of Hematology/Oncology, Department of Medicine, UW Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Fotis Asimakopoulos
- Division of Blood and Marrow Transplantation, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Demin Wang
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA.
| | - Jing Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
27
|
Paterek A, Oknińska M, Pilch Z, Sosnowska A, Ramji K, Mackiewicz U, Golab J, Nowis D, Mączewski M. Arginase Inhibition Mitigates Bortezomib-Exacerbated Cardiotoxicity in Multiple Myeloma. Cancers (Basel) 2023; 15:cancers15072191. [PMID: 37046852 PMCID: PMC10093116 DOI: 10.3390/cancers15072191] [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: 03/01/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is associated with increased cardiovascular morbidity and mortality, while MM therapies also result in adverse cardiac effects. Endothelial dysfunction and impaired nitric oxide (NO) pathway is their possible mediator. OBJECTIVE Since MM is associated with increased arginase expression, resulting in the consumption of ʟ-arginine, precursor for NO synthesis, our aim was to test if cardiotoxicity mediated by MM and MM therapeutic, bortezomib (a proteasome inhibitor), can be ameliorated by an arginase inhibitor through improved endothelial function. METHODS We used a mouse Vĸ*MYC model of non-light chain MM. Cardiac function was assessed by echocardiography. RESULTS MM resulted in progressive left ventricular (LV) systolic dysfunction, and bortezomib exacerbated this effect, leading to significant impairment of LV performance. An arginase inhibitor, OAT-1746, protected the heart against bortezomib- or MM-induced toxicity but did not completely prevent the effects of the MM+bortezomib combination. MM was associated with improved endothelial function (assessed as NO production) vs. healthy controls, while bortezomib did not affect it. OAT-1746 improved endothelial function only in healthy mice. NO plasma concentration was increased by OAT-1746 but was not affected by MM or bortezomib. CONCLUSIONS Bortezomib exacerbates MM-mediated LV systolic dysfunction in a mouse model of MM, while an arginase inhibitor partially prevents it. Endothelium does not mediate either these adverse or beneficial effects. This suggests that proteasome inhibitors should be used with caution in patients with advanced myeloma, where the summation of cardiotoxicity could be expected. Therapies aimed at the NO pathway, in particular arginase inhibitors, could offer promise in the prevention/treatment of cardiotoxicity in MM.
Collapse
Affiliation(s)
- Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Zofia Pilch
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Anna Sosnowska
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Kavita Ramji
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
- Centre of Preclinical Research, Medical University of Warsaw, 1B Banacha Street, 02-097 Warsaw, Poland
| | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
- Laboratory of Experimental Medicine, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| |
Collapse
|
28
|
Marques-Mourlet C, Di Iorio R, Fairfield H, Reagan MR. Obesity and myeloma: Clinical and mechanistic contributions to disease progression. Front Endocrinol (Lausanne) 2023; 14:1118691. [PMID: 36909335 PMCID: PMC9996186 DOI: 10.3389/fendo.2023.1118691] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Obesity and obesogenic behaviors are positively associated with both monoclonal gammopathy of unknown significance (MGUS) and multiple myeloma (MM). As the only known modifiable risk factor, this association has emerged as a new potential target for MM prevention, but little is known about the mechanistic relationship of body weight with MM progression. Here we summarize epidemiological correlations between weight, body composition, and the various stages of myeloma disease progression and treatments, as well as the current understanding of the molecular contributions of obesity-induced changes in myeloma cell phenotype and signaling. Finally, we outline groundwork for the future characterization of the relationship between body weight patterns, the bone marrow microenvironment, and MM pathogenesis in animal models, which have the potential to impact our understanding of disease pathogenesis and inform MM prevention messages.
Collapse
Affiliation(s)
- Constance Marques-Mourlet
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of Strasbourg, Pharmacology Department, Strasbourg, France
| | - Reagan Di Iorio
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of New England, College of Osteopathic Medicine, Biddeford, ME, United States
| | - Heather Fairfield
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME, United States
- Tufts University, School of Medicine, Boston, MA, United States
| | - Michaela R. Reagan
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME, United States
- Tufts University, School of Medicine, Boston, MA, United States
| |
Collapse
|
29
|
Piskor EM, Winkler R, Kosan C. Analyzing Lymphoma Development and Progression Using HDACi in Mouse Models. Methods Mol Biol 2023; 2589:3-15. [PMID: 36255614 DOI: 10.1007/978-1-0716-2788-4_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Besides the physiological role of histone deacetalylases in maintaining normal cellular integrity, the acetylation landscape is changed in cancer cells, which has been implicated as a potential target in cancer therapy. The overexpression of certain HDACs correlates with specific cancer types. Therefore, the development of specific HDAC inhibitors may extend the therapeutic strategy for cancer therapy. Here, we describe how to investigate the therapeutic potential of specific HDACi by treatment in a mouse model for B-cell lymphoma, exemplified by the HDAC6 inhibitor Marbostat-100.
Collapse
Affiliation(s)
- Eva-Maria Piskor
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
| | - René Winkler
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Christian Kosan
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
30
|
Targeting arginase-1 exerts antitumor effects in multiple myeloma and mitigates bortezomib-induced cardiotoxicity. Sci Rep 2022; 12:19660. [PMID: 36385153 PMCID: PMC9668840 DOI: 10.1038/s41598-022-24137-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Multiple myeloma (MM) remains an incurable malignancy of plasma cells despite constantly evolving therapeutic approaches including various types of immunotherapy. Increased arginase activity has been associated with potent suppression of T-cell immune responses in different types of cancer. Here, we investigated the role of arginase 1 (ARG1) in Vκ*MYC model of MM in mice. ARG1 expression in myeloid cells correlated with tumor progression and was accompanied by a systemic drop in ʟ-arginine levels. In MM-bearing mice antigen-induced proliferation of adoptively transferred T-cells was strongly suppressed and T-cell proliferation was restored by pharmacological arginase inhibition. Progression of Vκ*MYC tumors was significantly delayed in mice with myeloid-specific ARG1 deletion. Arginase inhibition effectively inhibited tumor progression although it failed to augment anti-myeloma effects of bortezomib. However, arginase inhibitor completely prevented development of bortezomib-induced cardiotoxicity in mice. Altogether, these findings indicate that arginase inhibitors could be further tested as a complementary strategy in multiple myeloma to mitigate adverse cardiac events without compromising antitumor efficacy of proteasome inhibitors.
Collapse
|
31
|
Markouli M, Strepkos D, Piperi C. Impact of Histone Modifications and Their Therapeutic Targeting in Hematological Malignancies. Int J Mol Sci 2022; 23:13657. [PMID: 36362442 PMCID: PMC9654260 DOI: 10.3390/ijms232113657] [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: 09/30/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Hematologic malignancies are a large and heterogeneous group of neoplasms characterized by complex pathogenetic mechanisms. The abnormal regulation of epigenetic mechanisms and specifically, histone modifications, has been demonstrated to play a central role in hematological cancer pathogenesis and progression. A variety of epigenetic enzymes that affect the state of histones have been detected as deregulated, being either over- or underexpressed, which induces changes in chromatin compaction and, subsequently, affects gene expression. Recent advances in the field of epigenetics have revealed novel therapeutic targets, with many epigenetic drugs being investigated in clinical trials. The present review focuses on the biological impact of histone modifications in the pathogenesis of hematologic malignancies, describing a wide range of therapeutic agents that have been discovered to target these alterations and are currently under investigation in clinical trials.
Collapse
Affiliation(s)
| | | | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.M.); (D.S.)
| |
Collapse
|
32
|
Minnie SA, Waltner OG, Ensbey KS, Nemychenkov NS, Schmidt CR, Bhise SS, Legg SRW, Campoy G, Samson LD, Kuns RD, Zhou T, Huck JD, Vuckovic S, Zamora D, Yeh A, Spencer A, Koyama M, Markey KA, Lane SW, Boeckh M, Ring AM, Furlan SN, Hill GR. Depletion of exhausted alloreactive T cells enables targeting of stem-like memory T cells to generate tumor-specific immunity. Sci Immunol 2022; 7:eabo3420. [PMID: 36240285 PMCID: PMC10184646 DOI: 10.1126/sciimmunol.abo3420] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Some hematological malignancies such as multiple myeloma are inherently resistant to immune-mediated antitumor responses, the cause of which remains unknown. Allogeneic bone marrow transplantation (alloBMT) is the only curative immunotherapy for hematological malignancies due to profound graft-versus-tumor (GVT) effects, but relapse remains the major cause of death. We developed murine models of alloBMT where the hematological malignancy is either sensitive [acute myeloid leukemia (AML)] or resistant (myeloma) to GVT effects. We found that CD8+ T cell exhaustion in bone marrow was primarily alloantigen-driven, with expression of inhibitory ligands present on myeloma but not AML. Because of this tumor-independent exhaustion signature, immune checkpoint inhibition (ICI) in myeloma exacerbated graft-versus-host disease (GVHD) without promoting GVT effects. Administration of post-transplant cyclophosphamide (PT-Cy) depleted donor T cells with an exhausted phenotype and spared T cells displaying a stem-like memory phenotype with chromatin accessibility present in cytokine signaling genes, including the interleukin-18 (IL-18) receptor. Whereas ICI with anti-PD-1 or anti-TIM-3 remained ineffective after PT-Cy, administration of a decoy-resistant IL-18 (DR-18) strongly enhanced GVT effects in both myeloma and leukemia models, without exacerbation of GVHD. We thus defined mechanisms of resistance to T cell-mediated antitumor effects after alloBMT and described an immunotherapy approach targeting stem-like memory T cells to enhance antitumor immunity.
Collapse
Affiliation(s)
- Simone A. Minnie
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Olivia G. Waltner
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Kathleen S. Ensbey
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Nicole S. Nemychenkov
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Christine R. Schmidt
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Shruti S. Bhise
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Samuel RW. Legg
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Gabriela Campoy
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Luke D. Samson
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Rachel D. Kuns
- QIMR Berghofer Medical Research Institute; Brisbane, QLD, 4006, AUSTRALIA
| | - Ting Zhou
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, 06519, UNITED STATES
| | - John D. Huck
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, 06519, UNITED STATES
| | - Slavica Vuckovic
- QIMR Berghofer Medical Research Institute; Brisbane, QLD, 4006, AUSTRALIA
| | - Danniel Zamora
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Albert Yeh
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
| | - Andrew Spencer
- Australian Center for Blood Diseases, Monash University/The Alfred Hospital; Melbourne, VIC, 3004, AUSTRALIA
- Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital; Melbourne, VIC, 3004, AUSTRALIA
- Department of Clinical Haematology, Monash University; Melbourne, VIC, 3800, AUSTRALIA
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Kate A. Markey
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
| | - Steven W. Lane
- QIMR Berghofer Medical Research Institute; Brisbane, QLD, 4006, AUSTRALIA
| | - Michael Boeckh
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Aaron M. Ring
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, 06519, UNITED STATES
| | - Scott N. Furlan
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Pediatrics, University of Washington; WA, 98105, UNITED STATES
| | - Geoffrey R. Hill
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
| |
Collapse
|
33
|
Hypoxia induces chemoresistance to proteasome inhibitors through orchestrating deSUMOylation and ubiquitination of SRC-3 in multiple myeloma. Oncogene 2022; 41:4971-4979. [PMID: 36209257 DOI: 10.1038/s41388-022-02494-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022]
Abstract
The bone marrow microenvironment in multiple myeloma (MM) is hypoxic and provides multi-advantages for the initiation of chemoresistance, but the underlying mechanisms and key regulators are still indistinct. In the current study, we found that hypoxia stimulus easily induced chemoresistance to proteasome inhibitors (PIs), and the steroid receptor coactivator 3 (SRC-3) expression was remarkably augmented at posttranslational level. Protein interactome analysis identified SENP1 as a key modifier of SRC-3 stability, as SENP1-mediated deSUMOylation attenuated the K11-linked polyubiquitination of SRC-3. SENP1 depletion in the SENP1fl/flCD19Cre/+ B cells showed impaired SRC3 stability, and knockdown of SENP1 in MM cells by CRISPR/cas9 sgRNA accelerated the degradation of SRC-3 and remarkably overcame the resistance to PIs. In the Vk*Myc and 5TGM1 mouse models as well as patient-derived xenograft (PDX) of myeloma, SENP1 inhibitor Momordin Ιc (Mc) increased the sensitivity to PIs in MM cells. Importantly, SENP1 level was positively correlated with SRC-3 level in the tissues from refractory/relapsed MM, as well as in xenograft tissues from mice treated with bortezomib and Mc. Taken together, our findings suggest that hypoxia-induced SENP1 is a crucial regulator of chemoresistance to PIs, and shed light on developing therapeutic strategies to overcome chemoresistance by using small molecules targeting SENP1 or SRC-3.
Collapse
|
34
|
Yamamoto J, Ito T, Yamaguchi Y, Handa H. Discovery of CRBN as a target of thalidomide: a breakthrough for progress in the development of protein degraders. Chem Soc Rev 2022; 51:6234-6250. [PMID: 35796627 DOI: 10.1039/d2cs00116k] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Progress in strategies aimed at breaking down therapeutic target proteins has led to a paradigm shift in drug discovery. Thalidomide and its derivatives are the only protein degraders currently used in clinical practice. Our understanding of the molecular mechanism of action of thalidomide and its derivatives has advanced dramatically since the identification of cereblon (CRBN) as their direct target. The binding of thalidomide derivatives to CRBN, a substrate recognition receptor for Cullin 4 RING E3 ubiquitin ligase (CRL4), induces the recruitment of non-native substrates to CRL4CRBN and their subsequent degradation. This discovery was a breakthrough in the current rapid development of protein-degrading agents because clarification of the mechanism of action of thalidomide derivatives has demonstrated the clinical value of these compounds. This review provides an overview of the mechanism of action of thalidomide and its derivatives and describes perspectives for protein degraders.
Collapse
Affiliation(s)
- Junichi Yamamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Takumi Ito
- Institute of Medical Science, Tokyo Medical University, Shinjuku, Tokyo 160-8402, Japan
| | - Yuki Yamaguchi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Hiroshi Handa
- Center for Future Medical Research, Tokyo Medical University, Shinjuku, Tokyo 160-8402, Japan.
| |
Collapse
|
35
|
Flietner E, Wen Z, Rajagopalan A, Jung O, Watkins L, Wiesner J, You X, Zhou Y, Sun Y, Kingstad-Bakke B, Callander NS, Rapraeger A, Suresh M, Asimakopoulos F, Zhang J. Ponatinib sensitizes myeloma cells to MEK inhibition in the high-risk VQ model. Sci Rep 2022; 12:10616. [PMID: 35739276 PMCID: PMC9226136 DOI: 10.1038/s41598-022-14114-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/01/2022] [Indexed: 11/08/2022] Open
Abstract
Multiple myeloma (MM) is a malignant plasma cell cancer. Mutations in RAS pathway genes are prevalent in advanced and proteasome inhibitor (PI) refractory MM. As such, we recently developed a VQ MM mouse model recapitulating human advanced/high-risk MM. Using VQ MM cell lines we conducted a repurposing screen of 147 FDA-approved anti-cancer drugs with or without trametinib (Tra), a MEK inhibitor. Consistent with its high-risk molecular feature, VQ MM displayed reduced responses to PIs and de novo resistance to the BCL2 inhibitor, venetoclax. Ponatinib (Pon) is the only tyrosine kinase inhibitor that showed moderate MM killing activity as a single agent and strong synergism with Tra in vitro. Combined Tra and Pon treatment significantly prolonged the survival of VQ MM mice regardless of treatment schemes. However, this survival benefit was moderate compared to that of Tra alone. Further testing of Tra and Pon on cytotoxic CD8+ T cells showed that Pon, but not Tra, blocked T cell function in vitro, suggesting that the negative impact of Pon on T cells may partially counteract its MM-killing synergism with Tra in vivo. Our study provides strong rational to comprehensively evaluate agents on both MM cells and anti-MM immune cells during therapy development.
Collapse
Affiliation(s)
- Evan Flietner
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Room 7453, WIMR II, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Zhi Wen
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Room 7453, WIMR II, 1111 Highland Avenue, Madison, WI, 53705, USA
- Center for Precision Medicine Research and Integrated Research and Development Laboratories, Marshfield Clinic Research Institute, Marshfield, WI, 54449, USA
| | - Adhithi Rajagopalan
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Room 7453, WIMR II, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Oisun Jung
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Lyndsay Watkins
- Center for Precision Medicine Research and Integrated Research and Development Laboratories, Marshfield Clinic Research Institute, Marshfield, WI, 54449, USA
| | - Joshua Wiesner
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Xiaona You
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Room 7453, WIMR II, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Yun Zhou
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Room 7453, WIMR II, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Yuqian Sun
- Department of Biology, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Natalie S Callander
- Division of Hematology/Oncology, Department of Medicine, UW Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Alan Rapraeger
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - M Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Fotis Asimakopoulos
- Division of Hematology/Oncology, Department of Medicine, UW Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California-San Diego, La Jolla, CA, 92093, USA
| | - Jing Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Room 7453, WIMR II, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
36
|
Ocio EM, Nadeem O, Schjesvold F, Gay F, Touzeau C, Dimopoulos MA, Richardson PG, Mateos MV. Melflufen for the treatment of multiple myeloma. Expert Rev Clin Pharmacol 2022; 15:371-382. [PMID: 35723075 DOI: 10.1080/17512433.2022.2075847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Melphalan flufenamide (melflufen) is a first-in-class peptide-drug conjugate that takes advantage of increased aminopeptidase activity inside tumor cells to rapidly release alkylating agents therein. Melflufen in combination with dexamethasone has been evaluated in multiple clinical trials in patients with relapsed/refractory multiple myeloma (MM). AREAS COVERED This profile covers the unique mechanism of action of melflufen, the preclinical results supporting its activity in cellular models of resistance to chemotherapy, its activity in animal models of MM, and the clinical pharmacokinetics of melflufen. Findings from clinical trials evaluating melflufen, including the pivotal phase II HORIZON study and the phase III OCEAN study, are discussed. EXPERT OPINION Although MM treatment has improved, patients with disease refractory to multiple standard-of-care drug classes face a dismal prognosis. Melflufen demonstrated efficacy and tolerability in select populations, with an initial approval in the United States in patients with ≥ four previous lines of therapy and triple-class-refractory MM. Results from the phase III OCEAN study - currently under discussion with regulatory agencies in the United States and Europe - are more complex and have been put into context herein. Lastly, melflufen provides a proof-of-concept for the utility of the peptide-drug conjugate platform in relapsed/refractory MM.
Collapse
Affiliation(s)
- Enrique M Ocio
- Department of Hematology, University Hospital Marqués de Valdecilla (IDIVAL), University of CantabriaUniversity Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, Santander, Spain
| | - Omar Nadeem
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fredrik Schjesvold
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Cyrille Touzeau
- Hematology Department, Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université d'Angers, Université de Nantes, Nantes, France; Site de Recherche Intégrée sur le Cancer (SIRIC), Imaging and Longitudinal Investigations to Ameliorate Decision-making (ILIAD), Nantes, France; Service d'hématologie Clinique, Centre Hospitalier Universitaire, Place Alexis Ricordeau, Nantes, France
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Paul G Richardson
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria-Victoria Mateos
- Department of Haematology, Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cancer (IBMCC-USAL, CSIC), Salamanca, Spain
| |
Collapse
|
37
|
Villa NY, Rahman MM, Mamola J, Sharik ME, de Matos AL, Kilbourne J, Lowe K, Daggett-Vondras J, D'Isabella J, Goras E, Chesi M, Bergsagel PL, McFadden G. Transplantation of autologous bone marrow pre-loaded ex vivo with oncolytic myxoma virus is efficacious against drug-resistant Vk*MYC mouse myeloma. Oncotarget 2022; 13:490-504. [PMID: 35251496 PMCID: PMC8893797 DOI: 10.18632/oncotarget.28205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/14/2022] [Indexed: 11/25/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of plasma cells that remains incurable despite significant progress with myeloablative regimens and autologous stem cell transplantation for eligible patients and, more recently with T cell redirected immunotherapy. Recently, we reported that ex vivo virotherapy with oncolytic myxoma virus (MYXV) improved MM-free survival in an autologous-transplant Balb/c mouse model. Here, we tested the Vk*MYC transplantable C57BL/6 mouse MM model that more closely recapitulates human disease. In vitro, the murine bortezomib-resistant Vk12598 cell line is fully susceptible to MYXV infection. In vivo results demonstrate: (i) autologous bone marrow (BM) leukocytes armed ex vivo with MYXV exhibit moderate therapeutic effects against MM cells pre-seeded into recipient mice; (ii) Cyclophosphamide in combination with BM/MYXV delays the onset of myeloma in mice seeded with Vk12598 cells; (iii) BM/MYXV synergizes with the Smac-mimetics LCL161 and with immune checkpoint inhibitor α-PD-1 to control the progression of established MM in vivo, resulting in significant improvement of survival rates and decreased of tumor burden; (iv) Survivor mice from (ii) and (iii), when re-challenged with fresh Vk12598 cells, developed acquired anti-MM immunity. These results highlight the utility of autologous BM grafts armed ex vivo with oncolytic MYXV alone or in combination with chemotherapy/immunotherapy to treat drug-resistant MM in vivo.
Collapse
Affiliation(s)
- Nancy Y. Villa
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
- Division of Hematology/Oncology, School of Medicine, Emory University, Atlanta, GA 32322, USA
| | - Masmudur M. Rahman
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Joseph Mamola
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | | | - Ana Lemos de Matos
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Jacquelyn Kilbourne
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Kenneth Lowe
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Juliane Daggett-Vondras
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Julia D'Isabella
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Elizabeth Goras
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| | - Marta Chesi
- Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | | | - Grant McFadden
- Biodesign Institute, Center for Immunotherapy, Vaccines and Virotherapy (CIVV), Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
38
|
SAR442085, a novel anti-CD38 antibody with enhanced antitumor activity against multiple myeloma. Blood 2022; 139:1160-1176. [PMID: 35201323 DOI: 10.1182/blood.2021012448] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/23/2021] [Indexed: 12/28/2022] Open
Abstract
Anti-CD38 monoclonal antibodies (mAbs) represent a breakthrough in the treatment of multiple myeloma (MM), yet some patients fail to respond or progress quickly with this therapy, highlighting the need for novel approaches. In this study we compared the preclinical efficacy of SAR442085, a next-generation anti-CD38 mAb with enhanced affinity for activating Fcγ receptors (FcγR), with first-generation anti-CD38 mAb daratumumab and isatuximab. In surface plasmon resonance and cellular binding assays, we found that SAR442085 had higher binding affinity than daratumumab and isatuximab for FcγRIIa (CD32a) and FcγRIIIa (CD16a). SAR442085 also exhibited better in vitro antibody-dependent cellular cytotoxicity (ADCC) against a panel of MM cells expressing variable CD38 receptor densities including MM patients' primary plasma cells. The enhanced ADCC of SAR442085 was confirmed using NK-92 cells bearing low and high affinity FcγRIIIa (CD16a)-158F/V variants. Using MM patients' primary bone marrow cells, we confirmed that SAR442085 had an increased ability to engage FcγRIIIa, resulting in higher natural killer (NK) cell activation and degranulation against primary plasma cells than preexisting Fc wild-type anti-CD38 mAbs. Finally, using huFcgR transgenic mice that express human Fcγ receptors under the control of their human regulatory elements, we demonstrated that SAR442085 had higher NK cell-dependent in vivo antitumor efficacy and better survival than daratumumab and isatuximab against EL4 thymoma or VK*MYC myeloma cells overexpressing human CD38. These results highlight the preclinical efficacy of SAR442085 and support the current evaluation of this next-generation anti-CD38 antibody in phase I clinical development in patients with relapsed/refractory MM.
Collapse
|
39
|
Lee HF, Lacbay CM, Boutin R, Matralis AN, Park J, Waller DD, Guan TL, Sebag M, Tsantrizos YS. Synthesis and Evaluation of Structurally Diverse C-2-Substituted Thienopyrimidine-Based Inhibitors of the Human Geranylgeranyl Pyrophosphate Synthase. J Med Chem 2022; 65:2471-2496. [PMID: 35077178 DOI: 10.1021/acs.jmedchem.1c01913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Novel analogues of C-2-substituted thienopyrimidine-based bisphosphonates (C2-ThP-BPs) are described that are potent inhibitors of the human geranylgeranyl pyrophosphate synthase (hGGPPS). Members of this class of compounds induce target-selective apoptosis of multiple myeloma (MM) cells and exhibit antimyeloma activity in vivo. A key structural element of these inhibitors is a linker moiety that connects their (((2-phenylthieno[2,3-d]pyrimidin-4-yl)amino)methylene)bisphosphonic acid core to various side chains. The structural diversity of this linker moiety, as well as the side chains attached to it, was investigated and found to significantly impact the toxicity of these compounds in MM cells. The most potent inhibitor identified was evaluated in mouse and rat for liver toxicity and systemic exposure, respectively, providing further optimism for the potential value of such compounds as human therapeutics.
Collapse
Affiliation(s)
- Hiu-Fung Lee
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Cyrus M Lacbay
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Rebecca Boutin
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Alexios N Matralis
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Jaeok Park
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Daniel D Waller
- Department of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
- Division of Hematology, McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Tian Lai Guan
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Michael Sebag
- Department of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
- Division of Hematology, McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Youla S Tsantrizos
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
40
|
Richard S, Jagannath S. Targeting Nuclear Export Proteins in Multiple Myeloma Therapy. BioDrugs 2022; 36:13-25. [PMID: 35113384 DOI: 10.1007/s40259-021-00514-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2021] [Indexed: 12/23/2022]
Abstract
Nuclear export proteins such as exportin-1 (XPO1) transport tumor-suppressor proteins and other growth-regulatory proteins from the nucleus to the cytoplasm. Overexpression of XPO1 has been observed in several cancers and correlates with shorter event-free and overall survival in multiple myeloma. Selinexor was developed as an oral first-in-class selective inhibitor of nuclear export (SINE) that inhibits XPO1. Preclinical studies in tumor cell lines and mouse models have demonstrated the efficacy of selinexor both as a single agent and in various combinations with known active antimyeloma agents. Results from the pivotal phase II STORM trial led to the US FDA approval of selinexor with dexamethasone in penta-refractory myeloma. Because of the feasibility of combining selinexor with other active antimyeloma agents, the multiarm STOMP trial was initiated and is ongoing, with impressive response rates reported in some of the combination arms thus far. The registrational phase III BOSTON trial demonstrated the superiority of selinexor in combination with bortezomib and dexamethasone as compared with bortezomib and dexamethasone in patients with relapsed refractory multiple myeloma (RRMM) who have received one to three prior anti-MM regimens. The toxicity profile of selinexor is well established and predictable and may be significant unless managed aggressively and preemptively. The most common side effects are thrombocytopenia, anemia, neutropenia, fatigue, nausea, anorexia, and weight loss. Hyponatremia and cataracts seem to be class effects. Other SINE compounds are now being studied in efforts to discover agents that will potentially be better tolerated. Eltanexor is an investigational SINE compound that has shown a more positive toxicity profile in preclinical studies, with reduced central nervous system penetration and gastrointestinal side effects, and is now undergoing clinical investigation. These and other trials will further clarify the role of these innovative agents in the therapeutic advancement of RRMM.
Collapse
Affiliation(s)
- Shambavi Richard
- Mount Sinai Medical Center, One Gustave L. Levy Place, Box 1185, New York, NY, 10029, USA
| | - Sundar Jagannath
- Mount Sinai Medical Center, One Gustave L. Levy Place, Box 1185, New York, NY, 10029, USA.
| |
Collapse
|
41
|
Wang Z, He J, Bach DH, Huang YH, Li Z, Liu H, Lin P, Yang J. Induction of m 6A methylation in adipocyte exosomal LncRNAs mediates myeloma drug resistance. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:4. [PMID: 34980213 PMCID: PMC8722039 DOI: 10.1186/s13046-021-02209-w] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/04/2021] [Indexed: 11/10/2022]
Abstract
Background Therapeutic resistance occurs in most patients with multiple myeloma (MM). One of the key mechanisms for MM drug resistance comes from the interaction between MM cells and adipocytes that inhibits drug-induced apoptosis in MM cells; MM cells reprogram adipocytes to morph into different characterizations, including exosomes, which are important for tumor-stroma cellular communication. However, the mechanism by which exosomes mediate the cellular machinery of the vicious cycle between MM cells and adipocytes remains unclear. Methods Adipocytes were either isolated from bone marrow aspirates of healthy donors or MM patients or derived from mesenchymal stem cells. Co-culturing normal adipocytes with MM cells was used to generate MM-associated adipocytes. Exosomes were collected from the culture medium of adipocytes. Annexin V-binding and TUNEL assays were performed to assess MM cell apoptosis. Methyltransferase activity assay and dot blotting were used to access the m6A methylation activity of methyltransferase like 7A (METTL7A). RIP, MeRIP-seq, and RNA–protein pull down for assessing the interaction between long non-cording RNAs (LncRNAs) and RNA binding proteins were performed. Adipocyte-specific enhancer of zeste homolog 2 (EZH2) knockout mice and MM-xenografted mice were used for evaluating MM therapeutic response in vivo. Results Exosomes collected from MM patient adipocytes protect MM cells from chemotherapy-induced apoptosis. Two LncRNAs in particular, LOC606724 and SNHG1, are significantly upregulated in MM cells after exposure to adipocyte exosomes. The raised LncRNA levels in MM cells are positively correlated to worse outcomes in patients, indicating their clinical relevancy in MM. The functional roles of adipocyte exosomal LOC606724 or SNHG1 in inhibition of MM cell apoptosis are determined by knockdown in adipocytes or overexpression in MM cells. We discovered the interactions between LncRNAs and RNA binding proteins and identified methyltransferase like 7A (METTL7A) as an RNA methyltransferase. MM cells promote LncRNA package into adipocyte exosomes through METTL7A-mediated LncRNA m6A methylation. Exposure of adipocytes to MM cells enhances METTL7A activity in m6A methylation through EZH2-mediated protein methylation. Conclusion This study elucidates an unexplored mechanism of how adipocyte-rich microenvironment exacerbates MM therapeutic resistance and indicates a potential strategy to improve therapeutic efficacy by blocking this vicious exosome-mediated cycle. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02209-w.
Collapse
Affiliation(s)
- Zhiming Wang
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Jin He
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Duc-Hiep Bach
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Yung-Hsing Huang
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Zongwei Li
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Huan Liu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Pei Lin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jing Yang
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
42
|
Fernández-Serrano M, Winkler R, Santos JC, Le Pannérer MM, Buschbeck M, Roué G. Histone Modifications and Their Targeting in Lymphoid Malignancies. Int J Mol Sci 2021; 23:253. [PMID: 35008680 PMCID: PMC8745418 DOI: 10.3390/ijms23010253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
In a wide range of lymphoid neoplasms, the process of malignant transformation is associated with somatic mutations in B cells that affect the epigenetic machinery. Consequential alterations in histone modifications contribute to disease-specific changes in the transcriptional program. Affected genes commonly play important roles in cell cycle regulation, apoptosis-inducing signal transduction, and DNA damage response, thus facilitating the emergence of malignant traits that impair immune surveillance and favor the emergence of different B-cell lymphoma subtypes. In the last two decades, the field has made a major effort to develop therapies that target these epigenetic alterations. In this review, we discuss which epigenetic alterations occur in B-cell non-Hodgkin lymphoma. Furthermore, we aim to present in a close to comprehensive manner the current state-of-the-art in the preclinical and clinical development of epigenetic drugs. We focus on therapeutic strategies interfering with histone methylation and acetylation as these are most advanced in being deployed from the bench-to-bedside and have the greatest potential to improve the prognosis of lymphoma patients.
Collapse
Affiliation(s)
- Miranda Fernández-Serrano
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain; (M.F.-S.); (J.C.S.)
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, 08014 Barcelona, Spain
| | - René Winkler
- Chromatin, Metabolism and Cell Fate Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain; (R.W.); (M.-M.L.P.)
| | - Juliana C. Santos
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain; (M.F.-S.); (J.C.S.)
| | - Marguerite-Marie Le Pannérer
- Chromatin, Metabolism and Cell Fate Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain; (R.W.); (M.-M.L.P.)
| | - Marcus Buschbeck
- Chromatin, Metabolism and Cell Fate Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain; (R.W.); (M.-M.L.P.)
- Program of Personalized and Predictive Medicine of Cancer, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Gaël Roué
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain; (M.F.-S.); (J.C.S.)
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, 08014 Barcelona, Spain
| |
Collapse
|
43
|
Croucher DC, Richards LM, Tsofack SP, Waller D, Li Z, Wei EN, Huang XF, Chesi M, Bergsagel PL, Sebag M, Pugh TJ, Trudel S. Longitudinal single-cell analysis of a myeloma mouse model identifies subclonal molecular programs associated with progression. Nat Commun 2021; 12:6322. [PMID: 34732728 PMCID: PMC8566524 DOI: 10.1038/s41467-021-26598-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 10/12/2021] [Indexed: 12/16/2022] Open
Abstract
Molecular programs that underlie precursor progression in multiple myeloma are incompletely understood. Here, we report a disease spectrum-spanning, single-cell analysis of the Vκ*MYC myeloma mouse model. Using samples obtained from mice with serologically undetectable disease, we identify malignant cells as early as 30 weeks of age and show that these tumours contain subclonal copy number variations that persist throughout progression. We detect intratumoural heterogeneity driven by transcriptional variability during active disease and show that subclonal expression programs are enriched at different times throughout early disease. We then show how one subclonal program related to GCN2 stress response is progressively activated during progression in myeloma patients. Finally, we use chemical and genetic perturbation of GCN2 in vitro to support this pathway as a therapeutic target in myeloma. These findings therefore present a model of precursor progression in Vκ*MYC mice, nominate an adaptive mechanism important for myeloma survival, and highlight the need for single-cell analyses to understand the biological underpinnings of disease progression.
Collapse
Affiliation(s)
- Danielle C Croucher
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Laura M Richards
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Serges P Tsofack
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Daniel Waller
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Zhihua Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ellen Nong Wei
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xian Fang Huang
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Marta Chesi
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - P Leif Bergsagel
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Michael Sebag
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Ontario Institute for Cancer Research, Toronto, ON, Canada.
| | - Suzanne Trudel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
44
|
Toll-like receptor 4 selective inhibition in medullar microenvironment alters multiple myeloma cell growth. Blood Adv 2021; 6:672-678. [PMID: 34714910 PMCID: PMC8791574 DOI: 10.1182/bloodadvances.2020003704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 10/08/2021] [Indexed: 11/20/2022] Open
Abstract
TLR4 is overexpressed on MM MSCs, and its activation promotes the crosstalk with MM cells in enhancing protumoral factors. Specifically targeting TLR4 on MM MSCs strengthens conventional MM therapy and weakens MM development in vivo.
Bone marrow (BM) mesenchymal stromal cells (MSCs) are abnormal in multiple myeloma (MM) and play a critical role by promoting growth, survival, and drug resistance of MM cells. We observed higher Toll-like receptor 4 (TLR4) gene expression in MM MSCs than in MSCs from healthy donors. At the clinical level, we highlighted that TLR4 expression in MM MSCs evolves in parallel with the disease stage. Thus, we reasoned that the TLR4 axis is pivotal in MM by increasing the protumor activity of MSCs. Challenging primary MSCs with TLR4 agonists increased the expression of CD54 and interleukin-6 (IL-6), 2 factors directly implicated in MM MSC-MM cell crosstalk. Then, we evaluated the therapeutic efficacy of a TLR4 antagonist combined or not with conventional treatment in vitro with MSC-MM cell coculture and in vivo with the Vk*MYC mouse model. Selective inhibition of TLR4 specifically reduced the MM MSC ability to support the growth of MM cells in an IL-6-dependent manner and delayed the development of MM in the Vk*MYC mouse model by altering the early disease phase in vivo. For the first time, we demonstrate that specific targeting of the pathological BM microenvironment via TLR4 signaling could be an innovative approach to alter MM pathology development.
Collapse
|
45
|
Sidhu I, Barwe SP, Pillai RK, Gopalakrishnapillai A. Harnessing the Power of Induced Pluripotent Stem Cells and Gene Editing Technology: Therapeutic Implications in Hematological Malignancies. Cells 2021; 10:2698. [PMID: 34685678 PMCID: PMC8534597 DOI: 10.3390/cells10102698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022] Open
Abstract
In vitro modeling of hematological malignancies not only provides insights into the influence of genetic aberrations on cellular and molecular mechanisms involved in disease progression but also aids development and evaluation of therapeutic agents. Owing to their self-renewal and differentiation capacity, induced pluripotent stem cells (iPSCs) have emerged as a potential source of short in supply disease-specific human cells of the hematopoietic lineage. Patient-derived iPSCs can recapitulate the disease severity and spectrum of prognosis dictated by the genetic variation among patients and can be used for drug screening and studying clonal evolution. However, this approach lacks the ability to model the early phases of the disease leading to cancer. The advent of genetic editing technology has promoted the generation of precise isogenic iPSC disease models to address questions regarding the underlying genetic mechanism of disease initiation and progression. In this review, we discuss the use of iPSC disease modeling in hematological diseases, where there is lack of patient sample availability and/or difficulty of engraftment to generate animal models. Furthermore, we describe the power of combining iPSC and precise gene editing to elucidate the underlying mechanism of initiation and progression of various hematological malignancies. Finally, we discuss the power of iPSC disease modeling in developing and testing novel therapies in a high throughput setting.
Collapse
Affiliation(s)
- Ishnoor Sidhu
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, Nemours Children’s Health, Wilmington, DE 19803, USA; (I.S.); (S.P.B.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| | - Sonali P. Barwe
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, Nemours Children’s Health, Wilmington, DE 19803, USA; (I.S.); (S.P.B.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| | - Raju K. Pillai
- National Medical Center, Department of Pathology, City of Hope, Duarte, CA 91105, USA;
| | - Anilkumar Gopalakrishnapillai
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, Nemours Children’s Health, Wilmington, DE 19803, USA; (I.S.); (S.P.B.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
46
|
Dahlhoff J, Manz H, Steinfatt T, Delgado-Tascon J, Seebacher E, Schneider T, Wilnit A, Mokhtari Z, Tabares P, Böckle D, Rasche L, Martin Kortüm K, Lutz MB, Einsele H, Brandl A, Beilhack A. Transient regulatory T-cell targeting triggers immune control of multiple myeloma and prevents disease progression. Leukemia 2021; 36:790-800. [PMID: 34584204 PMCID: PMC8885410 DOI: 10.1038/s41375-021-01422-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022]
Abstract
Multiple myeloma remains a largely incurable disease of clonally expanding malignant plasma cells. The bone marrow microenvironment harbors treatment-resistant myeloma cells, which eventually lead to disease relapse in patients. In the bone marrow, CD4+FoxP3+ regulatory T cells (Tregs) are highly abundant amongst CD4+ T cells providing an immune protective niche for different long-living cell populations, e.g., hematopoietic stem cells. Here, we addressed the functional role of Tregs in multiple myeloma dissemination to bone marrow compartments and disease progression. To investigate the immune regulation of multiple myeloma, we utilized syngeneic immunocompetent murine multiple myeloma models in two different genetic backgrounds. Analyzing the spatial immune architecture of multiple myeloma revealed that the bone marrow Tregs accumulated in the vicinity of malignant plasma cells and displayed an activated phenotype. In vivo Treg depletion prevented multiple myeloma dissemination in both models. Importantly, short-term in vivo depletion of Tregs in mice with established multiple myeloma evoked a potent CD8 T cell- and NK cell-mediated immune response resulting in complete and stable remission. Conclusively, this preclinical in-vivo study suggests that Tregs are an attractive target for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Julia Dahlhoff
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Hannah Manz
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Tim Steinfatt
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Julia Delgado-Tascon
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany
| | - Elena Seebacher
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany
| | - Theresa Schneider
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany
| | - Amy Wilnit
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany
| | - Zeinab Mokhtari
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany
| | - Paula Tabares
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany
| | - David Böckle
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Mildred Scheel Early Career Center, University Hospital of Würzburg, Würzburg, Germany
| | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Manfred B Lutz
- Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany.,Institute for Virology and Immunobiology, Würzburg University, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Brandl
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany. .,Center for Interdisciplinary Clinical Research, University of Würzburg, Würzburg, Germany. .,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
47
|
Expanded natural killer cells augment the antimyeloma effect of daratumumab, bortezomib, and dexamethasone in a mouse model. Cell Mol Immunol 2021; 18:1652-1661. [PMID: 33980993 PMCID: PMC8245645 DOI: 10.1038/s41423-021-00686-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/17/2021] [Indexed: 02/03/2023] Open
Abstract
The use of natural killer (NK) cells is a promising and safe immunotherapeutic approach in the field of cancer immunotherapy. However, combination treatments are required to enhance the effector functions and therapeutic efficacy of NK cells. In this study, we investigated the potential of daratumumab (Dara), bortezomib, and dexamethasone (Dvd) to augment the antitumor effects of NK cells in a multiple myeloma (MM) xenograft mouse model. NK cells were expanded and activated using the K562-OX40 ligand and membrane-bound IL-18 and IL-21 in the presence of IL-2 and IL-15 from peripheral blood mononuclear cells from MM patients. A human MM xenograft model was established using human RPMI8226-RFP-FLuc cells in NOD/SCID IL-2Rγnull (NSG) mice. Tumor-bearing mice were divided into six treatment groups: no treatment, expanded NK cells (eNKs), Dara, Dara + eNKs, Dvd, and Dvd + eNKs. Dvd treatment strongly enhanced the cytotoxicity of eNKs by upregulating expression of NK cell activation ligands, downregulating expression of NK cell inhibitory ligands, and promoting antibody-dependent cellular cytotoxicity. The combination of eNKs with Dvd significantly prolonged mouse survival and reduced the tumor burden and serum M-protein level. Furthermore, Dvd pretreatment significantly increased eNK persistence and homing to MM sites. Our findings suggest that Dvd treatment potentiates the antimyeloma effects of NK cells expanded and activated ex vivo by modulating immune responses in MM-bearing mice.
Collapse
|
48
|
Mehdi SH, Nafees S, Mehdi SJ, Morris CA, Mashouri L, Yoon D. Animal Models of Multiple Myeloma Bone Disease. Front Genet 2021; 12:640954. [PMID: 34163520 PMCID: PMC8215650 DOI: 10.3389/fgene.2021.640954] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a clonal B-cell disorder characterized by the proliferation of malignant plasma cells (PCs) in the bone marrow, the presence of monoclonal serum immunoglobulin, and osteolytic lesions. It is the second most common hematological malignancy and considered an incurable disease despite significant treatment improvements. MM bone disease (MMBD) is defined as the presence of one or more osteolytic bone lesions or diffused osteoporosis with compression fracture attributable to the underlying clonal PC disorder. MMBD causes severe morbidity and increases mortality. Cumulative evidence shows that the interaction of MM cells and bone microenvironment plays a significant role in MM progression, suggesting that these interactions may be good targets for therapy. MM animal models have been developed and studied in various aspects of MM tumorigenesis. In particular, MMBD has been studied in various models, and each model has unique features. As the general features of MM animal models have been reviewed elsewhere, the current review will focus on the features of MMBD animal models.
Collapse
Affiliation(s)
- Syed Hassan Mehdi
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sana Nafees
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Syed Jafar Mehdi
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Carol A Morris
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Ladan Mashouri
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Donghoon Yoon
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
49
|
Pisano M, Cheng Y, Sun F, Dhakal B, D’Souza A, Chhabra S, Knight JM, Rao S, Zhan F, Hari P, Janz S. Laboratory Mice - A Driving Force in Immunopathology and Immunotherapy Studies of Human Multiple Myeloma. Front Immunol 2021; 12:667054. [PMID: 34149703 PMCID: PMC8206561 DOI: 10.3389/fimmu.2021.667054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/28/2021] [Indexed: 11/13/2022] Open
Abstract
Mouse models of human cancer provide an important research tool for elucidating the natural history of neoplastic growth and developing new treatment and prevention approaches. This is particularly true for multiple myeloma (MM), a common and largely incurable neoplasm of post-germinal center, immunoglobulin-producing B lymphocytes, called plasma cells, that reside in the hematopoietic bone marrow (BM) and cause osteolytic lesions and kidney failure among other forms of end-organ damage. The most widely used mouse models used to aid drug and immunotherapy development rely on in vivo propagation of human myeloma cells in immunodeficient hosts (xenografting) or myeloma-like mouse plasma cells in immunocompetent hosts (autografting). Both strategies have made and continue to make valuable contributions to preclinical myeloma, including immune research, yet are ill-suited for studies on tumor development (oncogenesis). Genetically engineered mouse models (GEMMs), such as the widely known Vκ*MYC, may overcome this shortcoming because plasma cell tumors (PCTs) develop de novo (spontaneously) in a highly predictable fashion and accurately recapitulate many hallmarks of human myeloma. Moreover, PCTs arise in an intact organism able to mount a complete innate and adaptive immune response and tumor development reproduces the natural course of human myelomagenesis, beginning with monoclonal gammopathy of undetermined significance (MGUS), progressing to smoldering myeloma (SMM), and eventually transitioning to frank neoplasia. Here we review the utility of transplantation-based and transgenic mouse models of human MM for research on immunopathology and -therapy of plasma cell malignancies, discuss strengths and weaknesses of different experimental approaches, and outline opportunities for closing knowledge gaps, improving the outcome of patients with myeloma, and working towards a cure.
Collapse
Affiliation(s)
- Michael Pisano
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Yan Cheng
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Fumou Sun
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Binod Dhakal
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anita D’Souza
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Saurabh Chhabra
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jennifer M. Knight
- Departments of Psychiatry, Medicine, and Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sridhar Rao
- Division of Hematology, Oncology and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, United States
| | - Fenghuang Zhan
- Myeloma Center, Department of Internal Medicine and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Parameswaran Hari
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Siegfried Janz
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
50
|
Ito T, Yamaguchi Y, Handa H. Exploiting ubiquitin ligase cereblon as a target for small-molecule compounds in medicine and chemical biology. Cell Chem Biol 2021; 28:987-999. [PMID: 34033753 DOI: 10.1016/j.chembiol.2021.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/08/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022]
Abstract
Cereblon (CRBN), originally identified as a gene associated with intellectual disability, was identified as primary target of thalidomide. Accumulating evidence has shown that CRBN is a substrate receptor of Cullin Ring E3 ubiquitin ligase 4 (CRL4) containing DDB1, CUL4, and RBX1, which recognizes specific neosubstrates in the presence of thalidomide or its analogs and induces their ubiquitination and proteasomal degradation. A set of small-molecule, CRBN-binding drugs are known as molecular glue degraders because these compounds promote the interaction between CRBN and its neosubstrates. Moreover, CRBN-based proteolysis-targeting chimeras, heterobifunctional molecules hijacking CRBN and inducing degradation of proteins of interest, have emerged as a promising modality in drug development and are being actively investigated. Meanwhile, the original functions and regulations of CRBN are still largely elusive. In this review, we describe key findings surrounding CRBN since its discovery and then discuss a few unanswered issues.
Collapse
Affiliation(s)
- Takumi Ito
- Department of Chemical Biology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku 160-8402, Japan
| | - Yuki Yamaguchi
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Hiroshi Handa
- Department of Chemical Biology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku 160-8402, Japan.
| |
Collapse
|