Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 3844, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714690

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.


Subject(s)
Multiple Myeloma , Proto-Oncogene Proteins c-myc , Animals , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Humans , Mice , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Cell Transformation, Neoplastic/genetics , Mutation , Signal Transduction/genetics , Mice, Transgenic , NF-kappa B/metabolism , NF-kappa B/genetics , Mutagenesis, Insertional , DNA Copy Number Variations/genetics , Genomics/methods , Translocation, Genetic
2.
Blood Cancer Discov ; 5(1): 34-55, 2024 01 08.
Article in English | MEDLINE | ID: mdl-37767768

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.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Lenalidomide/pharmacology , Lenalidomide/therapeutic use , Transcription Factor AP-1/therapeutic use , Drug Combinations , Immunomodulating Agents
3.
bioRxiv ; 2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37546905

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.

4.
Oncotarget ; 13: 490-504, 2022.
Article in English | MEDLINE | ID: mdl-35251496

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.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Myxoma virus , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Bone Marrow , Bortezomib/pharmacology , Cyclophosphamide , Hematopoietic Stem Cell Transplantation/methods , Humans , Immune Checkpoint Inhibitors , Mice , Mice, Inbred C57BL , Multiple Myeloma/therapy , Oncolytic Virotherapy/methods , Programmed Cell Death 1 Receptor , Transplantation, Autologous
5.
Blood Cancer Discov ; 2(4): 354-369, 2021 07.
Article in English | MEDLINE | ID: mdl-34258584

ABSTRACT

BCMA-CD3-targeting bispecific antibodies (BsAb) are a recently developed immunotherapy class which shows potent tumor killing activity in multiple myeloma (MM). Here, we investigated a murine BCMA-CD3-targeting BsAb in the immunocompetent Vk*MYC and its IMiD-sensitive derivative Vk*MYChCRBN models of MM. The BCMA-CD3 BsAb was safe and efficacious in a subset of mice, but failed in those with high-tumor burden, consistent with clinical reports of BsAb in leukemia. The combination of BCMA-CD3 BsAb with pomalidomide expanded lytic T cells and improved activity even in IMiD resistant high-tumor burden cases. Yet, survival was only marginally extended due to acute toxicity and T cell exhaustion, which impaired T cell persistence. In contrast, the combination with cyclophosphamide was safe and allowed for a tempered pro-inflammatory response associated with long-lasting complete remission. Concurrent cytotoxic therapy with BsAb actually improved T cell persistence and function, offering a promising approach to patients with a large tumor burden.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Animals , Antibodies, Bispecific/pharmacology , Humans , Immunotherapy , Mice , Multiple Myeloma/drug therapy , T-Lymphocytes , Tumor Burden
6.
Blood Cancer Discov ; 1(1): 68-81, 2020 07.
Article in English | MEDLINE | ID: mdl-32954360

ABSTRACT

The most common genetic abnormality in multiple myeloma (MM) is the deletion of chromosome 13, seen in almost half of newly diagnosed patients. Unlike chronic lymphocytic leukemia, where a recurrent minimally deleted region including MIR15A/MIR16-1 has been mapped, the deletions in MM predominantly involve the entire chromosome and no specific driver gene has been identified. Additional candidate loci include RB1 and DIS3, but while biallelic deletion of RB1 is associated with disease progression, DIS3 is a common essential gene and complete inactivation is not observed. The Vk*MYC transgenic mouse model of MM spontaneously acquires del(14), syntenic to human chromosome 13, and Rb1 complete inactivation, but not Dis3 mutations. Taking advantage of this model, we explored the role in MM initiation and progression of two candidate loci on chromosome 13: RB1 and MIR15A/MIR16-1. Monoallelic deletion of Mir15a/Mir16-1 but not Rb1 was sufficient to accelerate the development of monoclonal gammopathy in wildtype mice, and the progression of MM in Vk*MYC mice, resulting in increased expression of Mir15a/Mir16-1 target genes and plasma cell proliferation, which was similarly observed in patients with MM.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , MicroRNAs , Monoclonal Gammopathy of Undetermined Significance , Multiple Myeloma , Animals , Cell Proliferation/genetics , Disease Progression , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mice , MicroRNAs/genetics , Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/genetics , Multiple Myeloma/pathology
7.
Haematologica ; 105(6): 1641-1649, 2020 06.
Article in English | MEDLINE | ID: mdl-31582538

ABSTRACT

The cellular cytotoxicity of APY0201, a PIKfyve inhibitor, against multiple myeloma was initially identified in an unbiased in vitro chemical library screen. The activity of APY0201 was confirmed in all 25 cell lines tested and in 40% of 100 ex vivo patient-derived primary samples, with increased activity in primary samples harboring trisomies and lacking t(11;14). The broad anti-multiple myeloma activity of PIKfyve inhibitors was further demonstrated in confirmatory screens and showed the superior potency of APY0201 when compared to the PIKfyve inhibitors YM201636 and apilimod, with a mid-point half maximal effective concentration (EC50) at nanomolar concentrations in, respectively, 65%, 40%, and 5% of the tested cell lines. Upregulation of genes in the lysosomal pathway and increased cellular vacuolization were observed in vitro following APY0201 treatment, although these cellular effects did not correlate well with responsiveness. We confirm that PIKfyve inhibition is associated with activation of the transcription factor EB, a master regulator of lysosomal biogenesis and autophagy. Furthermore, we established an assay measuring autophagy as a predictive marker of APY0201 sensitivity. Overall, these findings indicate promising activity of PIKfyve inhibitors secondary to disruption of autophagy in multiple myeloma and suggest a strategy to enrich for likely responders.


Subject(s)
Multiple Myeloma , Autophagy , Humans , Lysosomes , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors
8.
Nat Commun ; 9(1): 4832, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30510245

ABSTRACT

The gut microbiota has been causally linked to cancer, yet how intestinal microbes influence progression of extramucosal tumors is poorly understood. Here we provide evidence implying that Prevotella heparinolytica promotes the differentiation of Th17 cells colonizing the gut and migrating to the bone marrow (BM) of transgenic Vk*MYC mice, where they favor progression of multiple myeloma (MM). Lack of IL-17 in Vk*MYC mice, or disturbance of their microbiome delayed MM appearance. Similarly, in smoldering MM patients, higher levels of BM IL-17 predicted faster disease progression. IL-17 induced STAT3 phosphorylation in murine plasma cells, and activated eosinophils. Treatment of Vk*MYC mice with antibodies blocking IL-17, IL-17RA, and IL-5 reduced BM accumulation of Th17 cells and eosinophils and delayed disease progression. Thus, in Vk*MYC mice, commensal bacteria appear to unleash a paracrine signaling network between adaptive and innate immunity that accelerates progression to MM, and can be targeted by already available therapies.


Subject(s)
Eosinophils/immunology , Gastrointestinal Microbiome/immunology , Interleukin-17/immunology , Multiple Myeloma/immunology , Th17 Cells/immunology , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Differentiation/immunology , Cell Movement/immunology , Disease Progression , Eosinophils/metabolism , Humans , Interleukin-17/genetics , Interleukin-17/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Prevotella/immunology , Th17 Cells/metabolism
9.
Nat Med ; 22(12): 1411-1420, 2016 12.
Article in English | MEDLINE | ID: mdl-27841872

ABSTRACT

The cellular inhibitors of apoptosis (cIAP) 1 and 2 are amplified in about 3% of cancers and have been identified in multiple malignancies as being potential therapeutic targets as a result of their role in the evasion of apoptosis. Consequently, small-molecule IAP antagonists, such as LCL161, have entered clinical trials for their ability to induce tumor necrosis factor (TNF)-mediated apoptosis of cancer cells. However, cIAP1 and cIAP2 are recurrently homozygously deleted in multiple myeloma (MM), resulting in constitutive activation of the noncanonical nuclear factor (NF)-κB pathway. To our surprise, we observed robust in vivo anti-myeloma activity of LCL161 in a transgenic myeloma mouse model and in patients with relapsed-refractory MM, where the addition of cyclophosphamide resulted in a median progression-free-survival of 10 months. This effect was not a result of direct induction of tumor cell death, but rather of upregulation of tumor-cell-autonomous type I interferon (IFN) signaling and a strong inflammatory response that resulted in the activation of macrophages and dendritic cells, leading to phagocytosis of tumor cells. Treatment of a MM mouse model with LCL161 established long-term anti-tumor protection and induced regression in a fraction of the mice. Notably, combination of LCL161 with the immune-checkpoint inhibitor anti-PD1 was curative in all of the treated mice.


Subject(s)
Antineoplastic Agents/therapeutic use , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Thiazoles/therapeutic use , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cyclophosphamide/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Female , Gene Expression/drug effects , Humans , Interferon Type I/drug effects , Interferon Type I/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Transgenic , Middle Aged , Multiple Myeloma/immunology , Neoplasm Recurrence, Local/immunology , Phagocytosis/drug effects , Phagocytosis/immunology , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...