1
|
Uruno K, Iwano H, Hayashi T, Hatano R, Komuro K, Kawahatsu K, Doi T, Yuda S. Carfilzomib-induced pulmonary hypertension in a patient with multiple myeloma. J Cardiol Cases 2024; 30:172-175. [PMID: 39534307 PMCID: PMC11551454 DOI: 10.1016/j.jccase.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/04/2024] [Accepted: 07/29/2024] [Indexed: 11/16/2024] Open
Abstract
A 59-year-old female being treated for multiple myeloma (MM) was referred to the Division of Cardiology due to edema and dyspnea. She developed dyspnea on exertion 2 months previously when carfilzomib, a second-generation selective proteasome inhibitor which was approved for treatment of relapsed and refractory MM, was introduced, and facial edema appeared thereafter. The electrocardiogram showed sinus rhythm with T-wave inversion on extensive leads and the chest X-ray showed cardiomegaly. Although cancer therapeutics-related cardiac dysfunction was assumed to be the complication, echocardiogram revealed no evidence of elevated left ventricular filling pressure whereas elevated tricuspid regurgitation velocity (3.2 m/s) with right ventricular systolic dysfunction suggested pre-capillary pulmonary hypertension (PH). Right heart catheterization demonstrated elevated mean pulmonary artery pressure (33 mmHg) along with high pulmonary vascular resistance (11.54 Wood Units) and normal pulmonary capillary wedge pressure (9 mmHg), confirming the echocardiographic findings. After ruling out other causes, PH associated with carfilzomib was diagnosed. Cessation of carfilzomib along with pulmonary vasodilator therapy led to improvement of symptoms and reduction of right heart size along with reduced estimated pulmonary systolic pressure 2 months later. Although carfilzomib-induced PH is rare, we need to consider its possibility when we find PH in patients receiving carfilzomib. Learning objective While adverse cardiovascular events are often found in patients with multiple myeloma (MM) after use of carfilzomib, the occurrence of pulmonary hypertension (PH) is reported to be rare. Because temporal association of echocardiographic findings to carfilzomib therapy plays a key role for the diagnosis of drug-associated PH, serial echocardiographic examinations should be performed when we start carfilzomib therapy in refractory MM patients.
Collapse
Affiliation(s)
- Kosuke Uruno
- Division of Cardiology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Hiroyuki Iwano
- Division of Cardiology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Toshiaki Hayashi
- Division of Hematology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Ryosuke Hatano
- Division of Cardiology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Kaoru Komuro
- Division of Cardiology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Kandoh Kawahatsu
- Division of Cardiology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Takahiro Doi
- Division of Cardiology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Satoshi Yuda
- Division of Cardiology, Teine Keijinkai Hospital, Sapporo, Japan
| |
Collapse
|
2
|
Sundaravel SH, Marar RI, Abbasi MA, Baljevic M, Stone JR. Bortezomib-Induced Reversible Cardiomyopathy: Recovered with Guideline-Directed Medical Therapy. Cureus 2021; 13:e20295. [PMID: 35024253 PMCID: PMC8742456 DOI: 10.7759/cureus.20295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 11/29/2022] Open
Abstract
Bortezomib (BTZ) is a proteasome inhibitor (PI) used for the treatment of several hematologic malignancies, including multiple myeloma (MM), and various lymphomas including mantle cell lymphoma (MCL). It acts via disruption of the ubiquitin-proteasome pathway which plays a major role in regulating cell cycle and inhibiting synthesis of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-KB). The ubiquitin-proteasome pathway is also important in maintaining the integral signaling in cardiac myocytes. By inhibiting this system, BTZ induces cellular apoptosis in cancer cells, and possibly the cardiomyocytes. BTZ-induced cardiotoxicity in monotherapy and combination treatments is not well described in the literature. We observed a series of three patients who developed cardiotoxicity after treatment with BTZ. All patients had echocardiograms every 3 months until recovery to assess ejection fraction (EF) and global longitudinal strain (GLS). Two of the patients had a cardiac MRI (CMR) conducted during follow-up to assess for late gadolinium enhancement (LGE). The median age of our patients was 55 years (range 37-74). Two of them had MM, while one patient had MCL. Table 1 demonstrates patient demographics, past medical histories, and the cumulative dose and duration of BTZ therapy. Of the three patients, only one had a heart failure exacerbation at diagnosis. The other two patients were diagnosed with asymptomatic left ventricular systolic dysfunction on routine pre-transplant echocardiograms. Most importantly, all three patients had improvement or normalization of cardiac function with discontinuation of BTZ and initiation of guideline-directed medical therapy (GDMT) for heart failure. The median duration to recovery was 5 months (range 3-13). One patient had underlying non-compaction cardiomyopathy, and although EF did not normalize, it recovered to his previous baseline. All 3 patients had improvement in GLS. Two patients underwent CMRI at the time of cardiomyopathy diagnosis and neither of them had any late gadolinium enhancement. Since there was no routine pre-treatment echocardiogram, using the GLS trend to detect subclinical cardiac dysfunction was not possible. This case series demonstrates that BTZ-induced cardiomyopathy is potentially reversible with discontinuation of the drug and early initiation of GDMT. Further studies are needed to determine the ideal surveillance strategy for BTZ-induced cardiomyopathy.
Collapse
|
3
|
Mitsiades CS. Biological and Translational Considerations regarding the Recent Therapeutic Successes and Upcoming Challenges for Multiple Myeloma. Cold Spring Harb Perspect Med 2021; 11:a034900. [PMID: 32928892 PMCID: PMC8247558 DOI: 10.1101/cshperspect.a034900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Even though multiple myeloma (MM) is still considered incurable, the therapeutic management of this disease has undergone a major transformation over the last two decades, with several new classes of therapeutics and diverse options for their combined use in many different regimens that have contributed to major improvement in overall survival of patients. This review discusses key themes underlying the pharmacological and immune-based therapies that represent the cornerstones of this progress. A major part of the clinical progress achieved by these classes' therapeutics has depended on the targeting of molecular pathways with distinct or preferential roles for the biology of plasma cells-normal or malignant-and the ability of many of these agents to be incorporated into combination regimens that exhibit enhanced antimyeloma responses, without precipitating acceptable levels of toxicity. This review also discusses why these advances have not yet translated into curative outcomes and how these remaining barriers could be overcome.
Collapse
Affiliation(s)
- Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Medicine, Harvard Medical School, Broad Institute of MIT & Harvard, Boston, Massachusetts 02215, USA
| |
Collapse
|
4
|
Patel VG, Cornell RF. Cardiovascular Complications Associated with Multiple Myeloma Therapies: Incidence, Pathophysiology, and Management. Curr Oncol Rep 2019; 21:29. [PMID: 30834998 DOI: 10.1007/s11912-019-0784-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW Multiple myeloma is a common hematologic malignancy characterized by recurrent relapsing disease course requiring use of various therapies. Over the past few decades, significant advancements in the treatment of myeloma have occurred including routine use of proteasome inhibitors and immunomodulatory drugs. These have effectively improved survival; however, some also have increased risk of cardiovascular toxicity. Here, we will review the incidence, pathophysiology, and management of cardiovascular complications associated with antimyeloma agents. RECENT FINDINGS Cardiovascular complications associated with myeloma treatment are common. These cardiovascular complications include accelerated hypertension, ischemic heart disease, congestive heart failure, arrhythmia, pulmonary hypertension, venous thromboembolism, and arterial thromboembolism. Thromboprophylactic strategies during treatment with immunomodulatory agents and screening strategies to detect changes in myocardial function prior to the development of overt heart failure have occurred. Cardiovascular complications associated with proteasome inhibitors and immunomodulatory drugs are an important component in supportive care of patients with myeloma. The incidence of cardiotoxicity is high, and, as such, early intervention and collaborative efforts between cardiologists and oncologists to mitigate and effectively manage these complications are imperative. Additional studies are needed to clarify the underlying pathophysiology and evaluate effective strategies for prevention and treatment.
Collapse
Affiliation(s)
- Vivek G Patel
- Department of Internal Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Robert F Cornell
- Department of Internal Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
5
|
Guha A, Armanious M, Fradley MG. Update on cardio-oncology: Novel cancer therapeutics and associated cardiotoxicities. Trends Cardiovasc Med 2019; 29:29-39. [DOI: 10.1016/j.tcm.2018.06.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/22/2018] [Accepted: 06/03/2018] [Indexed: 02/08/2023]
|
6
|
Tian DW, Wu ZL, Jiang LM, Gao J, Wu CL, Hu HL. Neural precursor cell expressed, developmentally downregulated 8 promotes tumor progression and predicts poor prognosis of patients with bladder cancer. Cancer Sci 2018; 110:458-467. [PMID: 30407690 PMCID: PMC6317957 DOI: 10.1111/cas.13865] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/31/2018] [Accepted: 11/05/2018] [Indexed: 12/15/2022] Open
Abstract
Neddylation has been researched in many different human carcinomas. However, the roles of neural precursor cell expressed, developmentally downregulated 8 (NEDD8) in bladder cancer are still unknown. Our study was the first study which systematically investigated the possible functions of NEDD8 in bladder cancer (BC) progression. We carried out immunohistochemistry to explore associations between the expression of NEDD8 in tumor tissues and clinical outcomes of patients. RT‐qPCR and western blot were used to detect the expressional levels of genes. The biological abilities of cell proliferation, migration and invasion were researched by in vitro and in vivo experiments. Results were as follows: Data from The Cancer Genome Atlas (TCGA) database showed that NEDD8 was overexpressed in BC tissues and was associated with poor patient survival. Results of immunohistochemistry found that NEDD8 was significantly associated with poor clinical outcomes of BC patients. Suppression of NEDD8 could inhibit the proliferation, migration and invasion of tumor cells. Knocking down NEDD8 could induce apoptosis and G2 phase arrest of cell cycle progression. In vivo, suppression of NEDD8 restricted growth and metastasis of tumors in mice. In conclusion, NEDD8 has important roles in regulating the progression of BC cells and was associated with poor prognosis of patients; hence, it may become a potential therapeutic target of BC.
Collapse
Affiliation(s)
- Da-Wei Tian
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.,Sino-Singapore Eco-City Hospital of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, China
| | - Zhou-Liang Wu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.,Sino-Singapore Eco-City Hospital of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, China
| | - Li-Ming Jiang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.,Sino-Singapore Eco-City Hospital of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, China
| | - Jie Gao
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.,Sino-Singapore Eco-City Hospital of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, China
| | - Chang-Li Wu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.,Sino-Singapore Eco-City Hospital of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, China
| | - Hai-Long Hu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.,Sino-Singapore Eco-City Hospital of Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, China
| |
Collapse
|
7
|
Lee DH, Fradley MG. Cardiovascular Complications of Multiple Myeloma Treatment: Evaluation, Management, and Prevention. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2018; 20:19. [DOI: 10.1007/s11936-018-0618-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
8
|
Oerlemans R, Berkers CR, Assaraf YG, Scheffer GL, Peters GJ, Verbrugge SE, Cloos J, Slootstra J, Meloen RH, Shoemaker RH, Dijkmans BAC, Scheper RJ, Ovaa H, Jansen G. Proteasome inhibition and mechanism of resistance to a synthetic, library-based hexapeptide. Invest New Drugs 2018; 36:797-809. [PMID: 29442210 PMCID: PMC6153520 DOI: 10.1007/s10637-018-0569-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/31/2018] [Indexed: 12/14/2022]
Abstract
Background The hexapeptide 4A6 (Ac-Thr(tBu)-His(Bzl)-Thr(Bzl)-Nle-Glu(OtBu)-Gly-Bza) was isolated from a peptide library constructed to identify peptide-based transport inhibitors of multidrug resistance (MDR) efflux pumps including P-glycoprotein and Multidrug Resistance-associated Protein 1. 4A6 proved to be a substrate but not an inhibitor of these MDR efflux transporters. In fact, 4A6 and related peptides displayed potent cytotoxic activity via an unknown mechanism. Objective To decipher the mode of cytotoxic activity of 4A6. Methods Screening of 4A6 activity was performed against the NCI60 panel of cancer cell lines. Possible interactions of 4A6 with the 26S proteasome were assessed via proteasome activity and affinity labeling, and cell growth inhibition studies with leukemic cells resistant to the proteasome inhibitor bortezomib (BTZ). Results The NCI60 panel COMPARE analysis revealed that 4A6 had an activity profile overlapping with BTZ. Consistently, 4A6 proved to be a selective and reversible inhibitor of β5 subunit (PSMB5)-associated chymotrypsin-like activity of the 26S proteasome. This conclusion is supported by several lines of evidence: (i) inhibition of chymotrypsin-like proteasome activity by 4A6 and related peptides correlated with their cell growth inhibition potencies; (ii) 4A6 reversibly inhibited functional β5 active site labeling with the affinity probe BodipyFL-Ahx3L3VS; and (iii) human myeloid THP1 cells with acquired BTZ resistance due to mutated PSMB5 were highly (up to 287-fold) cross-resistant to 4A6 and its related peptides. Conclusion 4A6 is a novel specific inhibitor of the β5 subunit-associated chymotrypsin-like proteasome activity. Further exploration of 4A6 as a lead compound for development as a novel proteasome-targeted drug is warranted.
Collapse
Affiliation(s)
- Ruud Oerlemans
- Departments of Rheumatology, Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Rm 2.46, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Celia R Berkers
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - George L Scheffer
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sue Ellen Verbrugge
- Departments of Rheumatology, Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Rm 2.46, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | | | - Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ben A C Dijkmans
- Departments of Rheumatology, Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Rm 2.46, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Rik J Scheper
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Huib Ovaa
- Division of Cell Biology II, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Chemical Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerrit Jansen
- Departments of Rheumatology, Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Rm 2.46, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Eugênio AIP, Fook-Alves VL, de Oliveira MB, Fernando RC, Zanatta DB, Strauss BE, Silva MRR, Porcionatto MA, Colleoni GWB. Proteasome and heat shock protein 70 (HSP70) inhibitors as therapeutic alternative in multiple myeloma. Oncotarget 2017; 8:114698-114709. [PMID: 29383113 PMCID: PMC5777725 DOI: 10.18632/oncotarget.22815] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/05/2017] [Indexed: 11/25/2022] Open
Abstract
HSP70 connects multiple signaling pathways that work synergistically to protect tumor cells from death by proteotoxic stress and represents a possible target to establish a new approach for multiple myeloma treatment. Therefore, bioluminescent cell lines RPMI8226-LUC-PURO and U266-LUC-PURO were treated with HSP70 (VER155008) and/or proteasome (bortezomib) inhibitors and immunodeficient mice were used for subcutaneous xenograft models to evaluate tumor growth reduction and tumor growth inhibition after treatment. Bioluminescence imaging was used to follow tumor response. Treatment with bortezomib showed ∼60% of late apoptosis in RPMI8226-LUC-PURO (without additional benefit of VER155008 in this cell line). However, U266-LUC-PURO showed ∼60% of cell death after treatment with VER155008 (alone or with bortezomib). RPMI8226-LUC-PURO xenograft presented tumor reduction by bioluminescence imaging after treatment with bortezomib, VER155008 or drug combination compared to controls. Treatment with bortezomib, alone or combined with VER155008, showed inhibition of tumor growth assessed by bioluminescence imaging after one week in both RPMI8226-LUC-PURO and U266-LUC-PURO cell lines when compared to controls. In conclusion, our study shows that the combination of proteasome and HSP70 inhibitors induced cell death in tumor cells in vitro (late apoptosis induction) and in vivo (inhibition of tumor growth) with special benefit in U266-LUC-PURO, bearing 17p deletion.
Collapse
Affiliation(s)
- Angela Isabel Pereira Eugênio
- Discipline of Hematology e Hemotherapy, Department of Clinical and Experimental Oncology, Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brazil
| | - Veruska Lia Fook-Alves
- Discipline of Hematology e Hemotherapy, Department of Clinical and Experimental Oncology, Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brazil
| | - Mariana Bleker de Oliveira
- Discipline of Hematology e Hemotherapy, Department of Clinical and Experimental Oncology, Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brazil
| | - Rodrigo Carlini Fernando
- Discipline of Hematology e Hemotherapy, Department of Clinical and Experimental Oncology, Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brazil
| | - Daniela B Zanatta
- Center of Translational Investigation in Oncology, Cancer Institute of the State of São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Bryan Eric Strauss
- Center of Translational Investigation in Oncology, Cancer Institute of the State of São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Gisele Wally Braga Colleoni
- Discipline of Hematology e Hemotherapy, Department of Clinical and Experimental Oncology, Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brazil
| |
Collapse
|
10
|
Fradley MG, Groarke JD, Laubach J, Alsina M, Lenihan DJ, Cornell RF, Maglio M, Shain KH, Richardson PG, Moslehi J. Recurrent cardiotoxicity potentiated by the interaction of proteasome inhibitor and immunomodulatory therapy for the treatment of multiple myeloma. Br J Haematol 2017; 180:271-275. [PMID: 29048105 DOI: 10.1111/bjh.14970] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/14/2017] [Indexed: 02/02/2023]
Abstract
Patients with multiple myeloma (MM) have improved treatment options, including immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs). Despite their efficacy, increased rates of cardiovascular (CV) complications occur in patients exposed to some of these therapies. While previous research has focused on identifying the toxicities inherent to each specific agent, the CV side effects may be potentiated by the combination of PIs and IMiDs plus dexamethasone. We present a patient with MM with recurrent cardiotoxicity only when exposed to combination PI and IMiD-based therapy. We also review the literature in this context, and propose a potential algorithm for cardiotoxicity prevention in this population.
Collapse
Affiliation(s)
- Michael G Fradley
- Cardio-Oncology Program, Department of Cardiovascular Sciences, University of South Florida Morsani College of Medicine and H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - John D Groarke
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Jacob Laubach
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Melissa Alsina
- H. Lee Moffitt Cancer Center and Research Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Daniel J Lenihan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Cardio-Oncology Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert F Cornell
- Cardio-Oncology Program, Vanderbilt University School of Medicine, Nashville, TN, USA.,Division of Hematology-Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michelle Maglio
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth H Shain
- H. Lee Moffitt Cancer Center and Research Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Paul G Richardson
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Javid Moslehi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Cardio-Oncology Program, Vanderbilt University School of Medicine, Nashville, TN, USA.,Division of Hematology-Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
11
|
Lai G, Sun R, Wu J, Zhang B, Zhao Y. 20-HETE regulated PSMB5 expression via TGF-β/Smad signaling pathway. Prostaglandins Other Lipid Mediat 2017; 134:123-130. [PMID: 28807746 DOI: 10.1016/j.prostaglandins.2017.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/09/2017] [Accepted: 08/08/2017] [Indexed: 10/19/2022]
Abstract
We previously found that 20-hydroxyeicosatetraeonic acid (20-HETE) showed an effect on proteasome activity in cytochrome P450 F2 (CYP4F2) transgenic mice. Proteasome subunit β5 (PSMB5) is a primary subunit of the proteasome. In the current study, we examine whether 20-HETE has any affect on PSMB5. We found that PSMB5 was upregulated in the liver, but downregulated in the kidney of transgenic mice, when compared with wild-type mice. Luciferase reporter gene experiments and electrophoretic mobility shift assays (EMSA) suggested that Smad3 directly associated with the putative Smad binding element (SBE) of the Psmb5 promoter. Furthermore, the binding affinity was different between the liver and kidney, and can be regulated by 20-HETE. Compared to wild mice, both TGF-β1 and Smad3 phosphorylation were increased in the liver but decreased in the kidney of transgenic mice. SB431542, an inhibitor of TGF-β receptor I kinase activity, can reverse the changes induced in PSMB5 by 20-HETE in vitro. Taken together, our data demonstrated that 20-HETE upregulated the expression of PSMB5 by activating the TGF-β/Smad signaling pathway in the liver, but downregulated the expression of PSMB5 by inhibiting the TGF-β/Smad signaling pathway in the kidney of transgenic mice.
Collapse
Affiliation(s)
- Guangrui Lai
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ru Sun
- Department of Genetics, Shenyang Women's and Children's Hospital, Shenyang, Liaoning, China
| | - Jingjing Wu
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning, China
| | - Bijun Zhang
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yanyan Zhao
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China; Department of Medical Genetics, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
12
|
Schäfer J, Welti L, Seckinger A, Burhenne J, Theile D, Weiss J. Cellular effect and efficacy of carfilzomib depends on cellular net concentration gradient. Cancer Chemother Pharmacol 2017; 80:71-79. [DOI: 10.1007/s00280-017-3335-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/05/2017] [Indexed: 12/16/2022]
|
13
|
Hervé M, Ibrahim EC. Proteasome inhibitors to alleviate aberrant IKBKAP mRNA splicing and low IKAP/hELP1 synthesis in familial dysautonomia. Neurobiol Dis 2017; 103:113-122. [PMID: 28404519 DOI: 10.1016/j.nbd.2017.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/23/2017] [Accepted: 04/05/2017] [Indexed: 12/27/2022] Open
Abstract
FD is a rare neurodegenerative disorder caused by a mutation of the IKBKAP gene, which induces low expression levels of the Elongator subunit IKAP/hELP1 protein. A rational strategy for FD treatment could be to identify drugs increasing IKAP/hELP1 expression levels by blocking protein degradation pathways such as the 26S proteasome. Proteasome inhibitors are promising molecules emerging in cancer treatment and could thus constitute an enticing pharmaceutical strategy for FD treatment. Therefore, we tested three proteasome inhibitors on FD human olfactory ecto-mesenchymal stem cells (hOE-MSCs): two approved by the Food and Drug Administration (FDA) and European Medicines Agency (EMA), bortezomib and carfilzomib, as well as epoxomicin. Although all 3 inhibitors demonstrated activity in correcting IKBKAP mRNA aberrant splicing, carfilzomib was superior in enhancing IKAP/hELP1 quantity. Moreover, we observed a synergistic effect of suboptimal doses of carfilzomib on kinetin in improving IKBKAP isoforms ratio and IKAP/hELP1 expression levels allowing to counterbalance carfilzomib toxicity. Finally, we identified several dysregulated miRNAs after carfilzomib treatment that target proteasome-associated mRNAs and determined that IKAP/hELP1 deficiency in FD pathology is correlated to an overactivity of the 26S proteasome. Altogether, these results reinforce the rationale for using chemical compounds inhibiting the 26S proteasome as an innovative option for FD and a promising therapeutic pathway for many other neurodegenerative diseases.
Collapse
Affiliation(s)
- Mylène Hervé
- Aix-Marseille Univ, CNRS, CRN2M, 13344 Marseille Cedex 15, France
| | - El Chérif Ibrahim
- Aix-Marseille Univ, CNRS, CRN2M, 13344 Marseille Cedex 15, France; Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, 13385 Marseille Cedex 5, France.
| |
Collapse
|
14
|
Matthews GM, de Matos Simoes R, Dhimolea E, Sheffer M, Gandolfi S, Dashevsky O, Sorrell JD, Mitsiades CS. NF-κB dysregulation in multiple myeloma. Semin Cancer Biol 2016; 39:68-76. [PMID: 27544796 DOI: 10.1016/j.semcancer.2016.08.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 12/29/2022]
Abstract
The nuclear factor-κB (NF-κB) transcription factor family plays critical roles in the pathophysiology of hematologic neoplasias, including multiple myeloma. The current review examines the roles that this transcription factor system plays in multiple myeloma cells and the nonmalignant accessory cells of the local microenvironment; as well as the evidence indicating that a large proportion of myeloma patients harbor genomic lesions which perturb diverse genes regulating the activity of NF-κB. This article also discusses the therapeutic targeting of the NF-κB pathway using proteasome inhibitors, a pharmacological class that has become a cornerstone in the therapeutic management of myeloma; and reviews some of the future challenges and opportunities for NF-κB-related research in myeloma.
Collapse
Affiliation(s)
- Geoffrey M Matthews
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Ricardo de Matos Simoes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Eugen Dhimolea
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Michal Sheffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Sara Gandolfi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Jeffrey D Sorrell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States.
| |
Collapse
|
15
|
Stewart AK. Carfilzomib for the treatment of patients with relapsed and/or refractory multiple myeloma. Future Oncol 2015; 11:2121-36. [PMID: 26125319 DOI: 10.2217/fon.15.123] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Carfilzomib is a proteasome inhibitor that irreversibly binds to its target, resulting in sustained proteasomal inhibition with minimal off-target effects. As a single agent, carfilzomib has demonstrated durable antimyeloma activity with manageable toxicities, which has resulted in its approval in Argentina, Israel, Mexico and the USA for the treatment of patients with relapsed and refractory multiple myeloma. Data from ongoing Phase III studies that are evaluating carfilzomib in earlier lines of therapy may facilitate an expanded indication for this agent, as well as for regulatory approval in the EU. This article summarizes the chemistry, pharmacokinetics, pharmacodynamics and available clinical data for carfilzomib in the treatment of patients with multiple myeloma.
Collapse
Affiliation(s)
- A Keith Stewart
- Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ 85259, USA
| |
Collapse
|