1
|
Wesley CD, Sansonetti A, Neutel CHG, Krüger DN, De Meyer GRY, Martinet W, Guns PJ. Short-Term Proteasome Inhibition: Assessment of the Effects of Carfilzomib and Bortezomib on Cardiac Function, Arterial Stiffness, and Vascular Reactivity. BIOLOGY 2024; 13:844. [PMID: 39452152 PMCID: PMC11504385 DOI: 10.3390/biology13100844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Proteasome inhibitors such as bortezomib and carfilzomib induce apoptosis and are a cornerstone in the treatment of relapsed or refractory multiple myeloma. However, concerns have emerged concerning their link to cancer therapy-related cardiovascular dysfunction (CTRCD). Bortezomib, a reversible first-generation inhibitor, and carfilzomib, a second-generation irreversible inhibitor, are associated with hypertension, heart failure, and cardiac arrhythmias. The current study investigated the effects of bortezomib and carfilzomib on cardiac (left ventricular ejection fraction, LVEF) and vascular (arterial stiffness, vascular reactivity) function. Cardiac function assessment aimed to build upon existing evidence of proteasome inhibitors CTRCD, while arterial stiffness served as an early indicator of potential vascular remodeling. Groups of 12-week-old C57BL/6J male mice (n = 8 per group) were randomly assigned to receive vehicle, carfilzomib (8 mg/kg I.P.), or bortezomib (0.5 mg/kg I.P.). Additionally, proteasome inhibition was assessed in mice treated with L-NAME (0.5 mg/kg) to induce hypertension. Cardiac and vascular parameters were evaluated via echocardiography on days 0 and 3. On day 6, mice were sacrificed for ex vivo analysis of arterial stiffness and vascular reactivity. Overall, no changes in arterial stiffness were detected either in vivo or ex vivo at basal pressures. However, a steeper pressure-stiffness curve was observed for carfilzomib in normotensive (p < 0.01) and hypertensive (p < 0.0001) mice ex vivo. Additionally, in hypertensive mice, carfilzomib decreased LVEF (p = 0.06), with bortezomib exhibiting similar trends. Vascular reactivity remained largely unchanged, but proteasome inhibition tended to enhance endothelial-independent relaxations in both control and hypertensive mice. In conclusion, short-term treatment with carfilzomib and bortezomib is considered relatively safe for the protocols assessed in the study.
Collapse
Affiliation(s)
- Callan D. Wesley
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences University of Antwerp, Campus Drie Eiken, 2610 Antwerp, Belgium; (A.S.); (C.H.G.N.); (D.N.K.); (G.R.Y.D.M.); (W.M.); (P.-J.G.)
| | | | | | | | | | | | | |
Collapse
|
2
|
Grynblat J, Khouri C, Hlavaty A, Jaïs X, Savale L, Chaumais MC, Kularatne M, Jevnikar M, Boucly A, Antigny F, Perros F, Simonneau G, Sitbon O, Humbert M, Montani D. Characteristics and outcomes of patients developing pulmonary hypertension associated with proteasome inhibitors. Eur Respir J 2024; 63:2302158. [PMID: 38697649 DOI: 10.1183/13993003.02158-2023] [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: 12/01/2023] [Accepted: 04/15/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) has been described in patients treated with proteasome inhibitors (PIs). Our objective was to evaluate the association between PIs and PAH. METHODS Characteristics of incident PAH cases previously treated with carfilzomib or bortezomib were analysed from the French pulmonary hypertension registry and the VIGIAPATH programme from 2004 to 2023, concurrently with a pharmacovigilance disproportionality analysis using the World Health Organization (WHO) global database (VigiBase) and a meta-analysis of randomised controlled trials. RESULTS 11 incident cases of PI-associated PAH were identified (six with carfilzomib and five with bortezomib) with a female:male ratio of 2.7:1, a median age of 61 years, and a median delay between PI first exposure and PAH of 6 months. Four patients died (two from right heart failure, one from respiratory distress and one from an unknown cause). At diagnosis, six were in New York Heart Association Functional Class III/IV with severe haemodynamic impairment (median mean pulmonary arterial pressure 39 mmHg, cardiac index 2.45 L·min-1·m-2 and pulmonary vascular resistance 7.2 WU). In the WHO pharmacovigilance database, 169 cases of PH associated with PI were reported since 2013 with significant signals of disproportionate reporting (SDR) for carfilzomib, regardless of the definition of cases or control group. However, SDR for bortezomib were inconsistent. The systematic review identified 17 clinical trials, and carfilzomib was associated with a significantly higher risk of dyspnoea, severe dyspnoea and PH compared with bortezomib. CONCLUSION PIs may induce PAH in patients undergoing treatment, with carfilzomib emitting a stronger signal than bortezomib, and these patients should be monitored closely.
Collapse
Affiliation(s)
- Julien Grynblat
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
- These authors contributed equally to this work
| | - Charles Khouri
- Univ. Grenoble Alpes, HP2 Laboratory, INSERM U 1300, Grenoble, France
- Pharmacovigilance Unit and Clinical Pharmacology Department, Grenoble Alpes University Hospital, Grenoble, France
- These authors contributed equally to this work
| | - Alex Hlavaty
- Univ. Grenoble Alpes, HP2 Laboratory, INSERM U 1300, Grenoble, France
- Pharmacovigilance Unit and Clinical Pharmacology Department, Grenoble Alpes University Hospital, Grenoble, France
| | - Xavier Jaïs
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Laurent Savale
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Marie Camille Chaumais
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- School of Pharmacy, University of Paris-Saclay, Saclay, France
- AP-HP, Department of Pharmacy, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Mithum Kularatne
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
- Division of Respiratory Medicine, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mitja Jevnikar
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Athénaïs Boucly
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Frédéric Perros
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Gérald Simonneau
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Olivier Sitbon
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - David Montani
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
3
|
Frenzel L, Decaux O, Macro M, Belhadj-Merzoug K, Manier S, Touzeau C, Leleu X, Frère C, Lecompte T, Perrot A, Avet-Loiseau H, Moreau P, Chalayer E. Venous thromboembolism prophylaxis and multiple myeloma patients in real-life: Results of a large survey and clinical guidance recommendations from the IFM group. Thromb Res 2024; 233:153-164. [PMID: 38064842 DOI: 10.1016/j.thromres.2023.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/30/2023] [Accepted: 11/20/2023] [Indexed: 01/01/2024]
Abstract
Venous thromboembolism (VTE) remains a critical issue in the management of patients with multiple myeloma (MM), particularly when immunomodulatory drugs (IMiDs) combined with dexamethasone therapy are being prescribed as first-line and relapse therapy. One possible explanation for the persistent high rates of VTE, is the use of inappropriate thromboprophylaxis strategies for patients starting antimyeloma treatment. To tackle the issue, the Intergroupe francophone du myélome (IFM) offered convenient guidance for VTE thromboprophylaxis in MM patients initiating systemic therapy. This guidance is mainly supported by the results of a large survey on the clinical habits regarding VTE of physicians who are substantially involved in daily care of MM patients. VTE prophylaxis should be considered for all patients treated with IMiDs in combination with dexamethasone, in the absence of significant comorbidities, such as renal failure or bleeding risk. Anticoagulant should be preferred to antiplatelet agents for thromboprophylaxis. Despite the absence of large randomized controlled trials comparing those attitudes/options, available data on direct oral anticoagulants, which are already used in daily management of MM patients, are consistent with their potential usefulness for VTE prophylaxis in such patients. However, in order to implement a personalized continuous improvement strategy, clinicians must to be organized to collect all the data regarding this management. In other situations, thromboprophylaxis should be evaluated by using risk models and after careful evaluation of the risk/benefit ratio.
Collapse
Affiliation(s)
- Laurent Frenzel
- Service d'Hématologie Adulte et unité d'hémostase clinique, Hôpital Necker, Institut IMAGINE - INSERM U 1163/CNRS ERL 8254, Paris, France.
| | | | - Margaret Macro
- Institut d'Hématologie de Basse Normandie (IHBN), CHU Côte de Nacre, Caen, France
| | | | - Salomon Manier
- Hematology department, CHU Lille, Lille University, INSERM UMR-S1277, Lille, France
| | - Cyrille Touzeau
- Department of Hematology, University Hospital Hôtel-Dieu, 44093 Nantes, France; Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers, INSERM UMR 1307, CNRS UMR 6075, Nantes, France
| | - Xavier Leleu
- Hématologie Biologique, Hôpital Pontchaillou University Hospital of Rennes, Rennes, France
| | - Corinne Frère
- Service d'Hématologie Biologique, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | | | - Aurore Perrot
- Institut Universitaire du Cancer de Toulouse-Oncopole and Centre de Recherches en Cancerologie de Toulouse Institut National de la Sante et de la Recherche Medicale, Toulouse, France
| | - Hervé Avet-Loiseau
- Institut Universitaire du Cancer de Toulouse-Oncopole and Centre de Recherches en Cancerologie de Toulouse Institut National de la Sante et de la Recherche Medicale, Toulouse, France
| | - Philippe Moreau
- Department of Hematology, University Hospital Hôtel-Dieu, 44093 Nantes, France; Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers, INSERM UMR 1307, CNRS UMR 6075, Nantes, France
| | - Emilie Chalayer
- Hématologie clinique, Institut de Cancérologie Hématologie Universitaire, CHU St Etienne Unité INSERM SAINBIOSE, U1059, Université Jean Monnet, St-Etienne, France
| |
Collapse
|
4
|
Habib CN, Ali AE, Anber NH, George MY. Lactoferrin ameliorates carfilzomib-induced renal and pulmonary deficits: Insights to the inflammasome NLRP3/NF-κB and PI3K/Akt/GSK-3β/MAPK axes. Life Sci 2023; 335:122245. [PMID: 37926296 DOI: 10.1016/j.lfs.2023.122245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 10/20/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023]
Abstract
AIMS Carfilzomib, an irreversible proteasome inhibitor, has been increasingly used to treat multiple myeloma worldwide. However, case studies showed its treatment has been associated with cardiac, renal, and pulmonary deleterious effects. Lactoferrin is an iron-binding glycoprotein present in milk. It is a multifunctional protein with antimicrobial activity, antitumor, antioxidant, and anti-inflammatory effects. Thus, this study aimed to assess the protective effects of lactoferrin against carfilzomib-induced nephrotoxicity and pulmonary toxicity, in addition to identifying the possible underlying molecular mechanisms. MAIN METHODS Mice were treated with lactoferrin (300 mg/kg/day) concomitantly with carfilzomib (4 mg/kg, i.p.) twice weekly for three weeks. Kidney and lung indices, serum creatinine, blood urea nitrogen (BUN), uric acid, kidney injury molecule-1 (KIM-1), lactate dehydrogenase (LDH), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and histological examination were assessed. In addition, biochemical analyses of the inflammasome NLRP3/NF-κB and PI3K/Akt/GSK-3β/MAPK axes were conducted. KEY FINDINGS Treatment with lactoferrin decreased serum levels of creatinine, BUN, uric acid, KIM-1, ALP, AST, and LDH and reversed carfilzomib-induced histological changes in both kidney and lung. The inflammatory markers NLRP3, p65 NF-kB, caspases1, interleukin-1β, and interleukin-18, as well as the MAPK signaling pathway, were significantly reduced in renal and pulmonary tissues of mice following lactoferrin administration. Moreover, lactoferrin significantly counteracted carfilzomib-induced reduced expression of pAkt and pGSK-3β in both renal and pulmonary tissues. SIGNIFICANCE The current study suggests lactoferrin might be a promising candidate for ameliorating carfilzomib-induced nephrotoxicity and pulmonary toxicity.
Collapse
Affiliation(s)
- Christine N Habib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Alaa E Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Nahla H Anber
- Department of Biochemistry, the Emergency Hospital, Mansoura University, Mansoura, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt.
| |
Collapse
|
5
|
Abdul-Rahman T, Dunham A, Huang H, Bukhari SMA, Mehta A, Awuah WA, Ede-Imafidon D, Cantu-Herrera E, Talukder S, Joshi A, Sundlof DW, Gupta R. Chemotherapy Induced Cardiotoxicity: A State of the Art Review on General Mechanisms, Prevention, Treatment and Recent Advances in Novel Therapeutics. Curr Probl Cardiol 2023; 48:101591. [PMID: 36621516 DOI: 10.1016/j.cpcardiol.2023.101591] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/08/2023]
Abstract
As medicine advances to employ sophisticated anticancer agents to treat a vast array of oncological conditions, it is worth considering side effects associated with several chemotherapeutics. One adverse effect observed with several classes of chemotherapy agents is cardiotoxicity which leads to reduced ejection fraction (EF), cardiac arrhythmias, hypertension and Ischemia/myocardial infarction that can significantly impact the quality of life and patient outcomes. Research into possible mechanisms has elucidated several mechanisms, such as ROS generation, calcium overload and apoptosis. However, there is a relative scarcity of literature detailing the relationship between the exact mechanism of cardiotoxicity for each anticancer agent and observed clinical effects. This review comprehensively describes cardiotoxicity associated with various classes of anticancer agents and possible mechanisms. Further research exploring possible mechanisms for cardiotoxicity observed with anticancer agents could provide valuable insight into susceptibility for developing symptoms and management guidelines. Chemotherapeutics are associated with several side effects. Several classes of chemotherapy agents cause cardiotoxicity leading to a reduced ejection fraction (EF), cardiac arrhythmias, hypertension, and Ischemia/myocardial infarction. Research into possible mechanisms has elucidated several mechanisms, such as ROS generation, calcium overload, and apoptosis. However, there is a relative scarcity of literature detailing the relationship between the exact mechanism of cardiotoxicity for each anticancer agent and observed clinical effects. This review describes cardiotoxicity associated with various classes of anticancer agents and possible mechanisms. Further research exploring mechanisms for cardiotoxicity observed with anticancer agents could provide insight that will guide management.
Collapse
Affiliation(s)
| | - Alden Dunham
- University of South Florida Morsani College of Medicine, FL
| | - Helen Huang
- Royal College of Surgeons in Ireland, University of Medicine and Health Science, Dublin, Ireland
| | | | - Aashna Mehta
- University of Debrecen-Faculty of Medicine, Debrecen, Hungary
| | - Wireko A Awuah
- Sumy State University, Toufik's World Medical Association, Ukraine
| | | | - Emiliano Cantu-Herrera
- Department of Clinical Sciences, Division of Health Sciences, University of Monterrey, San Pedro Garza García, Nuevo León, México
| | | | - Amogh Joshi
- Department of Cardiology, Lehigh Valley Health Network, Allentown, PA
| | - Deborah W Sundlof
- Department of Cardiology, Lehigh Valley Health Network, Allentown, PA
| | - Rahul Gupta
- Department of Cardiology, Lehigh Valley Health Network, Allentown, PA.
| | | |
Collapse
|
6
|
Maddipati V, Sankhyan P, Goswami DP, Mahajan A. Pulmonary hypertension in patients with multiple myeloma: A comprehensive review. Pulm Circ 2023; 13:e12210. [PMID: 37063748 PMCID: PMC10098295 DOI: 10.1002/pul2.12210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/02/2023] [Accepted: 03/08/2023] [Indexed: 04/18/2023] Open
Abstract
Multiple myeloma (MM) is a common hematological malignancy resulting from clonal proliferation of plasma cells and is defined by criteria set forth by the international myeloma working group. Pulmonary hypertension (PH) is defined by an elevated mean pulmonary artery pressure >20 mmHg measured during right heart catheterization. Echocardiography-diagnosed PH is relatively common in patients with MM and has been associated with increased mortality, morbidity, and poor stem cell transplant outcomes. PH in patients with MM (PH-MM) is usually multifactorial in origin. MM disease-specific factors, host comorbidities, and treatment-related adverse effects are the key factors for the development of PH-MM. Pragmatically, patients with PH-MM can be grouped into either (i) PH in patients with a new diagnosis of MM or (ii) PH that develops or worsens along the way of MM treatment. In the latter group, drug-induced PH, venous thromboembolism, pulmonary veno occlusive disease, and cardiotoxicity should be considered as possible causes. PH-MM should be evaluated and managed in a multidisciplinary setting. Select individuals with PH-MM could be considered for pulmonary vasodilators at PH-specialized centers.
Collapse
Affiliation(s)
- Veeranna Maddipati
- Division of Pulmonary and Critical Care Medicine, Pulmonary Vascular MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Pratyaksha Sankhyan
- Division of Pulmonary and Critical Care MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Durga P. Goswami
- Department of Internal MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | | |
Collapse
|
7
|
Zheng Y, Huang S, Xie B, Zhang N, Liu Z, Tse G, Liu T. Cardiovascular Toxicity of Proteasome Inhibitors in Multiple Myeloma Therapy. Curr Probl Cardiol 2023; 48:101536. [PMID: 36481392 DOI: 10.1016/j.cpcardiol.2022.101536] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
The treatment for multiple myeloma has advanced significantly over the past few decades. Proteasome inhibitors have become the cornerstone of the treatment of multiple myeloma. However, proteasome inhibitors have shown cardiovascular complications such as hypertension, pulmonary hypertension, heart failure, arrhythmias, ischaemic heart disease and thromboembolism. Detection, monitoring and management of proteasome inhibitor-related cardiovascular toxicity are essential to improve clinical outcomes for patients. Proposed mechanisms of proteasome inhibitor-related cardiovascular toxicity are apoptosis, prolonged inhibition of the ubiquitin-proteasome system, accumulation of improperly folded proteins within cardiomyocytes and higher protein phosphatase 2A activity. To better understand the mechanisms underlying cardiotoxicity, further in vitro and in vivo experiments are required to investigate these hypotheses. Combined use of metformin or angiotensin II receptor blockers with the proteasome inhibitor, carfilzomib, showed an emerging role as a prophylactic therapy because they can preserve heart function in multiple myeloma patients. Metformin is expected to be an effective therapeutic intervention for the management of carfilzomib-induced cardiotoxicity. There has been evidence that three compounds, apremilast, rutin, and dexrazoxane, can reverse carfilzomib-induced cardiotoxicity in rats. The future transition from animal experiments to clinical trials is worth waiting for.
Collapse
Affiliation(s)
- Yi Zheng
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin China
| | - Shan Huang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin China
| | - Bingxin Xie
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin China
| | - Nan Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin China
| | - Zhiqiang Liu
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China; The province and ministry co-sponsored collaborative innovation center for medical epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin China; Kent and Medway Medical School, University of Kent and Canterbury Christ Church University, Canterbury, UK; School of Nursing and Health Studies, Metropolitan University, Hong Kong, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin China.
| |
Collapse
|
8
|
Astarita A, Mingrone G, Airale L, Cesareo M, Colomba A, Catarinella C, Leone D, Gay F, Bringhen S, Veglio F, Milan A, Vallelonga F. Carfilzomib-Based Regimen and Cardiotoxicity in Multiple Myeloma: Incidence of Cardiovascular Events and Organ Damage in Carfilzomib-Dexamethasone versus Carfilzomib-Lenalidomide-Dexamethasone. A Real-Life Prospective Study. Cancers (Basel) 2023; 15:cancers15030955. [PMID: 36765915 PMCID: PMC9913715 DOI: 10.3390/cancers15030955] [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: 12/31/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Carfilzomib-mediated cardiotoxicity in multiple myeloma (MM) is a well-established adverse effect, however limited data are available on the comparison of cardiovascular complications in patients treated with Carfilzomib-dexamethasone (target dose of K 56 mg/m2) versus Carfilzomib-lenalidomide-dexamethasone (target dose of K 27 mg/m2) beyond controlled trials. A total of 109 patients were enrolled, 47 (43%) received Kd and 62 (57%) KRd. They then underwent a baseline and follow-up evaluation including trans-thoracic echocardiography and arterial stiffness estimation. All types of cardiovascular and hypertensive events occurred more frequently in the Kd group compared with the KRd (59% vs. 40% and 55% vs. 35.5% patients, respectively, p ≤ 0.05), with higher incidence of hypertensive. The time of onset of any type of CVAE, and of major and hypertensive events was shorter in the Kd regimen (p ≤ 0.05). At follow-up, Kd patients more frequently developed signs of cardiac (decline of global longitudinal strain) and vascular organ damage (rise of pulse wave velocity), as compared with KRd. Despite the older age, longer history of MM and longer period of pre-treatment of Kd patients, these factors did not increase the probability of incidence for all types of cardiovascular events at multivariate analysis (p > 0.05). In conclusion, the Kd regimen showed greater cardiovascular toxicity and earlier onset of events with respect to KRd. Thus, a closer and thorough follow-up should be considered.
Collapse
Affiliation(s)
- Anna Astarita
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Giulia Mingrone
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Lorenzo Airale
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Marco Cesareo
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Anna Colomba
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Cinzia Catarinella
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Dario Leone
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Francesca Gay
- Myeloma Unit, Department of Medical Sciences, Division of Hematology, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Sara Bringhen
- Myeloma Unit, Department of Medical Sciences, Division of Hematology, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Franco Veglio
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Alberto Milan
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
| | - Fabrizio Vallelonga
- Hypertension Unit, Department of Medical Sciences, Division of Internal Medicine, AO “Città Della Salute e Della Scienza” University Hospital, 10126 Turin, Italy
- Correspondence: ; Tel.: +39-011-633-6959
| |
Collapse
|
9
|
Alam MF, Hijri SI, Alshahrani S, Alqahtani SS, Jali AM, Ahmed RA, Adawi MM, Algassmi SM, Shaheen ES, Moni SS, Anwer T. Zingerone Attenuates Carfilzomib-Induced Cardiotoxicity in Rats through Oxidative Stress and Inflammatory Cytokine Network. Int J Mol Sci 2022; 23:ijms232415617. [PMID: 36555257 PMCID: PMC9779556 DOI: 10.3390/ijms232415617] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Carfilzomib (CFZ) is an anticancer medication acting as a selective proteasome inhibitor. However, it can cause cardiovascular problems, increasing mortality and morbidity. This study aimed to investigate whether zingerone (ZRN) could help reduce carfilzomib-induced cardiotoxicity in Wistar albino rats. Rats were divided into five groups of six animals each. The first group received normal saline as a control (NC); the second group received multiple doses (six) of CFZ (4 mg/kg) intraperitoneally (IP); the third and fourth groups received zingerone (50 mg/kg and 100 mg/kg oral) along with six doses of CFZ for 16 days; and the fifth group received only 100 mg/kg zingerone orally. Hematological, biochemical, oxidative stress, and histopathological studies confirmed the findings of CFZ-induced cardiotoxicity. We found that ZRN significantly attenuated the effects of CFZ on oxidative stress by enhancing the antioxidant properties of glutathione (GSH), catalase (CAT), and superoxide dismutase (SOD). Additionally, ZRN reduces inflammatory cytokines and apoptotic markers, such as IL-1β, IL-6, TNFα, and caspase-3. Overall, zingerone prevents carfilzomib-induced cardiotoxicity in rats, as evidenced by histopathological studies.
Collapse
Affiliation(s)
- Mohammad Firoz Alam
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Correspondence:
| | - Sami I. Hijri
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Saad S. Alqahtani
- Department of Pharmacy Practice, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Pharmacy Practice Research Unit, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Abdulmajeed M. Jali
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Rayan A. Ahmed
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Mansour M. Adawi
- Department of Histopathology, King Fahad Hospital, Jazan 45142, Saudi Arabia
| | - Sameeh M. Algassmi
- Department of Histopathology, King Fahad Hospital, Jazan 45142, Saudi Arabia
| | - Emad Sayed Shaheen
- Department of Animal House, Medical Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Sivakumar S. Moni
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Tarique Anwer
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
10
|
Mineralocorticoid Receptor Pathway Is a Key Mediator of Carfilzomib-induced Nephrotoxicity: Preventive Role of Eplerenone. Hemasphere 2022; 6:e791. [PMID: 36285072 PMCID: PMC9584194 DOI: 10.1097/hs9.0000000000000791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022] Open
Abstract
Carfilzomib is an irreversible proteasome inhibitor indicated for relapsed/refractory multiple myeloma. Carfilzomib toxicity includes renal adverse effects (RAEs) of obscure pathobiology. Therefore, we investigated the mechanisms of nephrotoxicity developed by Carfilzomib. In a first experimental series, we used our previously established in vivo mouse models of Carfilzomib cardiotoxicity, that incorporated 2 and 4 doses of Carfilzomib, to identify whether Carfilzomib affects renal pathways. Hematology and biochemical analyses were performed, while kidneys underwent histological and molecular analyses. In a second and third experimental series, the 4 doses protocol was repeated for 24 hours urine collection and proteomic/metabolomic analyses. To test an experimental intervention, primary murine collecting duct tubular epithelial cells were treated with Carfilzomib and/or Eplerenone and Metformin. Finally, Eplerenone was orally co-administered with Carfilzomib daily (165 mg/kg) in the 4 doses protocol. We additionally used material from 7 patients to validate our findings and patients underwent biochemical analysis and assessment of renal mineralocorticoid receptor (MR) axis activation. In vivo screening showed that Carfilzomib-induced renal histological deficits and increased serum creatinine, urea, NGAL levels, and proteinuria only in the 4 doses protocol. Carfilzomib decreased diuresis, altered renal metabolism, and activated MR axis. This was consistent with the cytotoxicity found in primary murine collecting duct tubular epithelial cells, whereas Carfilzomib + Eplerenone co-administration abrogated Carfilzomib-related nephrotoxic effects in vitro and in vivo. Renal SGK-1, a marker of MR activation, increased in patients with Carfilzomib-related RAEs. Conclusively, Carfilzomib-induced renal MR/SGK-1 activation orchestrates RAEs and water retention both in vivo and in the clinical setting. MR blockade emerges as a potential therapeutic approach against Carfilzomib-related nephrotoxicity.
Collapse
|
11
|
Insulin-Degrading Enzyme Is a Non Proteasomal Target of Carfilzomib and Affects the 20S Proteasome Inhibition by the Drug. Biomolecules 2022; 12:biom12020315. [PMID: 35204815 PMCID: PMC8869475 DOI: 10.3390/biom12020315] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/12/2022] [Accepted: 01/27/2022] [Indexed: 02/07/2023] Open
Abstract
Carfilzomib is a last generation proteasome inhibitor (PI) with proven clinical efficacy in the treatment of relapsed/refractory multiple myeloma. This drug is considered to be extremely specific in inhibiting the chymotrypsin-like activity of the 20S proteasome, encoded by the β5 subunit, overcoming some bortezomib limitations, the first PI approved for multiple myeloma therapy which is however burdened by a significant toxicity profile, due also to its off-target effects. Here, molecular approaches coupled with molecular docking studies have been used to unveil that the Insulin-Degrading Enzyme, a ubiquitous and highly conserved Zn2+ peptidase, often found to associate with proteasome in cell-based models, is targeted by carfilzomib in vitro. The drug behaves as a modulator of IDE activity, displaying an inhibitory effect over 10-fold lower than for the 20S. Notably, the interaction of IDE with the 20S enhances in vitro the inhibitory power of carfilzomib on proteasome, so that the IDE-20S complex is an even better target of carfilzomib than the 20S alone. Furthermore, IDE gene silencing after delivery of antisense oligonucleotides (siRNA) significantly reduced carfilzomib cytotoxicity in rMC1 cells, a validated model of Muller glia, suggesting that, in cells, the inhibitory activity of this drug on cell proliferation is somewhat linked to IDE and, possibly, also to its interaction with proteasome.
Collapse
|
12
|
Elucidating Carfilzomib's Induced Cardiotoxicity in an In Vivo Model of Aging: Prophylactic Potential of Metformin. Int J Mol Sci 2021; 22:ijms222010956. [PMID: 34681615 PMCID: PMC8537073 DOI: 10.3390/ijms222010956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Carfilzomib is a first-line proteasome inhibitor indicated for relapsed/refractory multiple myeloma (MM), with its clinical use being hampered by cardiotoxic phenomena. We have previously established a translational model of carfilzomib cardiotoxicity in young adult mice, in which metformin emerged as a prophylactic therapy. Considering that MM is an elderly disease and that age is an independent risk factor for cardiotoxicity, herein, we sought to validate carfilzomib’s cardiotoxicity in an in vivo model of aging. Methods: Aged mice underwent the translational two- and four-dose protocols without and with metformin. Mice underwent echocardiography and were subsequently sacrificed for molecular analyses in the blood and cardiac tissue. Results: Carfilzomib decreased proteasomal activity both in PBMCs and myocardium in both protocols. Carfilzomib induced mild cardiotoxicity after two doses and more pronounced cardiomyopathy in the four-dose protocol, while metformin maintained cardiac function. Carfilzomib led to an increased Bip expression and decreased AMPKα phosphorylation, while metformin coadministration partially decreased Bip expression and induced AMPKα phosphorylation, leading to enhanced myocardial LC3B-dependent autophagy. Conclusion: Carfilzomib induced cardiotoxicity in aged mice, an effect significantly reversed by metformin. The latter possesses translational importance as it further supports the clinical use of metformin as a potent prophylactic therapy.
Collapse
|
13
|
Yang JZ, Buckstaff T, Narezkina A, Fernandes TM. Carfilzomib-associated pulmonary arterial hypertension in multiple myeloma. Pulm Circ 2021; 11:20458940211049300. [PMID: 34603687 PMCID: PMC8485285 DOI: 10.1177/20458940211049300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/07/2021] [Indexed: 11/15/2022] Open
Abstract
Drug-induced pulmonary arterial hypertension (PAH) is constantly evolving as new drugs are developed. Carfilzomib is a recently approved therapy for relapsed and refractory multiple myeloma. While it has been associated with cardiovascular adverse events, such as ischemic heart disease and heart failure, PAH has not been a well-described side effect. We present two patients who developed PAH associated with initiation of carfilzomib. They both initially presented with severe dyspnea, had elevated right ventricular systolic pressure on transthoracic echocardiography and ultimately underwent right heart catheterization. With discontinuation of carfilzomib, both patients had improvement in hemodynamics. However, one patient required initiation of PAH-targeted therapies and has had worsening right ventricular function again despite permanent discontinuation of carfilzomib. It is important to recognize the association between carfilzomib and PAH. Echocardiography can be an important initial screening tool. PAH from carfilzomib therapy may be reversible, especially if diagnosed early; however, extended follow-up is essential.
Collapse
Affiliation(s)
- Jenny Z Yang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Taylor Buckstaff
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Anna Narezkina
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Timothy M Fernandes
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
14
|
Zhai Y, Ye X, Hu F, Xu J, Guo X, Cao Y, Lin Z, Zhou X, Guo Z, He J. Cardiovascular Toxicity of Carfilzomib: The Real-World Evidence Based on the Adverse Event Reporting System Database of the FDA, the United States. Front Cardiovasc Med 2021; 8:735466. [PMID: 34646873 PMCID: PMC8503541 DOI: 10.3389/fcvm.2021.735466] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/31/2021] [Indexed: 02/02/2023] Open
Abstract
Background: Carfilzomib, an effective proteasome inhibitor agent for the therapy of relapsed and refractory multiple myeloma, has been related to a significant number of cardiovascular events. However, patterns of cardiovascular complications associated with this agent remain poorly characterized in real-world settings. Objective: To gain further insight into the frequency, spectrum, clinical features, timing, and outcomes of carfilzomib-related cardiovascular toxicities. Methods: This disproportionality (case/non-case) study was conducted leveraging records from FAERS database from 2014 to 2019. Cardiovascular events were defined and broadly categorized eight entities using narrow version of the Standardized MedDRA Queries (SMQs). Reporting odds ratios (ROR) and information component (IC) were calculated to measure disproportionality. Additionally, statistical shrinkage was applied to reduce false-positive signals. Results: The final number of records involved was 28,479,963, with 3,370 records submitted for carfilzomib related cardiovascular events. Significant disproportionality association between carfilzomib administration and cardiovascular events was captured (IC025/ROR025 = 0.85/1.95) when exploring in the entire database. Upon further analysis, all eight broad categories of cardiovascular toxicities were disproportionately associated with carfilzomib with varying frequencies, time-to-onset, and severities. Cardiomyopathy-related complications (N = 1,301, 38.61%), embolic and thrombotic events (N = 821, 24.36%), and cardiac failure (N = 765, 22.70%) largely comprised the reported problems. Notably, the strongest signal was detected for cardiac failure (IC025/ROR025 = 1.33/2.59), followed by pulmonary hypertension (IC025/ROR025 = 1.19/2.34). Median onset time of cardiovascular events was 41days (Q1-Q3: 9-114 days), with the shortest median time being 16 days (Q1-Q3: 4-85 days) for ischemic heart disease, with the longest time being 68 days (Q1-Q3: 21-139 days) for embolic and thrombotic events. Torsade de pointes/QT prolongation was identified as a new complication (IC025/ROR025 = 0.33/1.29) and was particularly noteworthy for highest death proportion (44.11%). Conclusions: Treatment with carfilzomib can lead to severe and versatile cardiovascular events. Early and intensive monitoring is important, particularly in the first 3 months after carfilzomib initiation. Maximizing the benefit while reducing potential cardiovascular harms of carfilzomib should become a priority.
Collapse
Affiliation(s)
- Yinghong Zhai
- School of Medicine, Tongji University, Shanghai, China
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Xiaofei Ye
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Fangyuan Hu
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Jinfang Xu
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Xiaojing Guo
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Yang Cao
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Zhen Lin
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Xiang Zhou
- School of Medicine, Tongji University, Shanghai, China
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Zhijian Guo
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Jia He
- School of Medicine, Tongji University, Shanghai, China
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| |
Collapse
|
15
|
Ramirez RL, Pienkos SM, de Jesus Perez V, Zamanian RT. Pulmonary Arterial Hypertension Secondary to Drugs and Toxins. Clin Chest Med 2021; 42:19-38. [PMID: 33541612 DOI: 10.1016/j.ccm.2020.11.008] [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: 11/30/2022]
Abstract
Pulmonary arterial hypertension secondary to drugs and toxins is an important subgroup of group 1 pulmonary hypertension associated with significant morbidity and mortality. Many drugs and toxins have emerged as risk factors for pulmonary arterial hypertension, which include anorexigens, illicit agents, and several US Food and Drug Administration-approved therapeutic medications. Drugs and toxins are classified as possible or definite risk factors for pulmonary arterial hypertension. This article reviews agents that have been implicated in the development of pulmonary arterial hypertension, their pathologic mechanisms, and methods to prevent the next deadly outbreak of drug- and toxin-induced pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Ramon L Ramirez
- Division of Pulmonary, Allergy and Critical Care, Stanford University School of Medicine, 300 Pasteur Drive, Room S102, Stanford, CA 94305, USA
| | - Shaun M Pienkos
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Room S102, Stanford, CA 94305, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary, Allergy and Critical Care, Stanford University School of Medicine, 300 Pasteur Drive, Room S102, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford, CA, USA
| | - Roham T Zamanian
- Division of Pulmonary, Allergy and Critical Care, Stanford University School of Medicine, 300 Pasteur Drive, Room S102, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford, CA, USA.
| |
Collapse
|
16
|
Yee AJ. The role of carfilzomib in relapsed/refractory multiple myeloma. Ther Adv Hematol 2021; 12:20406207211019612. [PMID: 34163580 PMCID: PMC8191071 DOI: 10.1177/20406207211019612] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/04/2021] [Indexed: 12/20/2022] Open
Abstract
Carfilzomib is the second proteasome inhibitor approved for relapsed multiple myeloma. Since its approval in 2012, carfilzomib has been an active and versatile drug, based on its efficacy as a single agent; superiority as a doublet with dexamethasone compared with bortezomib and dexamethasone; and as a partner in diverse three drug combinations such as with lenalidomide or daratumumab. While it has an established place in relapsed disease, clinicians should be aware of its cardiovascular and renal adverse event profile, which is manageable, in order to optimize outcomes. This review will provide a perspective on the current and future role of carfilzomib in relapsed/refractory multiple myeloma.
Collapse
Affiliation(s)
- Andrew J. Yee
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical
School, 55 Fruit St, Boston, MA 02114, USA
| |
Collapse
|
17
|
Mathur P, Thanendrarajan S, Lopez-Candales A. Severe Right-Sided Heart Failure and Pulmonary Hypertension with Carfilzomib Treatment in Multiple Myeloma. HEART VIEWS : THE OFFICIAL JOURNAL OF THE GULF HEART ASSOCIATION 2021; 21:296-299. [PMID: 33986932 PMCID: PMC8104323 DOI: 10.4103/heartviews.heartviews_107_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 11/03/2020] [Indexed: 12/24/2022]
Abstract
Carfilzomib, a second-generation irreversible proteasome inhibitor, is currently considered the preferred therapy for relapsed and refractory multiple myeloma. There are several cardiovascular adverse effects described with carfilzomib chemotherapy most commonly being hypertension, dyspnea and decreased cardiac ejection fraction. We report a case of newonset pulmonary hypertension with right ventricular (RV) heart failure in a patient receiving carfilzomib. Awareness of this rare side effect of this drug is essential for prompt diagnosis and management. We also propose close monitoring of RV and pulmonary artery pressures along with left ventricular function in echocardiographic assessment in patients with carfilzomib chemotherapy.
Collapse
Affiliation(s)
- Pankaj Mathur
- Division of Cardiovascular Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sharmilan Thanendrarajan
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Angel Lopez-Candales
- Division of Cardiovascular Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
18
|
Mingrone G, Astarita A, Airale L, Maffei I, Cesareo M, Crea T, Bruno G, Leone D, Avenatti E, Catarinella C, Salvini M, Cetani G, Gay F, Bringhen S, Veglio F, Vallelonga F, Milan A. Effects of Carfilzomib Therapy on Left Ventricular Function in Multiple Myeloma Patients. Front Cardiovasc Med 2021; 8:645678. [PMID: 33969010 PMCID: PMC8096903 DOI: 10.3389/fcvm.2021.645678] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/22/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Carfilzomib improves the prognosis of multiple myeloma (MM) patients but significantly increases cardiovascular toxicity. The timing and effect of Carfilzomib therapy on the left ventricular function is still under investigation. We sought to assess the echocardiographic systo-diastolic changes, including global longitudinal strain (GLS), in patients treated with Carfilzomib and to identify predictors of increased risk of cardiovascular adverse events (CVAEs) during therapy. Methods: Eighty-eight patients with MM performed a baseline cardiovascular evaluation comprehensive of transthoracic echocardiogram (TTE) before the start of Carfilzomib therapy and after 6 months. All patients were clinically followed up to early identify the occurrence of CVAEs during the whole therapy duration. Results: After Carfilzomib treatment, mean GLS slightly decreased (−22.2% ± 2.6 vs. −21.3% ± 2.5; p < 0.001). Fifty-eight percent of patients experienced CVAEs during therapy: 71% of them had uncontrolled hypertension, and 29% had major CVAEs or CV events not related to arterial hypertension. GLS variation during therapy was not related to an increased risk of CVAEs; however, patients with baseline GLS ≥ −21% and/or left ventricular ejection fraction (LVEF) ≤ 60% had a greater risk of major CVAEs (OR = 6.2, p = 0.004; OR = 3.7, p = 0.04, respectively). Carfilzomib led to a higher risk of diastolic dysfunction (5.6 vs. 13.4%, p = 0.04) and to a rise in E/e′ ratio (8.9 ± 2.7 vs. 9.7 ± 3.7; p = 0.006). Conclusion: Carfilzomib leads to early LV function impairment early demonstrated by GLS changes and diastolic dysfunction. Baseline echocardiographic parameters, especially GLS and LVEF, might improve cardiovascular risk stratification before treatment.
Collapse
Affiliation(s)
- Giulia Mingrone
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Anna Astarita
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Lorenzo Airale
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Ilaria Maffei
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Marco Cesareo
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Teresa Crea
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Giulia Bruno
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Dario Leone
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Eleonora Avenatti
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Cinzia Catarinella
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Marco Salvini
- Myeloma Unit, Division of Haematology, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Giusy Cetani
- Myeloma Unit, Division of Haematology, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Francesca Gay
- Myeloma Unit, Division of Haematology, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Sara Bringhen
- Myeloma Unit, Division of Haematology, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Franco Veglio
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Fabrizio Vallelonga
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| | - Alberto Milan
- Department of Internal Medicine and Hypertension Division, "Città della Salute e della Scienza" Hospital, University of Turin, Turin, Italy
| |
Collapse
|
19
|
Multiple Myeloma Patients Undergoing Carfilzomib: Development and Validation of a Risk Score for Cardiovascular Adverse Events Prediction. Cancers (Basel) 2021; 13:cancers13071631. [PMID: 33915804 PMCID: PMC8036868 DOI: 10.3390/cancers13071631] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/19/2021] [Accepted: 03/28/2021] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Despite the relationship between Carfilzomib (CFZ) therapy in multiple myeloma (MM) and cardiovascular adverse events (CVAEs), no specific validated protocols on cardiovascular risk assessment are available. In this prospective study, we investigated major predictors of CVAEs prior to starting CFZ, applying the European Myeloma Network management protocol (EMN). Five predictors were identified: office systolic blood pressure, 24-h blood pressure variability, left ventricular hypertrophy, pulse wave velocity value and global longitudinal strain. The resulting ‘CVAEs risk score’ defined a low- and a high-risk group (negative predicting value for the high-risk group of 90%). 62 patients experienced one or more CVAEs: 17 major and 45 hypertension-related events. In conclusion, CVAEs are frequent and a specific management protocol is required. The EMN protocol and ‘CFZ risk score’ proved to be effective in estimating the baseline risk of CVAEs during CFZ therapy in MM patients, targeting the appropriate follow-up. Abstract Cardiovascular adverse events (CVAEs) are linked to Carfilzomib (CFZ) therapy in multiple myeloma (MM); however, no validated protocols on cardiovascular risk assessment are available. In this prospective study, the effectiveness of the European Myeloma Network protocol (EMN) in cardiovascular risk assessment was investigated, identifying major predictors of CVAEs. From January 2015 to March 2020, 116 MM patients who had indication for CFZ therapy underwent a baseline evaluation (including blood pressure measurements, echocardiography and arterial stiffness estimation) and were prospectively followed. The median age was 64.53 ± 8.42 years old, 56% male. Five baseline independent predictors of CVAEs were identified: office systolic blood pressure, 24-h blood pressure variability, left ventricular hypertrophy, pulse wave velocity value and global longitudinal strain. The resulting ‘CVAEs risk score’ distinguished a low- and a high-risk group, obtaining a negative predicting value for the high-risk group of 90%. 52 patients (44.9%) experienced one or more CVAEs: 17 (14.7%) had major and 45 (38.7%) had hypertension-related events. In conclusion, CVAEs are frequent and a specific management protocol is crucial. The EMN protocol and the risk score proved to be useful to estimate the baseline risk for CVAEs during CFZ therapy, allowing the identification of higher-risk patients.
Collapse
|
20
|
McGregor PC, Boosalis V, Aragam J. Carfilzomib-induced pulmonary hypertension with associated right ventricular dysfunction: A case report. SAGE Open Med Case Rep 2021; 9:2050313X21994031. [PMID: 33643656 PMCID: PMC7890743 DOI: 10.1177/2050313x21994031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/18/2021] [Indexed: 11/17/2022] Open
Abstract
Carfilzomib, a selective proteasome inhibitor, is approved for use in relapsed and refractory multiple myeloma. Its link to left ventricular dysfunction is well established but little is known about its effects on the right ventricle. One of its rare complications is pulmonary hypertension, which at its extreme may result in right ventricular dysfunction. Here, we present a case of an elderly male veteran with multiple myeloma status post various failed therapies who developed acute dyspnea after four cycles of carfilzomib and subsequently found to have severe pulmonary hypertension with resultant acute right ventricular failure, which recovered after cessation of carfilzomib. This case highlights the need for careful cardiovascular surveillance while on carfilzomib and the importance of knowing even its rarest complications as these cardiotoxicities are reversible with discontinuation of the drug.
Collapse
Affiliation(s)
- Pei-Chun McGregor
- Harvard Medical School, Boston, MA, USA.,Department of Cardiology, Veterans Affairs Boston Healthcare System, West Roxbury, MA, USA
| | - Valia Boosalis
- Harvard Medical School, Boston, MA, USA.,Department of Hematology and Oncology, Veterans Affairs Boston Healthcare System, West Roxbury, MA, USA
| | - Jayashri Aragam
- Harvard Medical School, Boston, MA, USA.,Department of Cardiology, Veterans Affairs Boston Healthcare System, West Roxbury, MA, USA
| |
Collapse
|
21
|
Rago A, Siniscalchi A, Tordi A, Andrizzi C, Campagna S, Caravita di Toritto T. Pulmonary arterial hypertension in a patient with multiple myeloma during carfilzomib treatment: in search of better management. TUMORI JOURNAL 2021; 107:NP37-NP40. [PMID: 33525991 DOI: 10.1177/0300891621990427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Multiple myeloma is a plasma cell dyscrasia accounting for 1% of neoplastic diseases and is the second most common hematologic malignancy after lymphoma. Pulmonary arterial hypertension is characterized by increased blood pressure in the pulmonary circulation and the development of pulmonary vascular remodeling. Increased resistance in the pulmonary vessels strains the right ventricle, leading to right heart failure. AIM To report a case of a 65-year-old man who presented in 2017 with immunoglobulin G-kappa multiple myeloma characterized by pulmonary arterial hypertension during carfilzomib and dexamethasone treatment that resolved after stopping therapy. DISCUSSION Pulmonary arterial hypertension represents one of the main problems of carfilzomib and dexamethasone treatment, especially in patients with predisposing conditions such as hypertension. Therefore, monitoring and management of patients starting therapy with carfilzomib remains a critical issue. Although cardiac and vascular related adverse events were not frequent, a preemptive strategy prior to initiating carfilzomib appears advisable, particularly in patients at risk. CONCLUSION The mechanism responsible for cardiac and vascular events during carfilzomib therapy is unclear. This case report highlights that it is important to define at baseline the appropriate screening of patients undergoing carfilzomib therapy with specific monitoring and symptom management and that future large prospective controlled studies are needed to define the correct monitoring strategy in refractory and relapsed multiple myeloma and the potential mechanism of vascular events.
Collapse
Affiliation(s)
- Angela Rago
- Haematology Unit, ASL ROMA 1, Santo Spirito Hospital of Rome, Italy
| | | | - Attilio Tordi
- Haematology Unit, ASL ROMA 1, Santo Spirito Hospital of Rome, Italy
| | | | - Selenia Campagna
- Haematology Unit, ASL ROMA 1, Santo Spirito Hospital of Rome, Italy
| | | |
Collapse
|
22
|
Bishnoi R, Xie Z, Shah C, Bian J, Murthy HS, Wingard JR, Farhadfar N. Real-world experience of carfilzomib-associated cardiovascular adverse events: SEER-Medicare data set analysis. Cancer Med 2021; 10:70-78. [PMID: 33169938 PMCID: PMC7826471 DOI: 10.1002/cam4.3568] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/23/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023] Open
Abstract
Carfilzomib was approved for the treatment of multiple myeloma in 2012 and since then there have been concerns for cardiovascular toxicity from its use. With this study, we aim to further study the hazards and underlying risk factors for cardiovascular adverse events associated with carfilzomib. This study was conducted using Surveillance, Epidemiology, and End Results (SEER)-Medicare data set of multiple myeloma from 2001 to 2015. Data were analyzed for hazards ratio of cardiovascular adverse events between carfilzomib users and nonusers. We identified 7330 patients with multiple myeloma of whom 815 were carfilzomib users. Carfilzomib users had a statistically significant hazard ratio of 1.41 with p < 0.0001 for all cardiovascular adverse events as compared to nonusers. Carfilzomib use was significantly associated with increased risk of heart failure (HR 1.47, p = 0.0002), ischemic heart disease (HR 1.45, p = 0.0002), and hypertension (HR 3.33, p < 0.0001), whereas there was no association between carfilzomib use and cardiac conduction disorders (arrhythmia and heart blocks). Carfilzomib users were at higher risk of new-onset edema (HR 5.09, p < 0.0001), syncope (HR 4.27, p < 0.0001), dyspnea (HR 1.33, p < 0.0001), and chest pain (HR 1.18, p < 0.0001) as compared to carfilzomib nonusers. Age above 75 years, preexisting cardiovascular disease, obesity, and twice a week carfilzomib schedule were significant risk factors associated with cardiovascular adverse events in carfilzomib users. The median time of the onset for all cardiovascular adverse events was 3.1 months. This study has identified a significantly higher likelihood of cardiovascular adverse events in elderly Medicare patients receiving carfilzomib.
Collapse
Affiliation(s)
- Rohit Bishnoi
- Division of Hematology and OncologyDepartment of MedicineUniversity of FloridaGainesvilleFLUSA
| | - Zhigang Xie
- Department of Health Services Research, Management and PolicyCollege of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFLUSA
| | - Chintan Shah
- Division of Hematology and OncologyDepartment of MedicineUniversity of FloridaGainesvilleFLUSA
| | - Jiang Bian
- Department of Health Outcomes and BioinformaticsCollege of MedicineUniversity of FloridaGainesvilleFLUSA
| | - Hemant S. Murthy
- Division of Hematology/OncologyMayo Clinic Alix School of MedicineMayo Clinic FloridaJacksonvilleFLUSA
| | - John R. Wingard
- Bone Marrow Transplant ProgramUniversity of Florida Health Cancer CenterGainesvilleFLUSA
- Division of Hematology/OncologyUniversity of Florida College of MedicineGainesvilleFLUSA
| | - Nosha Farhadfar
- Division of Hematology/OncologyUniversity of Florida College of MedicineGainesvilleFLUSA
| |
Collapse
|
23
|
Carfilzomib-Induced Pulmonary Hypertension. Am J Ther 2020; 27:e693-e694. [PMID: 31724976 DOI: 10.1097/mjt.0000000000001051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
24
|
Bottinor W, Parikh A, Jahangir E. Emerging cancer therapies and cardiovascular risk. J Thromb Thrombolysis 2020; 51:837-845. [PMID: 32886244 DOI: 10.1007/s11239-020-02263-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The cardiovascular (CV) toxicity profiles of traditional cancer therapies such as anthracyclines and radiation therapy are familiar to many cardiologists. With the development and widespread use of additional cancer therapeutics, CV toxicities related to these agents are becoming more common. Cardiovascular specialists are often integrated into the care team for individuals with cancer and knowledge of the CV toxicities of cancer therapeutics has become essential. In this review, we provide a clinically focused summary of the current data regarding CV toxicities of common cancer therapies and identify potential management strategies for the CV specialist.
Collapse
Affiliation(s)
- Wendy Bottinor
- Division of Cardiovascular Medicine, Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Gateway bldg. 1200 E Marshall St, Richmond, VA, 23298, USA. .,Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Amar Parikh
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eiman Jahangir
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
25
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
26
|
Hypertension management in cardio-oncology. J Hum Hypertens 2020; 34:673-681. [PMID: 32747676 PMCID: PMC7398285 DOI: 10.1038/s41371-020-0391-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/08/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022]
Abstract
Cancer is one of the leading causes of death worldwide. During the last few decades prognosis has improved dramatically and patients are living longer and suffering long-term cardiovascular consequences of chemotherapeutic agents. Cardiovascular disease is a leading cause of morbidity and mortality in cancer survivors second only to recurrent cancer. In some types of cancer, cardiovascular disease is a more common cause of death than the cancer itself. This has led to a new sub-specialty of cardiology coined cardio-oncology to manage this specific population. Hypertension is one of the most common cardiovascular disease seen in this cohort. The aetiology of hypertension in cardio-oncology is complex and multifactorial based on the type of chemotherapy, type of malignancy and intrinsic patient factors such as age and pre-existing comorbidities. A variety of different oncological treatments have been implicated in causing hypertension. The effect can be transient whilst undergoing treatment or can be delayed occurring decades after treatment. A tailored management plan is recommended given the plethora of agents and their differing underlying mechanisms and speed of this mechanism in causing hypertension. Management by a multidisciplinary team consisting of oncology, general practice and cardiology is advised. There are currently no trials comparing antihypertensives in this specific cohort of patients. In the absence of evidence demonstrating otherwise, hypertension in cardio-oncology should be managed utilising the same treatment guidelines for the general population.
Collapse
|
27
|
Investigating the Vascular Toxicity Outcomes of the Irreversible Proteasome Inhibitor Carfilzomib. Int J Mol Sci 2020; 21:ijms21155185. [PMID: 32707866 PMCID: PMC7432349 DOI: 10.3390/ijms21155185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Carfilzomib’s (Cfz) adverse events in myeloma patients include cardiovascular toxicity. Since carfilzomib’s vascular effects are elusive, we investigated the vascular outcomes of carfilzomib and metformin (Met) coadministration. Methods: Mice received: (i) saline; (ii) Cfz; (iii) Met; (iv) Cfz+Met for two consecutive (acute) or six alternate days (subacute protocol). Leucocyte-derived reactive oxygen species (ROS) and serum NOx levels were determined and aortas underwent vascular and molecular analyses. Mechanistic experiments were recapitulated in aged mice who received similar treatment to young animals. Primary murine (prmVSMCs) and aged human aortic smooth muscle cells (HAoSMCs) underwent Cfz, Met and Cfz+Met treatment and viability, metabolic flux and p53-LC3-B expression were measured. Experiments were recapitulated in AngII, CoCl2 and high-glucose stimulated HAoSMCs. Results: Acutely, carfilzomib alone led to vascular hypo-contraction and increased ROS release. Subacutely, carfilzomib increased ROS release without vascular manifestations. Cfz+Met increased PGF2α-vasoconstriction and LC3-B-dependent autophagy in both young and aged mice. In vitro, Cfz+Met led to cytotoxicity and autophagy, while Met and Cfz+Met shifted cellular metabolism. Conclusion: Carfilzomib induces a transient vascular impairment and oxidative burst. Cfz+Met increased vascular contractility and synergistically induced autophagy in all settings. Therefore, carfilzomib cannot be accredited for a permanent vascular dysfunction, while Cfz+Met exert vasoprotective potency.
Collapse
|
28
|
Abstract
BACKGROUND Cardio-oncology aims to mitigate adverse cardiovascular manifestations in cancer survivors, but treatment-induced hypertension or aggravated hypertension has received less attention in these high cardiovascular risk patients. METHODS In this systematic review, we searched literature for contemporary data on the prevalence, pathophysiologic mechanisms, treatment implications and preventive strategies of hypertension in patients under antineoplastic therapy. RESULTS Several classes of antineoplastic drugs, including mainly vascular endothelial growth factor inhibitors, proteasome inhibitors, cisplatin derivatives, corticosteroids or radiation therapy were consistently associated with increased odds for new-onset hypertension or labile hypertensive status in previous controlled patients. Moreover, hypertension constitutes a major risk factor for chemotherapy-induced cardiotoxicity, which is the most serious cardiovascular adverse effect of antineoplastic therapy. Despite the heterogeneity of pooled studies, the pro-hypertensive profile of examined drug classes could be attributed to common structural and functional disorders. Importantly, certain antihypertensive drugs are considered to be more effective in the management of hypertension in this population and may partially attenuate indirect complications of cancer treatment, such as progressive development of cardiomyopathy and/or cardiovascular death. Nonpharmacological approaches to alleviate hypertension in cancer patients are also described, albeit adjudicated as less effective in general. CONCLUSION A growing body of evidence suggests that multiple antineoplastic agents increase the rate of progression of hypertension. Physicians need to balance the life-saving cancer treatment and the inflated risk of adverse cardiovascular events due to suboptimal management of hypertension in order to achieve improved clinical outcomes and sustained survival for their patients.
Collapse
|
29
|
Chavda SJ, Pocock R, Cheesman S, Lee KM, Dowling E, Marks DJB, Kyriakou C, Lee L, Sive J, Wechalekar A, Rabin N, Yong K, Popat R. Association of hypertension and cardiac events in patients with multiple myeloma receiving carfilzomib: practical management recommendations. Br J Haematol 2020; 190:e312-e316. [PMID: 32535901 DOI: 10.1111/bjh.16889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
| | - Rachael Pocock
- Department of Haematology, UCL Cancer Institute, London, UK
| | - Simon Cheesman
- Department of Haematology, University College London Hospitals NHS Foundation Trusts, London, UK
| | - Kyung-Min Lee
- HCA Healthcare, Department of Haematology, University College London NHS Foundation Trusts, London, UK
| | - Emma Dowling
- HCA Healthcare, Department of Haematology, University College London NHS Foundation Trusts, London, UK
| | - Daniel J B Marks
- Department of Acute Medicine, University College London Hospitals NHS Foundation Trusts, London, UK
| | - Charalampia Kyriakou
- Department of Haematology, University College London Hospitals NHS Foundation Trusts, London, UK
| | - Lydia Lee
- Department of Haematology, University College London Hospitals NHS Foundation Trusts, London, UK
| | - Jonathan Sive
- Department of Haematology, University College London Hospitals NHS Foundation Trusts, London, UK
| | - Ashutosh Wechalekar
- Department of Haematology, University College London Hospitals NHS Foundation Trusts, London, UK
| | - Neil Rabin
- Department of Haematology, University College London Hospitals NHS Foundation Trusts, London, UK
| | - Kwee Yong
- Department of Haematology, UCL Cancer Institute, London, UK
| | - Rakesh Popat
- Department of Haematology, University College London Hospitals NHS Foundation Trusts, London, UK
| |
Collapse
|
30
|
Abstract
Patients with cancer are at a higher risk of cardiovascular disease, which contributes to significant morbidity and mortality. The rapid progress in the field of oncological treatments has led to a steady increase in long-term cancer survivors. Care for cardiovascular complications is therefore becoming increasingly important. In addition, the establishment of new oncological therapies has resulted in the identification of previously unknown cardiovascular side effects. Oncocardiology aims to detect and treat cardiovascular diseases associated with cancer and cancer therapy. Continuous scientific, clinical, and structural developments are necessary as the basis for the best care of the growing number of affected patients. This review summarizes current developments in the field of oncocardiology with regard to advances in cancer therapy and challenges in clinical oncocardiology work. Cardiovascular side effects by targeted cancer therapies are characterized and recent advances in the field of cardiovascular diagnostics are outlined. Developments to better integrate oncocardiology into the medical care system and perspectives for modern, patient-oriented care are shown. In light of the coronavirus disease 2019 (COVID-19) pandemic, current challenges and opportunities are highlighted. The relevance of profitable further advances in oncocardiology including standardized guidelines and educational programs is delineated as a mandatory requirement for the successful development of oncocardiology.
Collapse
|
31
|
Wu P, Oren O, Gertz MA, Yang EH. Proteasome Inhibitor-Related Cardiotoxicity: Mechanisms, Diagnosis, and Management. Curr Oncol Rep 2020; 22:66. [DOI: 10.1007/s11912-020-00931-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
32
|
Sandy EB, Weerasinghe C, Terjanian T. Carfilzomib Induced Tumor Lysis Syndrome and Other Adverse Events. J Pharm Pract 2020; 33:213-216. [DOI: 10.1177/0897190018802129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the area of multiple myeloma (MM) therapy, proteasome inhibitors (PI) have emerged with promising responses both in the first- and second-line setting. Carfilzomib (CFZ) is a second-generation, selective PI approved in 2012 for the treatment of relapsed/refractory multiple myeloma (RRMM) in patients who received 2 prior therapies or have evidence of disease progression within 60 days of completion of last therapy. Its safety profile reported adverse events (AEs) ranging from drug-related AEs (nausea and vomiting), hematologic AEs (neutropenia and thrombocytopenia), and nonhematologic AEs (electrolyte imbalances). As CFZ use is gaining popularity, various hematological, renal, cardiovascular, pulmonary, and neurological toxicities have been reported. We are presenting this case to describe a rare occurrence of tumor lysis syndrome (TLS) with the use of this novel targeted therapy.
Collapse
Affiliation(s)
- El Bitar Sandy
- Department of Internal Medicine, Northwell Health Staten Island University Hospital, Staten Island, NY, USA
| | - Chanudi Weerasinghe
- Department of Hematology and Oncology, Northwell Health Staten Island University Hospital, Staten Island, NY, USA
| | - Terenig Terjanian
- Department of Hematology and Oncology, Northwell Health Staten Island University Hospital, Staten Island, NY, USA
| |
Collapse
|
33
|
Cornell RF, Ky B, Weiss BM, Dahm CN, Gupta DK, Du L, Carver JR, Cohen AD, Engelhardt BG, Garfall AL, Goodman SA, Harrell SL, Kassim AA, Jadhav T, Jagasia M, Moslehi J, O’Quinn R, Savona MR, Slosky D, Smith A, Stadtmauer EA, Vogl DT, Waxman A, Lenihan D. Prospective Study of Cardiac Events During Proteasome Inhibitor Therapy for Relapsed Multiple Myeloma. J Clin Oncol 2019; 37:1946-1955. [PMID: 31188726 PMCID: PMC9798911 DOI: 10.1200/jco.19.00231] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Cardiovascular adverse events (CVAEs) can occur during proteasome inhibitor (PI) therapy. We conducted a prospective, observational, multi-institutional study to define risk factors and outcomes in patients with multiple myeloma (MM) receiving PIs. PATIENTS AND METHODS Patients with relapsed MM initiating carfilzomib- or bortezomib-based therapy underwent baseline assessments and repeated assessments at regular intervals over 6 months, including cardiac biomarkers (troponin I or T, brain natriuretic peptide [BNP], and N-terminal proBNP), ECG, and echocardiography. Monitoring occurred over 18 months for development of CVAEs. RESULTS Of 95 patients enrolled, 65 received carfilzomib and 30 received bortezomib, with median 25 months of follow-up. Sixty-four CVAEs occurred, with 55% grade 3 or greater in severity. CVAEs occurred in 51% of patients treated with carfilzomib and 17% of those treated with bortezomib (P = .002). Median time to first CVAE from treatment start was 31 days, and 86% occurred within the first 3 months. Patients receiving carfilzomib-based therapy with a baseline elevated BNP level higher than 100 pg/mL or N-terminal proBNP level higher than 125 pg/mL had increased risk for CVAE (odds ratio, 10.8; P < .001). Elevated natriuretic peptides occurring mid-first cycle of treatment with carfilzomib were associated with a substantially higher risk of CVAEs (odds ratio, 36.0; P < .001). Patients who experienced a CVAE had inferior progression-free survival (log-rank P = .01) and overall survival (log-rank P < .001). PI therapy was safely resumed in 89% of patients, although 41% required chemotherapy modifications. CONCLUSION CVAEs are common during PI therapy for relapsed MM, especially with carfilzomib, particularly within the first 3 months of therapy. CVAEs were associated with worse overall outcomes, but usually, discontinuation of therapy was not required. Natriuretic peptides were highly predictive of CVAEs; however, validation of this finding is necessary before uniform incorporation into the routine management of patients receiving carfilzomib.
Collapse
Affiliation(s)
- Robert F. Cornell
- Vanderbilt University Medical Center, Nashville, TN,Robert F. Cornell, MS, MD, Division of Hematology and Oncology, Vanderbilt University Medical Center, 777 Preston Research Building, Nashville, TN 37232; Twitter: @myeloma_doc_com; e-mail:
| | - Bonnie Ky
- University of Pennsylvania, Philadelphia, PA
| | | | | | | | - Liping Du
- Vanderbilt University Medical Center, Nashville, TN
| | - Joseph R. Carver
- University of Pennsylvania, Philadelphia, PA,Abramson Cancer Center, Penn Medicine, Philadelphia, PA
| | - Adam D. Cohen
- Abramson Cancer Center, Penn Medicine, Philadelphia, PA
| | | | | | | | | | | | | | | | | | | | | | - David Slosky
- Vanderbilt University Medical Center, Nashville, TN
| | | | | | - Dan T. Vogl
- Abramson Cancer Center, Penn Medicine, Philadelphia, PA
| | - Adam Waxman
- Abramson Cancer Center, Penn Medicine, Philadelphia, PA
| | | |
Collapse
|
34
|
Ikoma T, Saotome M, Sano M, Suwa K, Naruse Y, Ohtani H, Urushida T, Nagata Y, Ono T, Maekawa Y. Cardiotoxicity of Carfilzomib in Two Japanese Patients with Relapsed Multiple Myeloma. Intern Med 2019; 58:1577-1581. [PMID: 30713316 PMCID: PMC6599935 DOI: 10.2169/internalmedicine.2194-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although multiple myeloma (MM) had been an incurable hematological malignancy with a poor prognosis, recent advances in novel anti-neoplastic agents, including carfilzomib (a proteasome inhibitor), have improved the prognosis. We herein report two cases of congestive heart failure in patients treated with carfilzomib. Although there are some reports on the cardiotoxicity of carfilzomib, to our knowledge, this is the first report on the cardiac involvement of carfilzomib in Japanese MM patients. We review the critical points from our two cases, with the aim of avoiding carfilzomib-associated heart failure in MM patients.
Collapse
Affiliation(s)
- Takenori Ikoma
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Masao Saotome
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Makoto Sano
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Kenichiro Suwa
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Yoshihisa Naruse
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Hayato Ohtani
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Tsuyoshi Urushida
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Yasuyuki Nagata
- Department of Hematology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Takaaki Ono
- Department of Hematology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Yuichiro Maekawa
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| |
Collapse
|
35
|
Abstract
Chemotherapy-associated myocardial toxicity is increasingly recognized with the expanding armamentarium of novel chemotherapeutic agents. The onset of cardiotoxicity during cancer therapy represents a major concern and often involves clinical uncertainties and complex therapeutic decisions, reflecting a compromise between potential benefits and harm. Furthermore, the improved cancer survival has led to cardiovascular complications becoming clinically relevant, potentially contributing to premature morbidity and mortality among cancer survivors. Specific higher-risk populations of cancer patients can benefit from prevention and screening measures during the course of cancer therapies. The pathobiology of chemotherapy-induced myocardial dysfunction is complex, and the individual patient risk for heart failure entails a multifactorial interaction between the selected chemotherapeutic regimen, traditional cardiovascular risk factors, and individual susceptibility. Treatment with several specific chemotherapeutic agents, including anthracyclines, proteasome inhibitors, epidermal growth factor receptor inhibitors, vascular endothelial growth factor inhibitors, and immune checkpoint inhibitors imparts increased risk for cardiotoxicity that results from specific therapy-related mechanisms. We review the pathophysiology, risk factors, and imaging considerations as well as patient surveillance, prevention, and treatment approaches to mitigate cardiotoxicity prior, during, and after chemotherapy. The complexity of decision-making in these patients requires viable discussion and partnership between cardiologists and oncologists aiming together to eradicate cancer while preventing cardiotoxic sequelae.
Collapse
Affiliation(s)
- Oren Caspi
- Department of Cardiology, Rambam Health Care Campus, Haifa, Israel
- The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Doron Aronson
- Department of Cardiology, Rambam Health Care Campus, Haifa, Israel
- The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
36
|
Ailawadhi S, DerSarkissian M, Duh MS, Lafeuille MH, Posner G, Ralston S, Zagadailov E, Ba-Mancini A, Rifkin R. Cost Offsets in the Treatment Journeys of Patients With Relapsed/Refractory Multiple Myeloma. Clin Ther 2019; 41:477-493.e7. [DOI: 10.1016/j.clinthera.2019.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/12/2018] [Accepted: 01/14/2019] [Indexed: 12/22/2022]
|
37
|
Late Presentation of Carfilzomib Associated Thrombotic Microangiopathy. AMERICAN JOURNAL OF MEDICAL CASE REPORTS 2019; 7:240-243. [PMID: 31457071 PMCID: PMC6711615 DOI: 10.12691/ajmcr-7-10-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Multiple Myeloma (MM) is a plasma cell disorder characterized by abnormal proliferation of plasma cells resulting in overproduction of paraprotein. Proteasome inhibitors (PI) have been a corner stone for the treatment of MM. Thrombotic Microangiopathy (TMA) is a recent hematological adverse event that has newly been recognized in multiple PI. TMA leads to end-organ damage and infarction by microthromobi. TMA pathophysiology is not well understood and has multiple etiologies. We present a case of PI-induced TMA, along with literature review of cases diagnosed from 2008-2018. Unique to our case is the onset of presentation, more than 24 months after initiating carfilzomib. Our case highlights the need for vigilant monitoring and the importance of clinical suspicion in patients at risk for TMA.
Collapse
|
38
|
Serra W, Fantin A, Longo C, Rabia G, De Rosa F, Plenteda C, Re F, Crisafulli E, Chetta A. Carfilzomib: A Tale of a Heartbreaking Moment: Case Report and Concise Review of the Literature. Cardiovasc Hematol Disord Drug Targets 2018; 19:253-258. [PMID: 30516116 DOI: 10.2174/1871529x19666181205100705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 11/12/2018] [Accepted: 11/28/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Carfilzomib, a proteasome inhibitor, known as a therapeutical option for people who have already received one or more previous treatments for multiple myeloma, has well known cardiac and systemic adverse effects. OBJECTIVE There is evidence supporting that adverse effects are dose dependent, yet there is no known patient phenotype characterized by worse associated consequences, nor are there widely accepted monitoring protocols. RESULTS In this article we describe two patients with cardiovascular adverse events related to carfilzomib treatment and their clinical course. Our goal was to present two cases of daily practice, which highlighted the complexity of their management and led to underline how baseline evaluation and close follow-up with echocardiography and cardiac biomarkers, including natriuretic peptides, remain an important tool for the cardiotoxicity surveillance. CONCLUSION These reflections should lead to further studies in order to identify high risk patients for cardiovascular adverse event and clarify the real incidence of cardiotoxicity of this drug and adequate follow-up timing. Finally further research is needed to evaluate strategies for prevention and attenuation of cardiovascular complications of cancer therapy.
Collapse
Affiliation(s)
- W Serra
- Cardiology Unit, Department of Surgery, University of Parma, Parma, Italy
| | - A Fantin
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - C Longo
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - G Rabia
- Cardiology Unit, Department of Surgery, University of Parma, Parma, Italy
| | - F De Rosa
- Cardiology Unit, Department of Surgery, University of Parma, Parma, Italy
| | - C Plenteda
- Emathologic Department, University of Parma, Parma, Italy
| | - F Re
- Emathologic Department, University of Parma, Parma, Italy
| | - E Crisafulli
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - A Chetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
39
|
Heckmann MB, Doroudgar S, Katus HA, Lehmann LH. Cardiovascular adverse events in multiple myeloma patients. J Thorac Dis 2018; 10:S4296-S4305. [PMID: 30701098 PMCID: PMC6328391 DOI: 10.21037/jtd.2018.09.87] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/18/2018] [Indexed: 12/15/2022]
Abstract
Multiple myeloma is a malignant disease, caused by an uncontrolled clonal proliferation of a specific group of white blood cells, the plasma cells. Clinical manifestations include bone pain due to osteolysis, hypercalcemia, anemia, and renal insufficiency. Proteasome inhibitors have substantially improved survival of patients suffering from multiple myeloma, providing an efficient treatment option mainly for relapsed and refractory multiple myeloma. Although constituting one substance class, bortezomib, carfilzomib, and ixazomib differ greatly regarding their non-hematologic side effects. This article reviews the clinical and preclinical data on approved proteasome inhibitors in an attempt to decipher the underlying pathomechanisms related to cardiovascular adverse events seen in clinical trials.
Collapse
Affiliation(s)
- Markus B. Heckmann
- Department of Cardiology, Angiology, and Pneumology, Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Shirin Doroudgar
- Department of Cardiology, Angiology, and Pneumology, Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Hugo A. Katus
- Department of Cardiology, Angiology, and Pneumology, Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Lorenz H. Lehmann
- Department of Cardiology, Angiology, and Pneumology, Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|
40
|
Strifler S, Knop S. The role of carfilzomib in treatment of newly diagnosed multiple myeloma. Future Oncol 2018; 14:3123-3134. [DOI: 10.2217/fon-2018-0040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite improvement of prognosis since approval of proteasome inhibitors and immunomodulatory drugs, myeloma remains largely incurable. The outcome of first-line treatment is known to be crucial for survival and, therefore, implementation of novel strategies remains one of the key aims of clinical myeloma research. Since approval of carfilzomib for relapsed and refractory multiple myeloma, a new therapeutic option with a favorable safety profile regarding neuropathy is available. Regarding its superior response rates and progression-free survival (PFS) when combined with other agents in heavily pretreated patients, the compound rapidly became a matter of great interest in search for first-line treatment. With an ORR up to 98% and promising PFS data, it might become an important partner in treatment of newly diagnosed myeloma.
Collapse
Affiliation(s)
- Susanne Strifler
- Department of Internal Medicine II, Division of Hematology & Medical Oncology, Wuerzburg University Medical Center, Wuerzburg, Germany
| | - Stefan Knop
- Department of Internal Medicine II, Division of Hematology & Medical Oncology, Wuerzburg University Medical Center, Wuerzburg, Germany
| |
Collapse
|
41
|
Imam F, Al-Harbi NO, Al-Harbi MM, Qamar W, Aljerian K, Belali OM, Alsanea S, Alanazi AZ, Alhazzani K. Apremilast ameliorates carfilzomib-induced pulmonary inflammation and vascular injuries. Int Immunopharmacol 2018; 66:260-266. [PMID: 30500623 DOI: 10.1016/j.intimp.2018.11.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/29/2018] [Accepted: 11/15/2018] [Indexed: 01/29/2023]
Abstract
Acute lung injury (ALI) due to chemotherapy occurs frequently. It presents a challenge for clinicians managing therapies for different types of cancers. Carfilzomib (Kyprolis™) is a new proteasome inhibitor that shows promise for the treatment of relapsing multiple myeloma. However, several cases of severe ALI have raised concern about the use of carfilzomib against relapsed multiple myelomas. To improve the efficacy of carfilzomib, a new anti-inflammatory drug for psoriasis treatment, apremilast (Otezla™) was investigated for its protective effects against carfilzomib-induced ALI in rats. RT-PCR analyses revealed that carfilzomib administration in rats markedly increased the levels of tumor necrosis factor-alpha and nuclear factor-kappa B and myeloperoxidase activity with a concomitant increase in lipid peroxidation. The anti-inflammatory cytokine, interleukin-10, was downregulated following carfilzomib administration. Reduction in glutathione levels indicated diminished cellular antioxidant defenses in response to carfilzomib-induced ALI. ALI was confirmed by histopathological observations in lung tissue slices. Apremilast administration reduced lung inflammation in terms of reduction in myeloperoxidase activity and levels of tumor necrosis factor-alpha and alveolar infiltrating cells. Apremilast reversed all observed toxic effects of carfilzomib and prevented ALI in rats.
Collapse
Affiliation(s)
- Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Naif O Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohammed M Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Wajhul Qamar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Central Laboratory, Research Center, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Khaldoon Aljerian
- King Khalid University Hospital, College of Medicine, King Saud University, Forensic Medicine and Toxicology Unit, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Osamah Mohammed Belali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Sary Alsanea
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ahmed Z Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
42
|
Molecular mechanisms of carfilzomib-induced cardiotoxicity in mice and the emerging cardioprotective role of metformin. Blood 2018; 133:710-723. [PMID: 30482794 DOI: 10.1182/blood-2018-06-858415] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 11/09/2018] [Indexed: 12/21/2022] Open
Abstract
Carfilzomib (Cfz), an irreversible proteasome inhibitor licensed for relapsed/refractory myeloma, is associated with cardiotoxicity in humans. We sought to establish the optimal protocol of Cfz-induced cardiac dysfunction, to investigate the underlying molecular-signaling and, based on the findings, to evaluate the cardioprotective potency of metformin (Met). Mice were randomized into protocols 1 and 2 (control and Cfz for 1 and 2 consecutive days, respectively); protocols 3 and 4 (control and alternate doses of Cfz for 6 and 14 days, respectively); protocols 5A and 5B (control and Cfz, intermittent doses on days 0, 1 [5A] and 0, 1, 7, and 8 [5B] for 13 days); protocols 6A and 6B (pharmacological intervention; control, Cfz, Cfz+Met and Met for 2 and 6 days, respectively); and protocol 7 (bortezomib). Cfz was administered at 8 mg/kg (IP) and Met at 140 mg/kg (per os). Cfz resulted in significant reduction of proteasomal activity in heart and peripheral blood mononuclear cells in all protocols except protocols 5A and 5B. Echocardiography demonstrated that Cfz led to a significant fractional shortening (FS) depression in protocols 2 and 3, a borderline dysfunction in protocols 1 and 4, and had no detrimental effect on protocols 5A and 5B. Molecular analysis revealed that Cfz inhibited AMPKα/mTORC1 pathways derived from increased PP2A activity in protocol 2, whereas it additionally inhibited phosphatidylinositol 3-kinase/Akt/endothelial nitric oxide synthase pathway in protocol 3. Coadministration of Met prevented Cfz-induced FS reduction and restored AMPKα phosphorylation and autophagic signaling. Conclusively, Cfz decreased left ventricular function through increased PP2A activity and inhibition of AMPKα and its downstream autophagic targets, whereas Met represents a novel promising intervention against Cfz-induced cardiotoxicity.
Collapse
|
43
|
Kharel P, Uprety D, Chandra AB, Hu Y, Belur AA, Dhakal A. Bortezomib-Induced Pulmonary Toxicity: A Case Report and Review of Literature. Case Rep Med 2018; 2018:2913124. [PMID: 30595697 PMCID: PMC6286773 DOI: 10.1155/2018/2913124] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 11/04/2018] [Indexed: 01/10/2023] Open
Abstract
Bortezomib, a proteasome inhibitor, is an established therapy against multiple myeloma. Bortezomib-induced lung injury, although not appreciated during the introductory time of the medication, has now been highlighted in multiple case reports. The objective of this study is to report a case of bortezomib-induced lung injury, review current literature, and perform exploratory analysis.
Collapse
Affiliation(s)
- Prakash Kharel
- Department of Hospital Medicine, Geisinger Medical Center, 100 N Academy Ave., Danville, PA 17821, USA
| | - Deekchha Uprety
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Yirui Hu
- Center for Health Research, Geisinger Medical Center, 100 N Academy Ave., Danville, PA 17821, USA
| | | | - Ajay Dhakal
- Department of Medicine, Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA
| |
Collapse
|
44
|
Gavazzoni M, Lombardi CM, Vizzardi E, Gorga E, Sciatti E, Rossi L, Belotti A, Rossi G, Metra M, Raddino R. Irreversible proteasome inhibition with carfilzomib as first line therapy in patients with newly diagnosed multiple myeloma: Early in vivo cardiovascular effects. Eur J Pharmacol 2018; 838:85-90. [DOI: 10.1016/j.ejphar.2018.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 01/22/2023]
|
45
|
Chari A, Stewart AK, Russell SD, Moreau P, Herrmann J, Banchs J, Hajek R, Groarke J, Lyon AR, Batty GN, Ro S, Huang M, Iskander KS, Lenihan D. Analysis of carfilzomib cardiovascular safety profile across relapsed and/or refractory multiple myeloma clinical trials. Blood Adv 2018; 2:1633-1644. [PMID: 29991494 PMCID: PMC6039655 DOI: 10.1182/bloodadvances.2017015545] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 05/23/2018] [Indexed: 01/10/2023] Open
Abstract
Carfilzomib is a selective proteasome inhibitor approved for the treatment of relapsed and/or refractory multiple myeloma (RRMM). It has significantly improved outcomes, including overall survival (OS), and shown superiority vs standard treatment with lenalidomide plus dexamethasone and bortezomib plus dexamethasone. The incidence rate of cardiovascular (CV) events with carfilzomib treatment has varied across trials. This analysis evaluated phase 1-3 trials with >2000 RRMM patients exposed to carfilzomib to describe the incidence of CV adverse events (AEs). In addition, the individual CV safety data of >1000 patients enrolled in the carfilzomib arm of phase 3 studies were compared with the control arms to assess the benefit-risk profile of carfilzomib. Pooling data across carfilzomib trials, the CV AEs (grade ≥3) noted included hypertension (5.9%), dyspnea (4.5%), and cardiac failure (4.4%). Although patients receiving carfilzomib had a numeric increase in the rates of any-grade and grade ≥3 cardiac failure, dyspnea, and hypertension, the frequency of discontinuation or death due to these cardiac events was low and comparable between the carfilzomib and control arms. Serial echocardiography in a blinded cardiac substudy showed no objective evidence of cardiac dysfunction in the carfilzomib and control arms. Moreover, carfilzomib had no significant effect on cardiac repolarization. Our results, including the OS benefit, showed that the benefit of carfilzomib treatment in terms of reducing progression or death outweighed the risk for developing cardiac failure or hypertension in most patients. Appropriate carfilzomib administration and risk factor management are recommended for elderly patients and patients with underlying risk factors.
Collapse
Affiliation(s)
- Ajai Chari
- Hematology/Oncology, Mount Sinai School of Medicine, New York, NY
| | - A Keith Stewart
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ
| | - Stuart D Russell
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Philippe Moreau
- Department of Hematology, University Hospital Hotel-Dieu, Nantes, France
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine, Phoenix, AZ
| | - Jose Banchs
- Department of Cardiology, Division of Internal Medicine, MD Anderson Cancer Center, Houston, TX
| | - Roman Hajek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava and University Hospital Ostrava, Ostrava, Czech Republic
| | - John Groarke
- Department of Cardiology, Division of Internal Medicine, Partners/Brigham and Women's Hospital, Boston, MA
| | - Alexander R Lyon
- Department of Cardiology, Imperial College London, London, United Kingdom
- Cardiology, Royal Brompton Hospital, London, United Kingdom
| | | | | | | | | | - Daniel Lenihan
- Cardiovascular Division, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
46
|
Gavazzoni M, Vizzardi E, Gorga E, Bonadei I, Rossi L, Belotti A, Rossi G, Ribolla R, Metra M, Raddino R. Mechanism of cardiovascular toxicity by proteasome inhibitors: New paradigm derived from clinical and pre-clinical evidence. Eur J Pharmacol 2018; 828:80-88. [DOI: 10.1016/j.ejphar.2018.03.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/11/2018] [Accepted: 03/14/2018] [Indexed: 01/08/2023]
|
47
|
|
48
|
Małyszko J, Małyszko M, Kozlowski L, Kozlowska K, Małyszko J. Hypertension in malignancy-an underappreciated problem. Oncotarget 2018; 9:20855-20871. [PMID: 29755695 PMCID: PMC5945504 DOI: 10.18632/oncotarget.25024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 03/19/2018] [Indexed: 12/25/2022] Open
Abstract
Hypertension is one of the most common comorbidities in cancer patients with malignancy, in particular, in the elderly. On the other hand, hypertension is a long-term consequence of antineoplastic treatment, including both chemotherapy and targeted agents. Several chemotherapeutics and targeted drugs may be responsible for development or worsening of the hypertension. The most common side effect of anti-VEGF (vascular endothelial growth factor) treatment is hypertension. However, pathogenesis of hypertension in patients receiving this therapy appears to be associated with multiple pathways and is not yet fully understood. Development of hypertension was associated with improved antitumor efficacy in patients treated with anti-antiangiogenic drugs in some but not in all studies. Drugs used commonly as adjuvants such as steroids, erythropoietin stimulating agents etc, may also cause rise in blood pressure or exacerbate preexisiting hypertension. Hypotensive therapy is crucial to manage hypertension during certain antineoplastic treatment. The choice and dose of antihypertensive drugs depend upon the presence of organ dysfunction, comorbidities, and/or adverse effects. In addition, severity of the hypertension and the urgency of blood pressure control should also be taken into consideration. As there are no specific guidelines on the hypertension treatment in cancer patients we should follow the available guidelines to obtain the best possible outcomes and pay the attention to the individualization of the therapy according to the actual situation.
Collapse
Affiliation(s)
- Jolanta Małyszko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University in Bialystok, Bialystok, Poland
- Department of Nephrology, Dialysis and Internal Medicine, Warsaw Medical University, Warsaw, Poland
| | - Maciej Małyszko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University in Bialystok, Bialystok, Poland
| | - Leszek Kozlowski
- Department of Oncological Surgery, Regional Cancer Center, Bialystok, Poland
| | - Klaudia Kozlowska
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University in Bialystok, Bialystok, Poland
| | - Jacek Małyszko
- 1st Department of Nephrology and Transplantology with Dialysis Unit, Medical University in Bialystok, Bialystok, Poland
| |
Collapse
|
49
|
Iannaccone A, Bruno G, Ravera A, Gay F, Salvini M, Bringhen S, Sabia L, Avenatti E, Veglio F, Milan A. Evaluation of Cardiovascular Toxicity Associated with Treatments Containing Proteasome Inhibitors in Multiple Myeloma Therapy. High Blood Press Cardiovasc Prev 2018; 25:209-218. [PMID: 29582365 DOI: 10.1007/s40292-018-0256-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/19/2018] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Recently new treatment options have substantially increased survival for patients with relapsed and/or refractory multiple myeloma (RRMM). Among these, proteasome inhibitors (PI), such as bortezomib and carfilzomib, offer high response rate and prolonged survival. These agents are generally well tolerated but demonstrated a significant cardiovascular toxicity, mostly for regimen containing carfilzomib. AIM To assess the cardiovascular damage in patients treated with PI for RRMM. METHODS 28 consecutive subjects treated with PI for RRMM were evaluated and compared with a population of 22 control (Con) subjects, matched for age, sex and mean 24 h blood pressure (24hMBP). All individuals underwent trans-thoracic echocardiography, ambulatory blood pressure monitoring and pulse wave velocity (PVW) study. RESULTS PI patients did not have significant differences in blood pressure load and PWV compared to controls. Among echocardiographic parameters, the global longitudinal strain (GLS) was significantly decreased in PI subjects (p = 0.02). The GLS was significantly lower also considering only patients treated with carfilzomib. Moreover, among carfilzomib patients, we found increase values of left ventricle mass indexed by BSA (LVMi; p = 0.047). After correction for age, sex, BSA, 24hMBP and morphological and functional parameters of LV, treatment with PI and carfilzomib were significantly associated with GLS (p = 0.01; p = 0.036, respectively). CONCLUSIONS PI treatment is associated with subclinical LV dysfunction in patients with RRMM compared to controls, as demonstrated by lower GLS values. These results are confirmed also considering patients treated with carfilzomib. Moreover, in this subgroup of patients, the LVMi is also increased, suggesting higher cardiotoxicity with this treatment.
Collapse
Affiliation(s)
- Andrea Iannaccone
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy.
| | - G Bruno
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - A Ravera
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - F Gay
- Myeloma Unit, Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University Hospital 'Città della Salute e della Scienza di Torino', University of Torino, Turin, Italy
| | - M Salvini
- Myeloma Unit, Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University Hospital 'Città della Salute e della Scienza di Torino', University of Torino, Turin, Italy
| | - S Bringhen
- Myeloma Unit, Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University Hospital 'Città della Salute e della Scienza di Torino', University of Torino, Turin, Italy
| | - L Sabia
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - E Avenatti
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - F Veglio
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - A Milan
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| |
Collapse
|
50
|
McGee M, Whitehead N, Martin J, Collins N. Drug-associated pulmonary arterial hypertension. Clin Toxicol (Phila) 2018; 56:801-809. [PMID: 29508628 DOI: 10.1080/15563650.2018.1447119] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
INTRODUCTION While pulmonary arterial hypertension remains an uncommon diagnosis, various therapeutic agents are recognized as important associations. These agents are typically categorized into "definite", "likely", "possible", or "unlikely" to cause pulmonary arterial hypertension, based on the strength of evidence. OBJECTIVE This review will focus on those therapeutic agents where there is sufficient literature to adequately comment on the role of the agent in the pathogenesis of pulmonary arterial hypertension. METHODS A systematic search was conducted using PubMed covering the period September 1970- 2017. The search term utilized was "drug induced pulmonary hypertension". This resulted in the identification of 853 peer-reviewed articles including case reports. Each paper was then reviewed by the authors for its relevance. The majority of these papers (599) were excluded as they related to systemic hypertension, chronic obstructive pulmonary disease, human immunodeficiency virus, pulmonary fibrosis, alternate differential diagnosis, treatment, basic science, adverse effects of treatment, and pulmonary hypertension secondary to pulmonary embolism. Agents affecting serotonin metabolism (and related anorexigens): Anorexigens, such as aminorex, fenfluramine, benfluorex, phenylpropanolamine, and dexfenfluramine were the first class of medications recognized to cause pulmonary arterial hypertension. Although most of these medications have now been withdrawn worldwide, they remain important not only from a historical perspective, but because their impact on serotonin metabolism remains relevant. Selective serotonin reuptake inhibitors, tryptophan, and lithium, which affect serotonin metabolism, have also been implicated in the development of pulmonary arterial hypertension. Interferon and related medications: Interferon alfa and sofosbuvir have been linked to the development of pulmonary arterial hypertension in patients with other risk factors, such as human immunodeficiency virus co-infection. Antiviral therapies: Sofosbuvir has been associated with two cases of pulmonary artery hypertension in patients with multiple risk factors for its development. Its role in pathogenesis remains unclear. Small molecule tyrosine kinase inhibitors: Small molecule tyrosine kinase inhibitors represent a relatively new class of medications. Of these dasatinib has the strongest evidence in drug-induced pulmonary arterial hypertension, considered a recognized cause. Nilotinib, ponatinib, carfilzomib, and ruxolitinib are newer agents, which paradoxically have been linked to both cause and treatment for pulmonary arterial hypertension. Monoclonal antibodies and immune regulating medications: Several case reports have linked some monoclonal antibodies and immune modulating therapies to pulmonary arterial hypertension. There are no large series documenting an increased prevalence of pulmonary arterial hypertension complicating these agents; nonetheless, trastuzumab emtansine, rituximab, bevacizumab, cyclosporine, and leflunomide have all been implicated in case reports. Opioids and substances of abuse: Buprenorphine and cocaine have been identified as potential causes of pulmonary arterial hypertension. The mechanism by which this occurs is unclear. Tramadol has been demonstrated to cause severe, transient, and reversible pulmonary hypertension. Chemotherapeutic agents: Alkylating and alkylating-like agents, such as bleomycin, cyclophosphamide, and mitomycin have increased the risk of pulmonary veno-occlusive disease, which may be clinically indistinct from pulmonary arterial hypertension. Thalidomide and paclitaxel have also been implicated as potential causes. Miscellaneous medications: Protamine appears to be able to cause acute, reversible pulmonary hypertension when bound to heparin. Amiodarone is also capable of causing pulmonary hypertension by way of recognized side effects. CONCLUSIONS Pulmonary arterial hypertension remains a rare diagnosis, with drug-induced causes even more uncommon, accounting for only 10.5% of cases in large registry series. Despite several agents being implicated in the development of PAH, the supportive evidence is typically limited, based on case series and observational data. Furthermore, even in the drugs with relatively strong associations, factors that predispose an individual to PAH have yet to be elucidated.
Collapse
Affiliation(s)
- Michael McGee
- a Cardiovascular Department , John Hunter Hospital , Newcastle , Australia
| | - Nicholas Whitehead
- a Cardiovascular Department , John Hunter Hospital , Newcastle , Australia
| | - Jennifer Martin
- b Clinical Pharmacology, School of Medicine and Public Health , University of Newcastle , Newcastle , Australia
| | - Nicholas Collins
- a Cardiovascular Department , John Hunter Hospital , Newcastle , Australia
| |
Collapse
|