1
|
Gutierrez C, Rajendram P, Idowu O. Novel Cancer Therapeutics: Perioperative Implications and Challenges. Anesth Analg 2024:00000539-990000000-01013. [PMID: 39453847 DOI: 10.1213/ane.0000000000007210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Since the introduction of immunotherapy and targeted therapies, patients not only have adequate tumoral response to these treatments, but their quality of life has improved due to milder toxicities. However, due to their wide mechanisms of action, the toxicity profile for these therapies is broad, can have an insidious onset, and their recognition can be challenging. Rarely, some of these toxicities can cause significant morbidity if not diagnosed early and lead to intensive care unit (ICU) admission and death. Anesthesiologists are likely to encounter not only a wide spectrum of these toxicities but also a wide range of severity. In some cases, they could be the first to make the diagnosis and therefore need to be prepared to rapidly assess, establish differentials, perform a diagnostic workup, and evaluate the impact the toxicity could have on the patients' care during the perioperative period. In this article, we set to review toxicities of novel cancer therapies such as checkpoint inhibitors and targeted therapies, that could present in the perioperative setting. This article will help as a guide for anesthesiologists to recognize their clinical presentation, the approach to their diagnosis, and their impact on patient care.
Collapse
Affiliation(s)
- Cristina Gutierrez
- From the Department of Critical Care Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Prabalini Rajendram
- Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Olakunle Idowu
- Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
2
|
Zakaria ZZ, Suleiman M, Benslimane FM, Al-Badr M, Sivaraman S, Korashy HM, Ahmad F, Uddin S, Mraiche F, Yalcin HC. Imatinib‑ and ponatinib‑mediated cardiotoxicity in zebrafish embryos and H9c2 cardiomyoblasts. Mol Med Rep 2024; 30:187. [PMID: 39219269 PMCID: PMC11350628 DOI: 10.3892/mmr.2024.13311] [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: 08/04/2023] [Accepted: 04/26/2024] [Indexed: 09/04/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) offer targeted therapy for cancers but can cause severe cardiotoxicities. Determining their dose‑dependent impact on cardiac function is required to optimize therapy and minimize adverse effects. The dose‑dependent cardiotoxic effects of two TKIs, imatinib and ponatinib, were assessed in vitro using H9c2 cardiomyoblasts and in vivo using zebrafish embryos. In vitro, H9c2 cardiomyocyte viability, apoptosis, size, and surface area were evaluated to assess the impact on cellular health. In vivo, zebrafish embryos were analyzed for heart rate, blood flow velocity, and morphological malformations to determine functional and structural changes. Additionally, reverse transcription‑quantitative PCR (RT‑qPCR) was employed to measure the gene expression of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), established markers of cardiac injury. This comprehensive approach, utilizing both in vitro and in vivo models alongside functional and molecular analyses, provides a robust assessment of the potential cardiotoxic effects. TKI exposure decreased viability and surface area in H9c2 cells in a dose‑dependent manner. Similarly, zebrafish embryos exposed to TKIs exhibited dose‑dependent heart malformation. Both TKIs upregulated ANP and BNP expression, indicating heart injury. The present study demonstrated dose‑dependent cardiotoxic effects of imatinib and ponatinib in H9c2 cells and zebrafish models. These findings emphasize the importance of tailoring TKI dosage to minimize cardiac risks while maintaining therapeutic efficacy. Future research should explore the underlying mechanisms and potential mitigation strategies of TKI‑induced cardiotoxicities.
Collapse
Affiliation(s)
- Zain Z. Zakaria
- Vice President of Health and Medical Sciences Office, QU Health, Qatar University, Doha 2713, Qatar
| | - Muna Suleiman
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | | | - Mashael Al-Badr
- Department of Biology, College of Art and Science, Qatar University, Doha 2713, Qatar
- National Reference Laboratory, Ministry of Public Health, Doha 7744, Qatar
| | - Siveen Sivaraman
- Translational Research Institute, Hamad Medical Corporation, Doha 3050, Qatar
| | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Fareed Ahmad
- Translational Research Institute and Dermatology Institute, Hamad Medical Corporation, Doha 3050, Qatar
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Hamad Medical Corporation, Doha 3050, Qatar
- Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar
| | - Fatima Mraiche
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | | |
Collapse
|
3
|
Lin X, Ma X, Zhao S, Yao J, Han L, Jing Y, Xue X. Cardiovascular toxicity in antitumor therapy: biological and therapeutic insights. Trends Cancer 2024; 10:920-934. [PMID: 39097431 DOI: 10.1016/j.trecan.2024.07.004] [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: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/12/2024] [Indexed: 08/05/2024]
Abstract
The evolution of antitumor therapies has significantly improved cancer prognosis but has concurrently resulted in cardiovascular toxicities. Understanding the biological mechanisms behind these toxicities is crucial for effective management. Immunotherapy-related cardiovascular toxicities are primarily mediated by immune cells and secreted cytokines. Chemotherapy may cause cardiovascular damage through autophagy disruption and mitochondrial dysfunction. Targeted therapies can induce toxicity through endothelin-1 (ET-1) production and cardiac signaling disruption. Radiotherapy may lead to cardiomyopathy and myocardial fibrosis by affecting endothelial cells, triggering inflammatory responses and accelerating atherosclerosis. This review provides insights into these mechanisms and strategies, aiming to enhance the clinical prevention and treatment of cardiovascular toxicities.
Collapse
Affiliation(s)
- Xuwen Lin
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xidong Ma
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Sheng Zhao
- Department of Cardiology, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Yao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Leng Han
- Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Ying Jing
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou, China.
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
4
|
Shankar G, Jain A, Gupta A, Jandial A, Varma N, Jain A, Prakash G, Khadwal A, Malhotra P. Imatinib Mesylate Causes Potential Cardiac Dysfunction in Patients With Chronic Myeloid Leukemia: the Results of a Case-control Study. Indian J Hematol Blood Transfus 2024; 40:558-563. [PMID: 39469155 PMCID: PMC11513061 DOI: 10.1007/s12288-024-01797-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/22/2024] [Indexed: 10/30/2024] Open
Abstract
Unlike second-generation tyrosine kinase inhibitors, effects of imatinib on the cardiovascular (CV) system are debatable. The current case-control study aimed to evaluate the CV effects of imatinib in patients with chronic myeloid leukemia (CML) using non-invasive 2D-echocardiography testing. Patients with CML ≥ 13 years attending the adult haematology clinic of a tertiary care hospital in north India were prospectively enrolled over 1.5 years. The study population (n = 110) consisted of 35 newly diagnosed patients (treatment-naïve group) and 75 patients under imatinib therapy ≥ 1 year (treated group). All the eligible patients were subjected to 2D-echocardiography to calculate pulmonary artery systolic pressure (PASP), left ventricular ejection fraction (LVEF) and deceleration time (DT). These parameters were compared between the two groups. P-value < 0.05 was considered statistically significant. The median age of study population was 40 years (range, 13-73) and M:F ratio was 1.14:1. Both the groups had similar demographics at the diagnosis including CV risk factors. The median PASP of the treated group was 2 mm Hg higher than the treatment-naïve group (25 vs 23 mm Hg, p-value = 0.919). The median LVEF of the treated group was 3.2% lower than the treatment-naïve group (58.5% vs 61.72%, p-value = 0.577). The median DT of the treated group was 7 ms shorter than treatment-naïve group (211 vs 204 ms, p-value = 0.411). Imatinib causes potential cardiac dysfunction in patients with CML. Large scale prospective follow-up trials in the same cohort of patients are needed to validate the findings of our study.
Collapse
Affiliation(s)
- Gowri Shankar
- Department of Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ankur Jain
- Department of Haematology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, 110029 India
| | - Ankur Gupta
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aditya Jandial
- Department of Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Neelam Varma
- Department of Haematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arihant Jain
- Department of Clinical Haematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gaurav Prakash
- Department of Clinical Haematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Khadwal
- Department of Clinical Haematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Department of Clinical Haematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
5
|
Mattii L, Moscato S, Ippolito C, Polizzi E, Novo G, Zucchi R, De Caterina R, Ghelardoni S, Madonna R. Empagliflozin mitigates ponatinib-induced cardiotoxicity by restoring the connexin 43-autophagy pathway. Biomed Pharmacother 2024; 178:117278. [PMID: 39116784 DOI: 10.1016/j.biopha.2024.117278] [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: 05/22/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Empagliflozin (EMPA), a selective sodium-glucose cotransporter type 2 (SGLT2) inhibitor, has been shown to reduce major adverse cardiovascular events in patients with heart failure of different etiologies, although the underlying mechanism still remains unclear. Ponatinib (PON) is a multi-tyrosine kinase inhibitor successfully used against myeloid leukemia and other human malignancies, but its cardiotoxicity remains worrisome. Cardiac connexins (Cxs) are both substrates and regulators of autophagy and responsible for proper heart function. Alteration in connexin expression and localization have been described in patients with heart failure. AIMS To assess whether EMPA can mitigate PON-induced cardiac dysfunction by restoring the connexin 43-autophagy pathway. METHODS AND RESULTS Male C57BL/6 mice, randomized into four treatment groups (CNTRL, PON, EMPA, PON+EMPA) for 28 days, showed increased autophagy, decreased Cx43 expression as well as Cx43 lateralization, and attenuated systo-diastolic cardiac dysfunction after treatment with EMPA and PON compared with PON alone. Compared with CNTRL (DMSO), cardiomyocyte-differentiated H9c2 (dH9c2) cells treated with PON showed significantly reduced cell viability to approximately 20 %, decreased autophagy, increased cell senescence and reduced DNA binding activity of serum response factor (SRF) to serum response elements (SRE), which were paralleled by reduction in cardiac actin expression. Moreover, PON induced a significant increase of Cx43 protein and its S368-phosphorylated form (pS368-Cx43), as well as their displacement from the plasma membrane to the perinuclear and nuclear cellular region. All these effects were reverted by EMPA. CONCLUSION EMPA attenuates PON-induced cardiotoxicity by reducing senescence, enhancing the SRE-SRF binding and restoring the connexin 43-autophagy pathway. This effect may pave the way to use of SGLT2 inhibitors in attenuating tyrosine-kinase inhibitor cardiotoxicity.
Collapse
Affiliation(s)
- Letizia Mattii
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefania Moscato
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Enza Polizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Riccardo Zucchi
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Italy
| | | | - Sandra Ghelardoni
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Italy
| | - Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, Pisa, Italy.
| |
Collapse
|
6
|
Iwamoto Y, Kimura T, Iwamoto H, Sanada J, Fushimi Y, Katakura Y, Shimoda M, Kondo T, Nakanishi S, Mune T, Kaku K, Wada H, Kaneto H. Nilotinib-induced Diabetes in Japanese Patients with Chronic Myeloid Leukemia. Intern Med 2024:3740-24. [PMID: 39198174 DOI: 10.2169/internalmedicine.3740-24] [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: 09/01/2024] Open
Abstract
Objective This study aimed to examine the risk of diabetes mellitus induced by nilotinib, a second-generation tyrosine kinase inhibitor. Methods This retrospective study included 25 patients with chronic myeloid leukemia (CML) treated with nilotinib at our hospital. Four patients had diabetes mellitus at the start of nilotinib administration (prior DM group), and five patients were newly diagnosed with diabetes mellitus after the start of nilotinib administration (new DM group). Sixteen patients who were not diagnosed with diabetes mellitus were classified into the non-DM group. Changes in the blood glucose and HbA1c levels were evaluated in each group at the time of nilotinib administration and two years later. Results Molecular genetic remission of CML was achieved in 81.8% of patients with diabetes and 72.2% of patients without non-DM group. There were no cases in this study in which nilotinib was changed or discontinued owing to hyperglycemia. There was no difference in the blood glucose levels at the start of nilotinib treatment among the groups. Two years after starting nilotinib, the blood glucose levels in the new DM group (232 (186-296) mg/dL) and prior DM group (168 (123-269) mg/dL) were significantly higher than those in the non-DM group (100 (91-115) mg/dL). ΔHbA1c levels in the new DM group (1.3 (0.9-2.2) %) and prior DM group (1.6 (0.7-1.7) %) were significantly higher than those in the non-DM group (-0.2 (-0.3-0.1) %). Conclusion Nilotinib caused diabetes in 23.8% of the participants, but there were no hyperglycemia-related severe adverse events. Therefore, nilotinib may be safely continued with regular monitoring for the development of diabetes after nilotinib administration.
Collapse
Affiliation(s)
- Yuichiro Iwamoto
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | - Tomohiko Kimura
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | - Hideyuki Iwamoto
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | - Junpei Sanada
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | - Yoshiro Fushimi
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | - Yukino Katakura
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | - Masashi Shimoda
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | | | - Shuhei Nakanishi
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | - Tomoatsu Mune
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | - Kohei Kaku
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| | - Hideho Wada
- Department of Hematology, Kawasaki Medical School, Japan
| | - Hideaki Kaneto
- Department of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Japan
| |
Collapse
|
7
|
Spannbauer A, Bergler-Klein J. Cardio-Oncology: A New Discipline in Medicine and Its Relevance to Hematology. Hamostaseologie 2024; 44:255-267. [PMID: 38723641 DOI: 10.1055/a-2284-5855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2024] Open
Abstract
Cardio-oncology, a burgeoning subspecialty, addresses the complex interplay between cardiology and oncology, particularly in light of increased cardiovascular (CV) disease mortality in cancer patients. This review provides a comprehensive overview of cardio-oncology with a focus on the therapies used in hematological malignancies. We explore the bidirectional relationship between heart failure and cancer, emphasizing the need for collaborative care. The review discusses risk stratification, highlighting the importance of baseline CV risk assessment and personalized surveillance regimens. Primary and secondary prevention strategies, including pharmacological interventions, are outlined. The review also delves into the cardiotoxicity associated with hematological cancer therapies, focusing on anthracyclines, Bruton kinase inhibitors, BCR-ABL tyrosine kinase inhibitors, CAR-T cell therapy, immune checkpoint inhibitors, multiple myeloma treatments, and hematopoietic stem cell transplantation. We then highlight the high risk of venous and arterial thromboembolisms in cancer patients and the challenges of anticoagulation management in cardio-oncology. Finally, the review touches on the importance of long-term follow-up and appropriate screening in cancer survivors at high risk of CV morbidity and mortality, based on their CV risk profile and the type and dose of cardiotoxic therapies they received such as anthracyclines or high radiation doses.
Collapse
|
8
|
Liu Y, Yang Z, Zhang J, Guo N, Liu N, Zhang Q, Dang X, Li Y, Zhang J, Pan X. Integrating amino acids into Bcr-Abl inhibitors: design, synthesis, biological evaluation, and in silico studies. RSC Med Chem 2024:d4md00417e. [PMID: 39246751 PMCID: PMC11376069 DOI: 10.1039/d4md00417e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/20/2024] [Indexed: 09/10/2024] Open
Abstract
Bcr-Abl is successfully applied to drug discovery as a CML therapeutic target, but point mutation resistance has become a major challenge in the clinical treatment of CML. Our previous studies have shown that the introduction of amino acids as flexible linkers and heterocyclic structures as HBMs can achieve potent inhibition of Bcr-AblT315I. In continuation of these studies, we further enriched the linker types by developing a library of compounds with tert-leucine or serine as a linker. Biological results showed that these compounds exhibited enhanced inhibition against Bcr-AblWT and Bcr-AblT315I kinases as well as improved antiproliferative activity in leukemia cell assays compared to previously disclosed compounds. In particular, compounds TL8, TL10, BS4, BS10, SR5 and SR11 exhibited potent inhibitory activities against Ba/F3 cells bearing a T315I mutant. Additionally, compounds TL8, BS4 and SR5 effectively induced K562 cell apoptosis, arrested the cell cycle at the S or G2/M phase, and inhibited the phosphorylation of Bcr-Abl and STAT5 in a dose-dependent manner. Docking studies verified the rationality of tert-leucine or serine as a flexible linker and indicated that phenylpyridine with an amide side chain favored the potency of these inhibitors. Moreover, ADME prediction suggested that the tested compounds had a favorable safety profile. Thus, tert-leucine or serine can be used as a promising class of flexible linkers for Bcr-Abl inhibitors with heterocyclic structures as HBMs, and compounds BS4, SR5, and especially TL8, can be used as starting points for further optimization.
Collapse
Affiliation(s)
- Yuying Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University Xi'an 710061 China
| | - Zeyu Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University Xi'an 710061 China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University Xi'an 710061 China
| | - Na Guo
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University Xi'an 710061 China
| | - Nanxin Liu
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University Xi'an 710061 China
| | - Qingqing Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University Xi'an 710061 China
| | - Xintao Dang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University Xi'an 710061 China
| | - Yanchen Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University Xi'an 710061 China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University Xi'an 710061 China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University Xi'an 710061 China
| |
Collapse
|
9
|
Xu Y, Carrier M, Kimpton M. Arterial Thrombosis in Patients with Cancer. Cancers (Basel) 2024; 16:2238. [PMID: 38927943 PMCID: PMC11201749 DOI: 10.3390/cancers16122238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Patients with cancer are at increased risk of arterial thromboembolic disease due to the presence of risk factors common to both the development of cancer and arterial thrombosis, the cancer itself, and the treatments provided to treat cancer. We review here the epidemiology and pathophysiology of arterial thromboembolic disease in cancer, along with its prevention and treatment strategies. We also propose a generalized approach for the management of arterial thromboembolic disease in this patient population.
Collapse
Affiliation(s)
| | - Marc Carrier
- Department of Medicine, The Ottawa Hospital Research Institute at University of Ottawa, Ottawa, ON K1H 8L6, Canada; (Y.X.); (M.K.)
| | | |
Collapse
|
10
|
Ravichandran A, Araque JC, Lawson JW. Predicting the functional state of protein kinases using interpretable graph neural networks from sequence and structural data. Proteins 2024; 92:623-636. [PMID: 38083830 DOI: 10.1002/prot.26641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 10/13/2023] [Accepted: 11/09/2023] [Indexed: 04/13/2024]
Abstract
Protein kinases are central to cellular activities and are actively pursued as drug targets for several conditions including cancer and autoimmune diseases. Despite the availability of a large structural database for kinases, methodologies to elucidate the structure-function relationship of these proteins (without manual intervention) are lacking. Such techniques are essential in structural biology and to accelerate drug discovery efforts. Here, we implement an interpretable graph neural network (GNN) framework for classifying the functionally active and inactive states of a large set of protein kinases by only using their tertiary structure and amino acid sequence. We show that the GNN models can classify kinase structures with high accuracy (>97%). We implement the Gradient-weighted Class Activation Mapping for graphs (Graph Grad-CAM) to automatically identify structurally important residues and residue-residue contacts of the kinases without any a priori input. We show that the motifs identified through the Graph Grad-CAM methodology are functionally critical, consistent with the existing kinase literature. Notably, the highly conserved DFG and HRD motifs of the well-known hydrophobic spine are identified by the interpretable framework in addition to some of the lesser known motifs. Further, using Grad-CAM maps as the vector embedding of the protein structures, we identify the subtle differences in the crystal structures among different sub-classes of kinases in the Protein Data Bank (PDB). Frameworks such as the one implemented here, for high-throughput identification of protein structure-function relationships are essential in designing targeted small molecules therapies as well as in engineering new proteins for novel applications.
Collapse
Affiliation(s)
- Ashwin Ravichandran
- KBR Inc., Intelligent Systems Division, NASA Ames Research Center, Moffett Field, California, USA
| | - Juan C Araque
- KBR Inc., Intelligent Systems Division, NASA Ames Research Center, Moffett Field, California, USA
| | - John W Lawson
- Intelligent Systems Division, NASA Ames Research Center, Moffett Field, California, USA
| |
Collapse
|
11
|
Wang Y, Liang ZJ, Gale RP, Liao HZ, Ma J, Gong TJ, Shao YQ, Liang Y. Chronic myeloid leukaemia: Biology and therapy. Blood Rev 2024; 65:101196. [PMID: 38604819 DOI: 10.1016/j.blre.2024.101196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
Chronic myeloid leukaemia (CML) is caused by BCR::ABL1. Tyrosine kinase-inhibitors (TKIs) are the initial therapy. Several organizations have reported milestones to evaluate response to initial TKI-therapy and suggest when a change of TKI should be considered. Achieving treatment-free remission (TFR) is increasingly recognized as the optimal therapy goal. Which TKI is the best initial therapy for which persons and what depth and duration of molecular remission is needed to achieve TFR are controversial. In this review we discuss these issues and suggest future research directions.
Collapse
MESH Headings
- Humans
- Protein Kinase Inhibitors/therapeutic use
- Fusion Proteins, bcr-abl/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Remission Induction
- Biology
Collapse
Affiliation(s)
- Yun Wang
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Centre for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhi-Jian Liang
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Centre for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Robert Peter Gale
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Centre for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Hua-Ze Liao
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Centre for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jun Ma
- Harbin Institute of Hematology and Oncology, Harbin First Hospital, Harbin 150010, China
| | - Tie-Jun Gong
- Harbin Institute of Hematology and Oncology, Harbin First Hospital, Harbin 150010, China.
| | - Ying-Qi Shao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China.
| | - Yang Liang
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Centre for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China.
| |
Collapse
|
12
|
Ameri P, Bertero E, Lombardi M, Porto I, Canepa M, Nohria A, Vergallo R, Lyon AR, López-Fernández T. Ischaemic heart disease in patients with cancer. Eur Heart J 2024; 45:1209-1223. [PMID: 38323638 DOI: 10.1093/eurheartj/ehae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/22/2023] [Accepted: 01/18/2024] [Indexed: 02/08/2024] Open
Abstract
Cardiologists are encountering a growing number of cancer patients with ischaemic heart disease (IHD). Several factors account for the interrelationship between these two conditions, in addition to improving survival rates in the cancer population. Established cardiovascular (CV) risk factors, such as hypercholesterolaemia and obesity, predispose to both IHD and cancer, through specific mechanisms and via low-grade, systemic inflammation. This latter is also fuelled by clonal haematopoiesis of indeterminate potential. Furthermore, experimental work indicates that IHD and cancer can promote one another, and the CV or metabolic toxicity of anticancer therapies can lead to IHD. The connections between IHD and cancer are reinforced by social determinants of health, non-medical factors that modify health outcomes and comprise individual and societal domains, including economic stability, educational and healthcare access and quality, neighbourhood and built environment, and social and community context. Management of IHD in cancer patients is often challenging, due to atypical presentation, increased bleeding and ischaemic risk, and worse outcomes as compared to patients without cancer. The decision to proceed with coronary revascularization and the choice of antithrombotic therapy can be difficult, particularly in patients with chronic coronary syndromes, necessitating multidisciplinary discussion that considers both general guidelines and specific features on a case by case basis. Randomized controlled trial evidence in cancer patients is very limited and there is urgent need for more data to inform clinical practice. Therefore, coexistence of IHD and cancer raises important scientific and practical questions that call for collaborative efforts from the cardio-oncology, cardiology, and oncology communities.
Collapse
Affiliation(s)
- Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
| | - Edoardo Bertero
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Marco Lombardi
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Roma, Italy
| | - Italo Porto
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
| | - Marco Canepa
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
| | - Anju Nohria
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Rocco Vergallo
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
| | | | - Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
- Cardiology Department, Quirón Pozuelo University Hospital, Madrid, Spain
| |
Collapse
|
13
|
Costanzo V, Ratre YK, Andretta E, Acharya R, Bhaskar LVKS, Verma HK. A Comprehensive Review of Cancer Drug-Induced Cardiotoxicity in Blood Cancer Patients: Current Perspectives and Therapeutic Strategies. Curr Treat Options Oncol 2024; 25:465-495. [PMID: 38372853 DOI: 10.1007/s11864-023-01175-z] [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] [Accepted: 12/19/2023] [Indexed: 02/20/2024]
Abstract
OPINION STATEMENT Cardiotoxicity has emerged as a serious outcome catalyzed by various therapeutic targets in the field of cancer treatment, which includes chemotherapy, radiation, and targeted therapies. The growing significance of cancer drug-induced cardiotoxicity (CDIC) and radiation-induced cardiotoxicity (CRIC) necessitates immediate attention. This article intricately unveils how cancer treatments cause cardiotoxicity, which is exacerbated by patient-specific risks. In particular, drugs like anthracyclines, alkylating agents, and tyrosine kinase inhibitors pose a risk, along with factors such as hypertension and diabetes. Mechanistic insights into oxidative stress and topoisomerase-II-B inhibition are crucial, while cardiac biomarkers show early damage. Timely intervention and prompt treatment, especially with specific agents like dexrazoxane and beta-blockers, are pivotal in the proactive management of CDIC.
Collapse
Affiliation(s)
- Vincenzo Costanzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | | | - Emanuela Andretta
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Rakesh Acharya
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - L V K S Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764, Munich, Germany.
| |
Collapse
|
14
|
Pinheiro EA, DeKeyser JM, Lenny B, Sapkota Y, Burridge PW. Nilotinib-induced alterations in endothelial cell function recapitulate clinical vascular phenotypes independent of ABL1. Sci Rep 2024; 14:7123. [PMID: 38532120 DOI: 10.1038/s41598-024-57686-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/20/2024] [Indexed: 03/28/2024] Open
Abstract
Nilotinib is a highly effective treatment for chronic myeloid leukemia but has been consistently associated with the development of nilotinib-induced arterial disease (NAD) in a subset of patients. To date, which cell types mediate this effect and whether NAD results from on-target mechanisms is unknown. We utilized human induced pluripotent stem cells (hiPSCs) to generate endothelial cells and vascular smooth muscle cells for in vitro study of NAD. We found that nilotinib adversely affects endothelial proliferation and migration, in addition to increasing intracellular nitric oxide. Nilotinib did not alter endothelial barrier function or lipid uptake. No effect of nilotinib was observed in vascular smooth muscle cells, suggesting that NAD is primarily mediated through endothelial cells. To evaluate whether NAD results from enhanced inhibition of ABL1, we generated multiple ABL1 knockout lines. The effects of nilotinib remained unchanged in the absence of ABL1, suggesting that NAD results from off- rather than on-target signaling. The model established in the present study can be applied to future mechanistic and patient-specific pharmacogenomic studies.
Collapse
Affiliation(s)
- Emily A Pinheiro
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, 320 E Superior St, Searle 8-525, Chicago, IL, 60611, USA
| | - Jean-Marc DeKeyser
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, 320 E Superior St, Searle 8-525, Chicago, IL, 60611, USA
| | - Brian Lenny
- Department of Epidemiology and Cancer Control, St. Jude Children's Hospital, Memphis, TN, 38105, USA
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children's Hospital, Memphis, TN, 38105, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, 320 E Superior St, Searle 8-525, Chicago, IL, 60611, USA.
| |
Collapse
|
15
|
Yan G, Han Z, Kwon Y, Jousma J, Nukala SB, Prosser BL, Du X, Pinho S, Ong SB, Lee WH, Ong SG. Integrated Stress Response Potentiates Ponatinib-Induced Cardiotoxicity. Circ Res 2024; 134:482-501. [PMID: 38323474 PMCID: PMC10940206 DOI: 10.1161/circresaha.123.323683] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/22/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Mitochondrial dysfunction is a primary driver of cardiac contractile failure; yet, the cross talk between mitochondrial energetics and signaling regulation remains obscure. Ponatinib, a tyrosine kinase inhibitor used to treat chronic myeloid leukemia, is among the most cardiotoxic tyrosine kinase inhibitors and causes mitochondrial dysfunction. Whether ponatinib-induced mitochondrial dysfunction triggers the integrated stress response (ISR) to induce ponatinib-induced cardiotoxicity remains to be determined. METHODS Using human induced pluripotent stem cells-derived cardiomyocytes and a recently developed mouse model of ponatinib-induced cardiotoxicity, we performed proteomic analysis, molecular and biochemical assays to investigate the relationship between ponatinib-induced mitochondrial stress and ISR and their role in promoting ponatinib-induced cardiotoxicity. RESULTS Proteomic analysis revealed that ponatinib activated the ISR in cardiac cells. We identified GCN2 (general control nonderepressible 2) as the eIF2α (eukaryotic translation initiation factor 2α) kinase responsible for relaying mitochondrial stress signals to trigger the primary ISR effector-ATF4 (activating transcription factor 4), upon ponatinib exposure. Mechanistically, ponatinib treatment exerted inhibitory effects on ATP synthase activity and reduced its expression levels resulting in ATP deficits. Perturbed mitochondrial function resulting in ATP deficits then acts as a trigger of GCN2-mediated ISR activation, effects that were negated by nicotinamide mononucleotide, an NAD+ precursor, supplementation. Genetic inhibition of ATP synthase also activated GCN2. Interestingly, we showed that the decreased abundance of ATP also facilitated direct binding of ponatinib to GCN2, unexpectedly causing its activation most likely because of a conformational change in its structure. Importantly, administering an ISR inhibitor protected human induced pluripotent stem cell-derived cardiomyocytes against ponatinib. Ponatinib-treated mice also exhibited reduced cardiac function, effects that were attenuated upon systemic ISRIB administration. Importantly, ISRIB does not affect the antitumor effects of ponatinib in vitro. CONCLUSIONS Neutralizing ISR hyperactivation could prevent or reverse ponatinib-induced cardiotoxicity. The findings that compromised ATP production potentiates GCN2-mediated ISR activation have broad implications across various cardiac diseases. Our results also highlight an unanticipated role of ponatinib in causing direct activation of a kinase target despite its role as an ATP-competitive kinase inhibitor.
Collapse
Affiliation(s)
- Gege Yan
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Zhenbo Han
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Youjeong Kwon
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Jordan Jousma
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Sarath Babu Nukala
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Benjamin L Prosser
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xiaoping Du
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Sandra Pinho
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Sang-Bing Ong
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, CUHK, Hong Kong SAR, China
- Hong Kong Hub of Pediatric Excellence (HK HOPE), Hong Kong Children’s Hospital (HKCH), Kowloon Bay, Hong Kong SAR, China
- Kunming Institute of Zoology – The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Neural, Vascular, and Metabolic Biology Thematic Research Program, School of Biomedical Sciences (SBS), Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, USA
| | - Sang-Ging Ong
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, USA
| |
Collapse
|
16
|
Vakilpour A, Lefebvre B, Lai C, Scherrer-Crosbie M. Heartbreaker: Detection and prevention of cardiotoxicity in hematological malignancies. Blood Rev 2024; 64:101166. [PMID: 38182490 DOI: 10.1016/j.blre.2023.101166] [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: 10/30/2023] [Revised: 12/12/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Cancer survivors are at significant risk of cardiovascular (CV) morbidity and mortality; patients with hematologic malignancies have a higher rate of death due to heart failure compared to all other cancer subtypes. The majority of conventional hematologic cancer treatments is associated with increased risk of acute and long-term CV toxicity. The incidence of cancer therapy induced CV toxicity depends on the combination of patient characteristics and on the type, dose, and duration of the therapy. Early diagnosis of CV toxicity, appropriate referral, more specific cardiac monitoring follow-up and timely interventions in target patients can decrease the risk of CV adverse events, the interruption of oncological therapy, and improve the patient's prognosis. Herein, we summarize the CV effects of conventional treatments used in hematologic malignancies with a focus on definitions and incidence of the most common CV toxicities, guideline recommended early detection approaches, and preventive strategies before and during cancer treatments.
Collapse
Affiliation(s)
- Azin Vakilpour
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| | - Bénédicte Lefebvre
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; The Thalheimer Center for Cardio-oncology, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Catherine Lai
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; The Thalheimer Center for Cardio-oncology, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Mozneb M, Jenkins A, Sances S, Pohlman S, Workman MJ, West D, Ondatje B, El-Ghazawi K, Woodbury A, Garcia VJ, Patel S, Arzt M, Dezem F, Laperle AH, Moser VA, Ho R, Yucer N, Plummer J, Barrett RJ, Svendsen CN, Sharma A. Multi-lineage heart-chip models drug cardiotoxicity and enhances maturation of human stem cell-derived cardiovascular cells. LAB ON A CHIP 2024; 24:869-881. [PMID: 38252454 DOI: 10.1039/d3lc00745f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Cardiovascular toxicity causes adverse drug reactions and may lead to drug removal from the pharmaceutical market. Cancer therapies can induce life-threatening cardiovascular side effects such as arrhythmias, muscle cell death, or vascular dysfunction. New technologies have enabled cardiotoxic compounds to be identified earlier in drug development. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) and vascular endothelial cells (ECs) can screen for drug-induced alterations in cardiovascular cell function and survival. However, most existing hiPSC models for cardiovascular drug toxicity utilize two-dimensional, immature cells grown in static culture. Improved in vitro models to mechanistically interrogate cardiotoxicity would utilize more adult-like, mature hiPSC-derived cells in an integrated system whereby toxic drugs and protective agents can flow between hiPSC-ECs that represent systemic vasculature and hiPSC-CMs that represent heart muscle (myocardium). Such models would be useful for testing the multi-lineage cardiotoxicities of chemotherapeutic drugs such as VEGFR2/PDGFR-inhibiting tyrosine kinase inhibitors (VPTKIs). Here, we develop a multi-lineage, fully-integrated, cardiovascular organ-chip that can enhance hiPSC-EC and hiPSC-CM functional and genetic maturity, model endothelial barrier permeability, and demonstrate long-term functional stability. This microfluidic organ-chip harbors hiPSC-CMs and hiPSC-ECs on separate channels that can be subjected to active fluid flow and rhythmic biomechanical stretch. We demonstrate the utility of this cardiovascular organ-chip as a predictive platform for evaluating multi-lineage VPTKI toxicity. This study may lead to the development of new modalities for the evaluation and prevention of cancer therapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Maedeh Mozneb
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Amelia Jenkins
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Samuel Sances
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Stephany Pohlman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael J Workman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Dylan West
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Briana Ondatje
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Kareem El-Ghazawi
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Amanda Woodbury
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Veronica J Garcia
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Shachi Patel
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Madelyn Arzt
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Felipe Dezem
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alex H Laperle
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - V Alexandra Moser
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Ritchie Ho
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Nur Yucer
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Jasmine Plummer
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robert J Barrett
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
18
|
Jiang C, Xu H, Wu Y. Effect of chemotherapy in tumor on coronary arteries: Mechanisms and management. Life Sci 2024; 338:122377. [PMID: 38135114 DOI: 10.1016/j.lfs.2023.122377] [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: 08/20/2023] [Revised: 11/29/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
Coronary artery disease (CAD) is an important contributor to the cardiovascular burden in cancer survivors. The development of coronary ischemia events, myocardial infarction, and heart failure has been associated with many conventional chemotherapeutic agents, new targeted therapies, and immunotherapy. The most frequent pathological manifestations of chemotherapy-mediated coronary damage include acute vasospasm, acute thrombosis, accelerated atherosclerosis development, and microvascular dysfunction. Potential screening techniques for CAD patients include baseline risk factor evaluation, polygenic risk factors, and coronary artery calcium scores. Determining the risk requires consideration of both the type of chemotherapy and the type of cancer being treated. Cardiology-oncology guidelines offer some suggestions for the care of coronary artery disease, which might involve medication, lifestyle changes, and coronary revascularization.
Collapse
Affiliation(s)
- Chengqing Jiang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haiyan Xu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Yongjian Wu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
19
|
Duijvelaar E, Gisby J, Peters JE, Bogaard HJ, Aman J. Longitudinal plasma proteomics reveals biomarkers of alveolar-capillary barrier disruption in critically ill COVID-19 patients. Nat Commun 2024; 15:744. [PMID: 38272877 PMCID: PMC10811341 DOI: 10.1038/s41467-024-44986-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
The pathobiology of respiratory failure in COVID-19 consists of a complex interplay between viral cytopathic effects and a dysregulated host immune response. In critically ill patients, imatinib treatment demonstrated potential for reducing invasive ventilation duration and mortality. Here, we perform longitudinal profiling of 6385 plasma proteins in 318 hospitalised patients to investigate the biological processes involved in critical COVID-19, and assess the effects of imatinib treatment. Nine proteins measured at hospital admission accurately predict critical illness development. Next to dysregulation of inflammation, critical illness is characterised by pathways involving cellular adhesion, extracellular matrix turnover and tissue remodelling. Imatinib treatment attenuates protein perturbations associated with inflammation and extracellular matrix turnover. These proteomic alterations are contextualised using external pulmonary RNA-sequencing data of deceased COVID-19 patients and imatinib-treated Syrian hamsters. Together, we show that alveolar capillary barrier disruption in critical COVID-19 is reflected in the plasma proteome, and is attenuated with imatinib treatment. This study comprises a secondary analysis of both clinical data and plasma samples derived from a clinical trial that was registered with the EU Clinical Trials Register (EudraCT 2020-001236-10, https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001236-10/NL ) and Netherlands Trial Register (NL8491, https://www.trialregister.nl/trial/8491 ).
Collapse
Affiliation(s)
- Erik Duijvelaar
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| | - Jack Gisby
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - James E Peters
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Baum J, Lax H, Lehmann N, Merkel-Jens A, Beelen DW, Jöckel KH, Dührsen U. Health-Related Complications during Follow-Up and Their Impact on Blood Cancer Survivors: Results from the "Aftercare in Blood Cancer Survivors" (ABC) Study. Acta Haematol 2024; 147:435-446. [PMID: 38228094 DOI: 10.1159/000536155] [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: 08/18/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024]
Abstract
INTRODUCTION Blood cancer survivors are at increased risk for medical complications. METHODS Our questionnaire-based study involved 1,551 blood cancer survivors with a ≥3-year interval since the last intense treatment. Its goal was to quantify health-related complications during follow-up and assess their impact on the patients' lives. RESULTS A total of 20.4% of the responding survivors reported a disease relapse, most often in indolent lymphomas. Second primary malignancies occurred in 14.1%, primarily in lymphoma and allogeneic transplantation survivors. The most frequent malignancy was basal cell carcinoma of the skin, but myeloid malignancies, melanoma, bladder, head-and-neck, and thyroid cancer also appeared disproportionately frequent. An increased infection rate was reported by 43.7%, most often after allogeneic transplantation. New cardiovascular diseases were reported by 30.2%, with a high rate of thromboembolic events in multiple myeloma (MM) and myeloproliferative diseases. Polyneuropathies were reported by 39.1%, most often by survivors with a history of MM or aggressive lymphoma. Disease relapse was perceived as the highest burden, followed by second primary malignancy, increased infection frequency, and polyneuropathy. In each area investigated, the range of perceived severities was wide. CONCLUSIONS Health-related complications are frequent during blood cancer follow-up, with significant repercussions on the patients' lives.
Collapse
Affiliation(s)
- Julia Baum
- Klinik für Hämatologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Hildegard Lax
- Institut für Medizinische Informatik, Biometrie und Epidemiologie, Universität Duisburg-Essen, Essen, Germany
| | - Nils Lehmann
- Institut für Medizinische Informatik, Biometrie und Epidemiologie, Universität Duisburg-Essen, Essen, Germany
| | - Anja Merkel-Jens
- Institut für Medizinische Informatik, Biometrie und Epidemiologie, Universität Duisburg-Essen, Essen, Germany
| | - Dietrich W Beelen
- Klinik für Knochenmarktransplantation, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Karl-Heinz Jöckel
- Institut für Medizinische Informatik, Biometrie und Epidemiologie, Universität Duisburg-Essen, Essen, Germany
| | - Ulrich Dührsen
- Klinik für Hämatologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| |
Collapse
|
21
|
Huang CE, Lee KD, Chang JJ, Tzeng HE, Huang SH, Yu LHL, Chen MC. Association of Nilotinib With Cardiovascular Diseases in Patients With Chronic Myelogenous Leukemia: A National Population-Based Cohort Study. Oncologist 2024; 29:e81-e89. [PMID: 37561957 PMCID: PMC10769786 DOI: 10.1093/oncolo/oyad225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Tyrosine kinase inhibitor (TKI) treatment has been identified to be a risk factor for metabolic syndrome and cardiovascular diseases (CVDs) in patients diagnosed with chronic myeloid leukemia (CML). However, the specific contribution of post-TKI metabolic syndrome and the individual TKIs, including imatinib, nilotinib, and dasatinib, contribute to the development of CVDs remains unclear. METHODS We conducted a nationwide database to investigate the incidence of post-TKI metabolic syndrome, including diabetes, hyperlipidemia, and hypertension, as well as their association with CVDs. To compare the risk of post-TKI comorbidities and CVDs among TKIs, we utilized the incidence rate ratio (IRR), and subdistribution hazard ratio (SHR) calculated from multiple Fine-Gray models. RESULTS A total of 1211 patients without diabetes, 1235 patients without hyperlipidemia, and 1074 patients without hypertension were enrolled in the study. The incidence rate of post-TKI diabetes and hyperlipidemia was the highest in patients treated with nilotinib compared to imatinib and dasatinib (IRRs ≥ 3.15, Ps ≤ .047). After adjusting for confounders, nilotinib remained a significant risk factor for post-TKI diabetes and hyperlipidemia at an SHR of 3.83 (P < .001) and 5.15 (P < .001), respectively. Regarding the occurrence of CVDs, patients treated with nilotinib were more likely to develop CVDs than those treated with imatinib in non-hyperlipidemic group (IRR = 3.21, P = .020). Pre-existing and post-TKI hyperlipidemia were found to have a stronger association with CVDs, with SHR values of 5.81 (P = .034) and 13.21 (P = .001), respectively. CONCLUSION The findings of this study indicate that nilotinib treatment is associated with increased risks of diabetes and hyperlipidemia, with hyperlipidemia being the most significant risk for CVDs. Therefore, we recommend that CML patients receiving nilotinib should undergo screening for diabetes and hyperlipidemia prior to initiating TKI treatment. Additionally, regular monitoring of lipid profiles during TKI therapy and implementing effective management strategies to control hyperlipidemia are crucial.
Collapse
Affiliation(s)
- Cih-En Huang
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuan-Der Lee
- Cell Therapy and Regenerative Medicine Center, Comprehensive Cancer Center, Taichung Veterans General Hospital, Taichung, Taiwan
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Jung-Jung Chang
- Division of Cardiology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Huey-En Tzeng
- Division of Transfusion Medicine, Department of Pathology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shih-Hao Huang
- Department of Public Health and Biostatistics Consulting Center, Chang Gung University, Taoyuan, Taiwan
| | | | - Min-Chi Chen
- Department of Public Health and Biostatistics Consulting Center, Chang Gung University, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| |
Collapse
|
22
|
Zhang Y, Wu X, Sun X, Yang J, Liu C, Tang G, Lei X, Huang H, Peng J. The Progress of Small Molecule Targeting BCR-ABL in the Treatment of Chronic Myeloid Leukemia. Mini Rev Med Chem 2024; 24:642-663. [PMID: 37855278 DOI: 10.2174/0113895575218335230926070130] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/14/2023] [Accepted: 07/14/2023] [Indexed: 10/20/2023]
Abstract
Chronic myelogenous leukemia (CML) is a malignant myeloproliferative disease. According to the American Cancer Society's 2021 cancer data report, new cases of CML account for about 15% of all leukemias. CML is generally divided into three stages: chronic phase, accelerated phase, and blast phase. Nearly 90% of patients are diagnosed as a chronic phase. Allogeneic stem cell transplantation and chemotherapeutic drugs, such as interferon IFN-α were used as the earliest treatments for CML. However, they could generate obvious side effects, and scientists had to seek new treatments for CML. A new era of targeted therapy for CML began with the introduction of imatinib, the first-generation BCR-ABL kinase inhibitor. However, the ensuing drug resistance and mutant strains led by T315I limited the further use of imatinib. With the continuous advancement of research, tyrosine kinase inhibitors (TKI) and BCR-ABL protein degraders with novel structures and therapeutic mechanisms have been discovered. From biological macromolecules to classical target protein inhibitors, a growing number of compounds are being developed to treat chronic myelogenous leukemia. In this review, we focus on summarizing the current situation of a series of candidate small-molecule drugs in CML therapy, including TKIs and BCR-ABL protein degrader. The examples provided herein describe the pharmacology activity of small-molecule drugs. These drugs will provide new enlightenment for future treatment directions.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Pharmacy, School of Pharmacy, Hengyang Medical School, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Xin Wu
- Department of Pharmacy, School of Pharmacy, Hengyang Medical School, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Xueyan Sun
- Department of Pharmacy, School of Pharmacy, Hengyang Medical School, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Jun Yang
- Department of Pharmacy, School of Pharmacy, Hengyang Medical School, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Chang Liu
- Department of Pharmacy, School of Pharmacy, Hengyang Medical School, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Guotao Tang
- Department of Pharmacy, School of Pharmacy, Hengyang Medical School, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaoyong Lei
- Department of Pharmacy, School of Pharmacy, Hengyang Medical School, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Honglin Huang
- Department of Pharmacy, School of Pharmacy, Hengyang Medical School, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Junmei Peng
- Department of Pharmacy, School of Pharmacy, Hengyang Medical School, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
23
|
El-Damasy AK, Jin H, Park JW, Kim HJ, Khojah H, Seo SH, Lee JH, Bang EK, Keum G. Overcoming the imatinib-resistant BCR-ABL mutants with new ureidobenzothiazole chemotypes endowed with potent and broad-spectrum anticancer activity. J Enzyme Inhib Med Chem 2023; 38:2189097. [PMID: 36927348 PMCID: PMC10026764 DOI: 10.1080/14756366.2023.2189097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
The design of kinase inhibitors targeting the oncogenic kinase BCR-ABL constitutes a promising paradigm for treating chronic myeloid leukaemia (CML). Nevertheless, the efficacy of imatinib, the first FDA-approved targeted therapy for CML, is curbed by the emergence of resistance. Herein, we report the identification of the 2-methoxyphenyl ureidobenzothiazole AK-HW-90 (2b) as a potent pan-BCR-ABL inhibitor against imatinib-resistant mutants, particularly T315I. A concise array of six compounds 2a-f was designed based on our previously reported benzothiazole lead AKE-5l to improve its BCR-ABLT315I inhibitory activity. Replacing the 6-oxypicolinamide moiety of AKE-5l with o-methoxyphenyl and changing the propyl spacer with phenyl afforded 2a and AK-HW-90 (2b) with IC50 values of 2.0 and 0.65 nM against BCR-ABLT315I, respectively. AK-HW-90 showed superior anticancer potency to imatinib against multiple cancer cells (NCI), including leukaemia K-562. The obtained outcomes offer AK-HW-90 as a promising candidate for the treatment of CML and other types of cancer.
Collapse
Affiliation(s)
- Ashraf K El-Damasy
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Heewon Jin
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Jung Woo Park
- Center for Supercomputing Applications, Div. of National Supercomputing R&D, Korea Institute of Science and Technology Information, Daejeon, Republic of Korea
| | - Hyun Ji Kim
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hanan Khojah
- Department of Pharmacognosy, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Seon Hee Seo
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Ju-Hyeon Lee
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Eun-Kyoung Bang
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Gyochang Keum
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| |
Collapse
|
24
|
Nakajima H, Miyake R, Hongo T, Takasaki M, Fujimoto Y, Io K. Moyamoya disease presenting with symptomatic ischemic stroke during new-generation tyrosine kinase inhibitor treatment: two illustrative cases. Acta Neurochir (Wien) 2023; 165:3637-3641. [PMID: 37932634 DOI: 10.1007/s00701-023-05858-2] [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: 09/11/2023] [Accepted: 10/16/2023] [Indexed: 11/08/2023]
Abstract
Tyrosine kinase inhibitors (TKIs) have been widely used to treat chronic myeloid leukemia. Nilotinib and ponatinib, which are second- and third-generation TKIs, have been reported to cause cerebrovascular arterial complications. Here, we present two cases of moyamoya disease presenting with symptomatic ischemic stroke during new-generation TKI treatment. We judged that new-generation TKI treatment was a factor in symptomatic ischemic stroke of unknown moyamoya disease in both cases. Noninvasive examinations using magnetic resonance imaging or carotid ultrasonography should be performed before and during new-generation TKI treatment in order to prevent symptomatic ischemic stroke.
Collapse
Affiliation(s)
- Hideki Nakajima
- Department of Neurosurgery, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-Ku, Osaka, 553-0003, Japan.
| | - Ryota Miyake
- Department of Neurosurgery, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-Ku, Osaka, 553-0003, Japan
| | - Taku Hongo
- Department of Neurosurgery, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-Ku, Osaka, 553-0003, Japan
| | - Morio Takasaki
- Department of Neurosurgery, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-Ku, Osaka, 553-0003, Japan
| | - Yasuhiro Fujimoto
- Department of Neurosurgery, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-Ku, Osaka, 553-0003, Japan
| | - Katsuhiro Io
- Department of Hematology, Kansai Electric Power Hospital, Osaka, Japan
| |
Collapse
|
25
|
Iurlo A, Cattaneo D, Bucelli C, Spallarossa P, Passamonti F. Cardiovascular Adverse Events of Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia: Clinical Relevance, Impact on Outcome, Preventive Measures and Treatment Strategies. Curr Treat Options Oncol 2023; 24:1720-1738. [PMID: 38047977 DOI: 10.1007/s11864-023-01149-1] [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] [Accepted: 11/12/2023] [Indexed: 12/05/2023]
Abstract
OPINION STATEMENT The introduction of TKIs into the therapeutic armamentarium of CML has changed the disease paradigm, increasing long-term survival from 20% to over 80%, with a life expectancy now approaching that of the general population. Although highly effective, TKIs also have a toxicity profile that is often mild to moderate, but sometimes severe, with multiple kinases involved in the development of adverse events (AEs). Among others, cardiovascular AEs observed in TKI-treated CML patients may represent a significant cause of morbidity and mortality, and their pathogenesis is still only partially understood. In view of the recent introduction into daily clinical practice of new TKIs, namely the STAMP inhibitor asciminib, with a distinct safety profile, hematologists now more than ever have the opportunity to select the most suitable TKI for each patient, an aspect that will be fundamental in terms of personalized preventive and therapeutic strategies. Furthermore, physicians should be aware of the feasibility of TKI dose modifications at all stages of the patients' treatment journey, both at diagnosis for frail or elderly subjects or with multiple comorbidities, and during follow-up for those patients who experience toxicity, as well as to prevent it, with the main objective of reducing side effects while maintaining the response. Consequently, preserving the cardiovascular health of CML patients will likely be a more urgent topic in the near future, with specific measures aimed at controlling cardiovascular risk factors through a multidisciplinary approach involving a panel of healthcare professionals together with the hematologist.
Collapse
Affiliation(s)
- Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milano, Italy.
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milano, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milano, Italy
| | - Paolo Spallarossa
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino - Italian IRCCS Cardiology Network, Genova, Italy
| | - Francesco Passamonti
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milano, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
26
|
Aghel N, Gustafson D, Delgado D, Atenafu EG, Fish JE, Lipton JH. High sensitivity c-reactive protein and circulating biomarkers of endothelial dysfunction in patients with chronic myeloid leukemia receiving tyrosine kinase inhibitors. Leuk Lymphoma 2023; 64:2008-2017. [PMID: 37554059 DOI: 10.1080/10428194.2023.2242990] [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: 01/10/2023] [Revised: 06/25/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023]
Abstract
Tyrosine kinase inhibitors (TKIs) have revolutionized the management of patients with chronic myelogenous leukemia (CML); however, they may cause cardiovascular (CV) toxicities. In this cross-sectional study, we explored whether high-sensitivity C-reactive protein (hsCRP) and novel markers of vascular dysfunction were associated with exposure to specific TKIs, in 262 CML patients. Hs-CRP level was not associated with CML disease activity or treatment with a specific TKI. Body mass index (OR: 1.15, 95% CI: 1.108-1.246; p < 0.001) and CML duration (OR: 1.004, 95% CI: 1.001-1.008; p = 0.024) were independently associated with higher hs-CRP. In exploratory analyses, novel endothelial-centric markers (e.g. ET-1 and VCAM-1) were differential across the various TKIs, particularly amongst nilotinib- and ponatinib-treated patients. While Levels of hs-CRP do not appear to be correlated with specific TKIs, circulating markers of vascular dysfunction were altered in patients treated with specific TKIs and should be explored as potential markers of TKI-associated CV risk.
Collapse
Affiliation(s)
- Nazanin Aghel
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, Ted Rogers Program in Cardiotoxicity Prevention University Health Network, Toronto, Canada
- Division of Cardiology, Cardio-Oncology Program, McMaster University, Hamilton, Canada
| | - Dakota Gustafson
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Diego Delgado
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, Ted Rogers Program in Cardiotoxicity Prevention University Health Network, Toronto, Canada
| | - Eshetu G Atenafu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network University of Toronto, Toronto, ON, Canada
| | - Jason E Fish
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, Ted Rogers Program in Cardiotoxicity Prevention University Health Network, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Jeffrey H Lipton
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Park H, Kim HJ, Sohn SK, Baik Y, Kim D, Lee SY, Kong JH, Kim H, Shin DY, Ahn JS, Park J, Park S, Kim I. Effect of BCR::ABL1 transcript type and droplet digital polymerase chain reaction on successful treatment-free remission in chronic myeloid leukemia patients who discontinued tyrosine kinase inhibitor. Ther Adv Hematol 2023; 14:20406207231205637. [PMID: 37929079 PMCID: PMC10624046 DOI: 10.1177/20406207231205637] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/05/2023] [Indexed: 11/07/2023] Open
Abstract
Background Droplet digital polymerase chain reaction (ddPCR) is an exact method of measurement. Objectives We conducted this study to identify the prognostic factors for successful treatment-free remission in patients with chronic-phase chronic myeloid leukemia who discontinued tyrosine kinase inhibitors (TKIs). We also aimed to validate ddPCR for predicting molecular relapse. Design This is a prospective, multicenter study. Methods We enrolled patients treated with TKIs for at least 3 years with a confirmed sustained deep molecular response (DMR) for at least 1 year. TKI was re-administered in patients who experienced the loss of major molecular response (MMR). Results A total of 66 patients from five institutions in South Korea were enrolled. During a median follow-up period of 16.5 months, 29/66 (43.9%) patients experienced molecular relapse; the probability of molecular relapse-free survival (RFS) at 6 or 12 months after TKI discontinuation was 65.6% or 57.8%, respectively, with most molecular relapses occurring within the first 7 months. All patients who lost MMR were re-treated with TKI, and all re-achieved MMR at a median of 2.8 months. E14a2 transcript type (p = 0.005) and longer DMR duration (⩾48 months) prior to TKI discontinuation (p = 0.002) were associated with prolonged molecular RFS and with sustained DMR. Patients with both e13a2 transcript type and detectable BCR::ABL1 (⩾MR5.0) by ddPCR at the time of TKI discontinuation showed shorter duration of molecular RFS (p = 0.015). Conclusion Our data suggest that transcript type and BCR::ABL1 transcript levels on ddPCR should be taken into consideration when deciding whether to discontinue TKI therapy.
Collapse
Affiliation(s)
- Hyunkyung Park
- Department of Internal Medicine, Seoul National University–Seoul Metropolitan Government Boramae Medical Center, Seoul, South Korea
| | - Hyeong-Joon Kim
- Department of Internal Medicine, Chonnam National University, Hwasun Hospital, Hwasun, South Korea
| | - Sang-Kyun Sohn
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | | | | | | | - Jee Hyun Kong
- Department of Internal Medicine, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Hawk Kim
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, South Korea
| | - Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jae-Sook Ahn
- Department of Internal Medicine, Chonnam National University, Hwasun Hospital, Hwasun, South Korea
| | - Jinny Park
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, South Korea
| | - Seonyang Park
- Department of Internal Medicine, Inje University, Haeundae Paik Hospital, Busan, South Korea
| | - Inho Kim
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul 03080, South Korea
| |
Collapse
|
28
|
Prousi GS, Joshi AM, Atti V, Addison D, Brown SA, Guha A, Patel B. Vascular Inflammation, Cancer, and Cardiovascular Diseases. Curr Oncol Rep 2023; 25:955-963. [PMID: 37261651 DOI: 10.1007/s11912-023-01426-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 06/02/2023]
Abstract
PURPOSE OF REVIEW Cancer and cardiovascular disease are among the leading causes of morbidity and mortality in the USA. Cancer and cardiovascular disease have inflammatory underpinnings that have been associated with both the development and progression of these disease states. RECENT FINDINGS Inflammatory signaling has been found to be a critical event in both cardiovascular disease and cancer formation and progression. Further, many chemotherapeutic agents potentiate inflammation exacerbating existing cardiovascular disease or leading to its presence. The exact mechanisms of these interactions remain poorly understood. The proinflammatory milieu observed in both cancer and cardiovascular disease likely plays an important role in the development and potentiation of both conditions. Further evaluation of this relationship will be critical in the development of new diagnostic and therapeutic modalities.
Collapse
Affiliation(s)
| | - Amogh M Joshi
- Department of Cardiology, Lehigh Valley Health Network, Allentown, PA, USA
| | - Varun Atti
- Heart and Vascular Institute, West Virginia University, 1 Medical Center Dr, Morgantown, WV, 26505, USA
| | - Daniel Addison
- Department of Cardiology, Ohio State University, Columbus, OH, USA
| | - Sherry-Ann Brown
- Department of Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Avirup Guha
- Department of Cardiology, Medical College of Georgia, Augusta, GA, USA
| | - Brijesh Patel
- Heart and Vascular Institute, West Virginia University, 1 Medical Center Dr, Morgantown, WV, 26505, USA.
| |
Collapse
|
29
|
Liang JP, Mi RH, Chen L, Wei XD. Nilotinib cause moyamoya disease in the treatment of chronic myelogenous leukemia during chronic phase: A case report. Asian J Surg 2023; 46:4001-4002. [PMID: 37137777 DOI: 10.1016/j.asjsur.2023.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 05/05/2023] Open
Affiliation(s)
- Jin-Ping Liang
- Department of Hematology, The Affiliated Tumor Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou, China
| | - Rui-Hua Mi
- Department of Hematology, The Affiliated Tumor Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou, China
| | - Lin Chen
- Department of Hematology, The Affiliated Tumor Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou, China
| | - Xu-Dong Wei
- Department of Hematology, The Affiliated Tumor Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou, China.
| |
Collapse
|
30
|
Sun S, Qin J, Liao W, Gao X, Shang Z, Luo D, Xiong S. Mitochondrial Dysfunction in Cardiotoxicity Induced by BCR-ABL1 Tyrosine Kinase Inhibitors -Underlying Mechanisms, Detection, Potential Therapies. Cardiovasc Toxicol 2023; 23:233-254. [PMID: 37479951 DOI: 10.1007/s12012-023-09800-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/08/2023] [Indexed: 07/23/2023]
Abstract
The advent of BCR-ABL tyrosine kinase inhibitors (TKIs) targeted therapy revolutionized the treatment of chronic myeloid leukemia (CML) patients. Mitochondria are the key organelles for the maintenance of myocardial tissue homeostasis. However, cardiotoxicity associated with BCR-ABL1 TKIs can directly or indirectly cause mitochondrial damage and dysfunction, playing a pivotal role in cardiomyocytes homeostatic system and putting the cancer survivors at higher risk. In this review, we summarize the cardiotoxicity caused by BCR-ABL1 TKIs and the underlying mechanisms, which contribute dominantly to the damage of mitochondrial structure and dysfunction: endoplasmic reticulum (ER) stress, mitochondrial stress, damage of myocardial cell mitochondrial respiratory chain, increased production of mitochondrial reactive oxygen species (ROS), and other kinases and other potential mechanisms of cardiotoxicity induced by BCR-ABL1 TKIs. Furthermore, detection and management of BCR-ABL1 TKIs will promote our rational use, and cardioprotection strategies based on mitochondria will improve our understanding of the cardiotoxicity from a mitochondrial perspective. Ultimately, we hope shed light on clinical decision-making. By integrate and learn from both research and practice, we will endeavor to minimize the mitochondria-mediated cardiotoxicity and reduce the adverse sequelae associated with BCR-ABL1 TKIs.
Collapse
Affiliation(s)
- Sheng Sun
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Medical Oncology, Hospital of Chengdu University of Traditioanal Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Jiqiu Qin
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenhao Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhoubiao Shang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dehua Luo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaoquan Xiong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Medical Oncology, Hospital of Chengdu University of Traditioanal Chinese Medicine, Chengdu, 610075, Sichuan Province, China.
| |
Collapse
|
31
|
Shyam Sunder S, Sharma UC, Pokharel S. Adverse effects of tyrosine kinase inhibitors in cancer therapy: pathophysiology, mechanisms and clinical management. Signal Transduct Target Ther 2023; 8:262. [PMID: 37414756 PMCID: PMC10326056 DOI: 10.1038/s41392-023-01469-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/06/2023] [Accepted: 04/23/2023] [Indexed: 07/08/2023] Open
Abstract
Since their invention in the early 2000s, tyrosine kinase inhibitors (TKIs) have gained prominence as the most effective pathway-directed anti-cancer agents. TKIs have shown significant utility in the treatment of multiple hematological malignancies and solid tumors, including chronic myelogenous leukemia, non-small cell lung cancers, gastrointestinal stromal tumors, and HER2-positive breast cancers. Given their widespread applications, an increasing frequency of TKI-induced adverse effects has been reported. Although TKIs are known to affect multiple organs in the body including the lungs, liver, gastrointestinal tract, kidneys, thyroid, blood, and skin, cardiac involvement accounts for some of the most serious complications. The most frequently reported cardiovascular side effects range from hypertension, atrial fibrillation, reduced cardiac function, and heart failure to sudden death. The potential mechanisms of these side effects are unclear, leading to critical knowledge gaps in the development of effective therapy and treatment guidelines. There are limited data to infer the best clinical approaches for the early detection and therapeutic modulation of TKI-induced side effects, and universal consensus regarding various management guidelines is yet to be reached. In this state-of-the-art review, we examine multiple pre-clinical and clinical studies and curate evidence on the pathophysiology, mechanisms, and clinical management of these adverse reactions. We expect that this review will provide researchers and allied healthcare providers with the most up-to-date information on the pathophysiology, natural history, risk stratification, and management of emerging TKI-induced side effects in cancer patients.
Collapse
Affiliation(s)
- Sunitha Shyam Sunder
- Cardio-Oncology Research Group, Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Umesh C Sharma
- Division of Cardiovascular Medicine, Jacob's School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Saraswati Pokharel
- Cardio-Oncology Research Group, Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
32
|
Ghafary I, Kim CK, Roth E, Lu M, Taub EM, Lee S, Cohen I, Lu Z. The association of QTc prolongation with cardiovascular events in cancer patients taking tyrosine kinase inhibitors (TKIs). CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:25. [PMID: 37208762 DOI: 10.1186/s40959-023-00178-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
OBJECTIVE To investigate the association between stages of QTc prolongation and the risk of cardiac events among patients on TKIs. METHODS This was a retrospective cohort study performed at an academic tertiary care center of cancer patients who were taking TKIs or not taking TKIs. Patients with two recorded ECGs between January 1, 2009, and December 31, 2019, were selected from an electronic database. The QTc duration > 450ms was determined as prolonged. The association between QTc prolongation progression and events of cardiovascular disease were compared. RESULTS This study included a total of 451 patients with 41.2% of patients taking TKIs. During a median follow up period of 3.1 years, 49.5% subjects developed CVD and 5.4% subjects suffered cardiac death in patient using TKIs (n = 186); the corresponding rates are 64.2% and 1.2% for patients not on TKIs (n = 265), respectively. Among patient on TKIs, 4.8% of subjects developed stroke, 20.4% of subjects suffered from heart failure (HF) and 24.2% of subjects had myocardial infarction (MI); corresponding incidence are 6.8%, 26.8% and 30.6% in non-TKIs. When patients were regrouped to TKIs versus non-TKIs with and without diabetes, there was no significant difference in the incidence of cardiac events among all groups. Adjusted Cox proportional hazards models were applied to estimate hazard ratios (HRs) with 95% confidence intervals (CIs). There is a significant increased risk of HF events (HR, 95% CI: 2.12, 1.36-3.32) and MI events (HR, 95% CI: 1.78, 1.16-2.73) during the 1st visit. There are also trends for an increased incidence of cardiac adverse events associated with QTc prolongation among patient with QTc > 450ms, however the difference is not statistically significant. Increased cardiac adverse events in patients with QTc prolongation were reproduced during the 2nd visit and the incidence of heart failure was significantly associated with QTc prolongation(HR, 95% CI: 2.94, 1.73-5.0). CONCLUSION There is a significant increased QTc prolongation in patients taking TKIs. QTc prolongation caused by TKIs is associated with an increased risk of cardiac events.
Collapse
Affiliation(s)
- Ismail Ghafary
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Chang-Kyung Kim
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Eric Roth
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Michael Lu
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Erin M Taub
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Susan Lee
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Ira Cohen
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Zhongju Lu
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
33
|
Chen Z, Xin Q, Wei W, Wu Y. The pathogenesis and development of targeted drugs in acute T lymphoblastic leukaemia. Br J Pharmacol 2023; 180:1017-1037. [PMID: 36623836 DOI: 10.1111/bph.16029] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Acute lymphoblastic leukaemia (ALL) is mainly classified into acute T- and B-lymphoblastic leukaemia according to the source of its lymphocytes, thymus and bone. Among them, the incidence of adult T-cell accounts for about 25% of adult acute lymphoblastic leukaemia, but the degree of malignancy is high and the treatment rate and prognosis are poor. At this stage, there are few targeted drugs and the commonly used broad-spectrum chemotherapeutic drugs have poor efficacy and many adverse drug reactions. Understanding and investigating the pathogenesis of T-acute lymphoblastic leukaemia is very important for further developing new targeting drugs and improving existing drugs. Dysregulated signalling pathways are the main aetiological factors of T-acute lymphoblastic leukaemia. They play crucial roles in promoting tumour initiation, progression, drug design and therapy responses. This is primarily because signalling pathways are indispensable for many cellular biological processes, including tumour growth, migration, invasion, metastasis and others. As a result, small molecule inhibitors targeting the major kinase components of the signalling pathway have received a lot of attention and have been developed and evaluated in preclinical models and clinical trials. Already marketed drugs are also being repurposed in combination therapies to further improve efficacy and overcome tumour cell resistance. In this review, we have aimed to examine the latest and most classical signalling pathways in the aetiology of T-acute lymphoblastic leukaemia and shed light on potential targets for novel therapeutic agents to act on.
Collapse
Affiliation(s)
- Zhaoying Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Qianling Xin
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Yujing Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Provincial Institute of Translational Medicine, Hefei, China
| |
Collapse
|
34
|
Amrati FEZ, Chebaibi M, Galvão de Azevedo R, Conte R, Slighoua M, Mssillou I, Kiokias S, de Freitas Gomes A, Soares Pontes G, Bousta D. Phenolic Composition, Wound Healing, Antinociceptive, and Anticancer Effects of Caralluma europaea Extracts. Molecules 2023; 28:molecules28041780. [PMID: 36838767 PMCID: PMC9961855 DOI: 10.3390/molecules28041780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Caralluma europaea (Guss.) is an important medicinal plant widely used in Morocco for various traditional purposes. Our work aimed to evaluate the phenolic composition, wound healing, antinociceptive, and anticancer activities of C. europaea extracts. Moreover, this study assessed the beneficial effect of C. europaea phytocompounds on the TRADD, cyclooxegenase-2, Wnt/β-catenin, and tyrosine kinase signaling pathways. The wound healing effect of C. europaea formulations against skin burn was evaluated for 21 days. The cytotoxic effect of the C. europaea extracts was evaluated against human leukemic (K562 and HL60) and liver cancer cell lines (Huh-7) using the MTT test. All the phytoconstituents identified by UHPLC in the polyphenols were docked for their inhibitory power on protein casein kinase-1, glycogen synthase kinase-3-β, cyclooxegenase-2, tyrosine kinase, and TRADD. Luteolin and kaempferol are the main compounds identified in C. europaea polyphenols. The group treated with polyphenols showed the greatest wound contractions and all tested extracts presented a significant antinociceptive effect. Polyphenols showed a remarkable antitumoral activity against the K562, HL60 and Huh-7 cell lines. Saponins exerted an important cytotoxic effect against the Huh-7 cell line, whereas no cytotoxicity was observed for the hydroethanolic and flavonoids extracts. Hesperetin and trimethoxyflavone presented the highest docking G-score on tyrosine kinase and cyclooxygenase, respectively.
Collapse
Affiliation(s)
- Fatima Ez-Zahra Amrati
- Laboratory of Biotechnology, Health, Agrofood and Environment (LBEAS), Faculty of Sciences Dhar El Mehraz, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco
| | - Mohamed Chebaibi
- Biomedical and Translational Research Laboratory, Faculty of Medicine and Pharmacy of the Fez, University of Sidi Mohamed Ben Abdellah, Fez 30000, Morocco
| | - Renata Galvão de Azevedo
- Laboratory of Virology, National Institute of Amazonian Research (INPA), Manaus 69067-375, Brazil
| | - Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, 80131 Naples, Italy
| | - Meryem Slighoua
- Laboratory of Biotechnology, Health, Agrofood and Environment (LBEAS), Faculty of Sciences Dhar El Mehraz, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco
| | - Ibrahim Mssillou
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco
| | - Sotirios Kiokias
- European Research Executive Agency (REA), 1210 Bruxelles, Belgium
- Correspondence: ; Tel.: +32-2-29-57656
| | - Alice de Freitas Gomes
- Laboratory of Virology, National Institute of Amazonian Research (INPA), Manaus 69067-375, Brazil
- Post-Graduate Program in Hematology, School of Health Sciences, University of the State of Amazonas, Manaus 69050-010, Brazil
| | - Gemilson Soares Pontes
- Laboratory of Virology, National Institute of Amazonian Research (INPA), Manaus 69067-375, Brazil
- Post-Graduate Program in Hematology, School of Health Sciences, University of the State of Amazonas, Manaus 69050-010, Brazil
| | - Dalila Bousta
- Laboratory of Biotechnology, Health, Agrofood and Environment (LBEAS), Faculty of Sciences Dhar El Mehraz, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco
| |
Collapse
|
35
|
Tousif S, Singh AP, Umbarkar P, Galindo C, Wheeler N, Coro AT, Zhang Q, Prabhu SD, Lal H. Ponatinib Drives Cardiotoxicity by S100A8/A9-NLRP3-IL-1β Mediated Inflammation. Circ Res 2023; 132:267-289. [PMID: 36625265 PMCID: PMC9898181 DOI: 10.1161/circresaha.122.321504] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND The tyrosine kinase inhibitor ponatinib is the only treatment option for chronic myelogenous leukemia patients with T315I (gatekeeper) mutation. Pharmacovigilance analysis of Food and Drug Administration and World Health Organization datasets has revealed that ponatinib is the most cardiotoxic agent among all Food and Drug Administration-approved tyrosine kinase inhibitors in a real-world scenario. However, the mechanism of ponatinib-induced cardiotoxicity is unknown. METHODS The lack of well-optimized mouse models has hampered the in vivo cardio-oncology studies. Here, we show that cardiovascular comorbidity mouse models evidence a robust cardiac pathological phenotype upon ponatinib treatment. A combination of multiple in vitro and in vivo models was employed to delineate the underlying molecular mechanisms. RESULTS An unbiased RNA sequencing analysis identified the enrichment of dysregulated inflammatory genes, including a multifold upregulation of alarmins S100A8/A9, as a top hit in ponatinib-treated hearts. Mechanistically, we demonstrate that ponatinib activates the S100A8/A9-TLR4 (Toll-like receptor 4)-NLRP3 (NLR family pyrin domain-containing 3)-IL (interleukin)-1β signaling pathway in cardiac and systemic myeloid cells, in vitro and in vivo, thereby leading to excessive myocardial and systemic inflammation. Excessive inflammation was central to the cardiac pathology because interventions with broad-spectrum immunosuppressive glucocorticoid dexamethasone or specific inhibitors of NLRP3 (CY-09) or S100A9 (paquinimod) nearly abolished the ponatinib-induced cardiac dysfunction. CONCLUSIONS Taken together, these findings uncover a novel mechanism of ponatinib-induced cardiac inflammation leading to cardiac dysfunction. From a translational perspective, our results provide critical preclinical data and rationale for a clinical investigation into immunosuppressive interventions for managing ponatinib-induced cardiotoxicity.
Collapse
Affiliation(s)
- Sultan Tousif
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Anand P. Singh
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Prachi Umbarkar
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Cristi Galindo
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA35294-1913, USA
| | - Nicholas Wheeler
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA35294-1913, USA
| | - Angelica Toro Coro
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Qinkun Zhang
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Sumanth D. Prabhu
- Division of Cardiology, Department of Medicine, Washington University in St. Louis
| | - Hind Lal
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
36
|
Mihalcea D, Memis H, Mihaila S, Vinereanu D. Cardiovascular Toxicity Induced by Vascular Endothelial Growth Factor Inhibitors. Life (Basel) 2023; 13:life13020366. [PMID: 36836722 PMCID: PMC9965690 DOI: 10.3390/life13020366] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Cardiotoxicity is an important side effect of vascular endothelial growth factor (VEGF) inhibitors therapy used in the treatment of various malignancies, leading to increased morbidity and mortality. Arterial hypertension, cardiac ischemia with the acceleration of atherosclerosis, arrhythmias, myocardial dysfunction and thromboembolic disease are the most feared cardiovascular adverse reactions due to VEGF inhibitors. Susceptibility for the occurrence of VEGF inhibitors-induced cardiotoxicity has multifactorial determinants, with a significant inter-individual variation. Baseline cardiovascular risk assessment of the patient, type and stage of cancer, dose and duration of VEGF inhibitors treatment and adjuvant chemotherapy or radiotherapy are the main predictors for cardiotoxicity. The role of the cardio-oncology team becomes essential for achieving maximum therapeutic anti-angiogenic effects with minimum cardiovascular side effects. This review will summarize the incidence, risk factors, mechanisms, management and treatment of VEGF inhibitors-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Diana Mihalcea
- Cardiology and Cardiovascular Surgery Department, University of Medicine and Pharmacy Carol Davila, Splaiul Independentei 169, 050098 Bucharest, Romania
- Cardiology Department, University and Emergency Hospital, 050098 Bucharest, Romania
| | - Hayat Memis
- Cardiology and Cardiovascular Surgery Department, University of Medicine and Pharmacy Carol Davila, Splaiul Independentei 169, 050098 Bucharest, Romania
| | - Sorina Mihaila
- Cardiology and Cardiovascular Surgery Department, University of Medicine and Pharmacy Carol Davila, Splaiul Independentei 169, 050098 Bucharest, Romania
- Cardiology Department, University and Emergency Hospital, 050098 Bucharest, Romania
| | - Dragos Vinereanu
- Cardiology and Cardiovascular Surgery Department, University of Medicine and Pharmacy Carol Davila, Splaiul Independentei 169, 050098 Bucharest, Romania
- Cardiology Department, University and Emergency Hospital, 050098 Bucharest, Romania
- Correspondence: ; Tel./Fax: +40-21-3180576
| |
Collapse
|
37
|
Zhou F, Zhu X, Liu Y, Sun Y, Zhang Y, Cheng D, Wang W. Coronary atherosclerosis and chemotherapy: From bench to bedside. Front Cardiovasc Med 2023; 10:1118002. [PMID: 36742069 PMCID: PMC9892653 DOI: 10.3389/fcvm.2023.1118002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular disease, particularly coronary artery disease, is the leading cause of death in humans worldwide. Coronary heart disease caused by chemotherapy affects the prognosis and survival of patients with tumors. The most effective chemotherapeutic drugs for cancer include proteasome inhibitors, tyrosine kinase inhibitors, immune checkpoint inhibitors, 5-fluorouracil, and anthracyclines. Animal models and clinical trials have consistently shown that chemotherapy is closely associated with coronary events and can cause serious adverse cardiovascular events. Adverse cardiovascular events after chemotherapy can affect the clinical outcome, treatment, and prognosis of patients with tumors. In recent years, with the development of new chemotherapeutic drugs, new discoveries have been made about the effects of drugs used for chemotherapy on cardiovascular disease and its related mechanisms, such as inflammation. This review article summarizes the effects of chemotherapeutic drugs on coronary artery disease and its related mechanisms to guide efforts in reducing cardiovascular adverse events during tumor chemotherapy, preventing the development of coronary heart disease, and designing new prevention and treatment strategies for cardiotoxicity caused by clinical tumor chemotherapy.
Collapse
Affiliation(s)
- Fanghui Zhou
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Zhu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yao Liu
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Sun
- Department of Blood and Endocrinology, The 962nd Hospital of the PLA Joint Logistic Support Force, Harbin, Heilongjiang, China
| | - Ying Zhang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | | | - Wei Wang
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,*Correspondence: Wei Wang,
| |
Collapse
|
38
|
Gao C, Zhang L, Xu Y, Ma X, Chen P, Chen ZS, Wei L. I13 overrides resistance mediated by the T315I mutation in chronic myeloid leukemia by direct BCR-ABL inhibition. Front Pharmacol 2023; 14:1183052. [PMID: 37124196 PMCID: PMC10130674 DOI: 10.3389/fphar.2023.1183052] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm caused by a BCR-ABL fusion gene. Imatinib has significantly improved the treatment of CML as a first-generation tyrosine kinase inhibitor (TKIs). The T315I mutant form of BCR-ABL is the most common mutation that confers resistance to imatinib or the second-generation TKIs, resulting in poor clinical prognosis. In this work, we assessed the effect of a potent histone deacetylase (HDAC) inhibitor, I13, on the differentiation blockade in CML cells harboring T315I-mutated and wild-type BCR-ABL by MTT assay, flow cytometery, cell colony formation assay, mRNA Sequencing, Quantitative real-time PCR and Western blotting analysis. We found that I13 possessed highly potent activity against T315I-mutated BCR-ABL mutant-expressing cells and wild-type BCR-ABL-expressing cells. I13 induced cell differentiation and significantly suppressed the proliferation of these CML cells via the cell cycle G0/G1-phase accumulation. Moreover, it was revealed that I13 triggered the differentiation of BaF3-T315I cells, which was attributed to the block of the chronic myeloid leukemia signaling pathway via the depletion of BCR-ABL that was mediated by the inhibition of HDAC activity presented by the acetylation of histones H3 and H4. Taken together, I13 efficiently depleted BCR-ABL in CML cells expressing the BCR-ABL-T315I mutation, which blocked its function, serving as a scaffold protein that modulated the chronic myeloid leukemia signaling pathway mediating cell differentiation. The present findings demonstrate that I13 is a BCR-ABL modulator for the development of CML therapy that can override resistance caused by T315I-mutated BCR-ABL.
Collapse
Affiliation(s)
- Congying Gao
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Lei Zhang
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Yun Xu
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Xiangyu Ma
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Peilei Chen
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
- *Correspondence: Zhe-Sheng Chen, ; Liuya Wei,
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, China
- *Correspondence: Zhe-Sheng Chen, ; Liuya Wei,
| |
Collapse
|
39
|
Dose optimization of tyrosine kinase inhibitor therapy in chronic myeloid leukemia. Int J Hematol 2023; 117:24-29. [PMID: 36087226 DOI: 10.1007/s12185-022-03431-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 01/07/2023]
Abstract
The therapeutic outcomes of chronic myeloid leukemia (CML) have improved dramatically since tyrosine kinase inhibitors (TKIs) became available in clinical practice. Life expectancy of patients with CML is now close to that of the general population. Patients with CML who achieve sustained deep molecular response may discontinue TKI therapy. However, most patients still require TKI therapy for long periods without sustained deep molecular response. Given the awareness of increased incidence of arterial occlusive events in patients on TKI therapy, the optimal TKI selection should be based on age, comorbidities, risk classification, and goals of treatment. Dose optimization of TKI therapy reduces the incidence of adverse events while maintaining efficacy in CML.
Collapse
|
40
|
Ahmed K, Kaddoura R, Yassin MA. A practical guide to managing hypertension, hyperlipidemia, and hyperglycemia in patients with chronic myeloid leukemia. Front Med (Lausanne) 2022; 9:1025392. [PMID: 36569145 PMCID: PMC9774492 DOI: 10.3389/fmed.2022.1025392] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have significantly improved the prognosis of chronic myeloid leukemia (CML) since their approval. Although safe in general, TKIs carry concerns about cardiovascular adverse events. Hypertension, diabetes, and dyslipidemia are among the most common baseline comorbidities among CML patients. Guidelines for the management of the existing comorbidities or those related to TKI therapy are lacking. This paper will review hypertension, hyperglycemia and hyperlipidemia reported in CML patients or associated with TKI therapy and then propose a simple guide on their management.
Collapse
Affiliation(s)
- Khalid Ahmed
- Department of Hematology, National Center for Cancer Care and Research (NCCCR), Hamad Medical Corporation (HMC), Doha, Qatar,*Correspondence: Khalid Ahmed
| | - Rasha Kaddoura
- Pharmacy Department, Heart Hospital, Hamad Medical Corporation (HMC), Doha, Qatar
| | - Mohamed A. Yassin
- Department of Hematology, National Center for Cancer Care and Research (NCCCR), Hamad Medical Corporation (HMC), Doha, Qatar,Mohamed A. Yassin
| |
Collapse
|
41
|
Spetz JKE, Florido MHC, Fraser CS, Qin X, Choiniere J, Yu SJ, Singh R, Friesen M, Rubin LL, Salem JE, Moslehi JJ, Sarosiek KA. Heightened apoptotic priming of vascular cells across tissues and life span predisposes them to cancer therapy-induced toxicities. SCIENCE ADVANCES 2022; 8:eabn6579. [PMID: 36351019 PMCID: PMC9645721 DOI: 10.1126/sciadv.abn6579] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 09/22/2022] [Indexed: 06/16/2023]
Abstract
Although major organ toxicities frequently arise in patients treated with cytotoxic or targeted cancer therapies, the mechanisms that drive them are poorly understood. Here, we report that vascular endothelial cells (ECs) are more highly primed for apoptosis than parenchymal cells across many adult tissues. Consequently, ECs readily undergo apoptosis in response to many commonly used anticancer agents including cytotoxic and targeted drugs and are more sensitive to ionizing radiation and BH3 mimetics than parenchymal cells in vivo. Further, using differentiated isogenic human induced pluripotent stem cell models of ECs and vascular smooth muscle cells (VSMCs), we find that these vascular cells exhibit distinct drug toxicity patterns, which are linked to divergent therapy-induced vascular toxicities in patients. Collectively, our results demonstrate that vascular cells are highly sensitive to apoptosis-inducing stress across life span and may represent a "weakest link" vulnerability in multiple tissues for development of toxicities.
Collapse
Affiliation(s)
- Johan K. E. Spetz
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mary H. C. Florido
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Cameron S. Fraser
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xingping Qin
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jonathan Choiniere
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Stacey J. Yu
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rumani Singh
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Max Friesen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Lee L. Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Joe-Elie Salem
- Clinical Investigation Center Paris-Est, CIC-1901, INSERM, UNICO-GRECO Cardio-Oncology Program, Department of Pharmacology, Pitié-Salpêtrière University Hospital, Sorbonne Université, Paris, France
- Cardio-Oncology Program, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Javid J. Moslehi
- Section of Cardio-Oncology and Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Kristopher A. Sarosiek
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Cancer Center, Boston, MA, USA
| |
Collapse
|
42
|
Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, Boriani G, Cardinale D, Cordoba R, Cosyns B, Cutter DJ, de Azambuja E, de Boer RA, Dent SF, Farmakis D, Gevaert SA, Gorog DA, Herrmann J, Lenihan D, Moslehi J, Moura B, Salinger SS, Stephens R, Suter TM, Szmit S, Tamargo J, Thavendiranathan P, Tocchetti CG, van der Meer P, van der Pal HJH. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J 2022; 43:4229-4361. [PMID: 36017568 DOI: 10.1093/eurheartj/ehac244] [Citation(s) in RCA: 897] [Impact Index Per Article: 448.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
43
|
Clinical features, diagnosis, and management of dasatinib-induced nephrotic syndrome. Invest New Drugs 2022; 40:1153-1159. [PMID: 35867286 DOI: 10.1007/s10637-022-01288-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/18/2022] [Indexed: 12/15/2022]
Abstract
Knowledge of dasatinib-induced nephrotic syndrome is largely based on case reports. The clinical features of dasatinib-induced nephrotic syndrome are unknown. We collected case reports of 25 patients with nephrotic syndrome and analyzed their clinical characteristics. Overall, the onset of nephrotic syndrome ranged from 10 days to 5 years after dasatinib administration. Nine patients (36.0%) had clinical symptoms, mainly periorbital edema and lower-extremity edema. Serum albumin ranged from 1.2 g/dL to 3.7 g/dL in 10 patients (38.5%). The 24-h urine protein values ranged from 3.54 g/day to 118 g/day. Kidney biopsy of 13 patients (52.0%) mainly showed focal foot process effacement, mesangial hyperplasia, endothelial cell damage and focal segmental glomerulosclerosis. Proteinuria resolved or recovered after dasatinib discontinuation or dose reduction or switching to other tyrosine kinase inhibitors (TKIs).
Collapse
|
44
|
Masbuchin AN, Widodo, Rohman MS, Liu PY. The two facets of receptor tyrosine kinase in cardiovascular calcification-can tyrosine kinase inhibitors benefit cardiovascular system? Front Cardiovasc Med 2022; 9:986570. [PMID: 36237897 PMCID: PMC9552878 DOI: 10.3389/fcvm.2022.986570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/29/2022] [Indexed: 01/09/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are widely used in cancer treatment due to their effectiveness in cancer cell killing. However, an off-target of this agent limits its success. Cardiotoxicity-associated TKIs have been widely reported. Tyrosine kinase is involved in many regulatory processes in a cell, and it is involved in cancer formation. Recent evidence suggests the role of tyrosine kinase in cardiovascular calcification, specifically, the calcification of heart vessels and valves. Herein, we summarized the accumulating evidence of the crucial role of receptor tyrosine kinase (RTK) in cardiovascular calcification and provided the potential clinical implication of TKIs-related ectopic calcification. We found that RTKs, depending on the ligand and tissue, can induce or suppress cardiovascular calcification. Therefore, RTKs may have varying effects on ectopic calcification. Additionally, in the context of cardiovascular calcification, TKIs do not always relate to an unfavored outcome-they might offer benefits in some cases.
Collapse
Affiliation(s)
- Ainun Nizar Masbuchin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Widodo
- Department of Biology, Faculty of Mathematics and Natural Science, Universitas Brawijaya, Malang, Indonesia
| | - Mohammad Saifur Rohman
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Ping-Yen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Division of Cardiology, Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
45
|
Involvement of Rho-Associated Coiled-Coil Containing Kinase (ROCK) in BCR-ABL1 Tyrosine Kinase Inhibitor Cardiovascular Toxicity. JACC CardioOncol 2022; 4:371-383. [PMID: 36213346 PMCID: PMC9537085 DOI: 10.1016/j.jaccao.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 10/31/2022] Open
Abstract
Background Second- and third-generation BCR-ABL1 tyrosine kinase inhibitors (TKIs) are associated with cardiovascular adverse events (CVAEs) in patients with Philadelphia chromosome-positive (Ph+) leukemia. Objectives We hypothesized that second- and third-generation BCR-ABL1 TKIs may cause CVAEs through the activation of Rho-associated coiled-coil containing kinase (ROCK). Methods Peripheral blood mononuclear cells from 53 Ph+ patients on TKIs and 15 control patients without Ph+ leukemia were assessed for ROCK activity through capillary electrophoresis (median follow-up = 26 months [Q1-Q3: 5-37 months]). We also investigated the effects of TKIs and ROCK on endothelial dysfunction in vitro, which could contribute to CVAEs. Results Patients receiving second- and third-generation TKIs had 1.6-fold greater ROCK activity compared with patients receiving imatinib and control patients. Elevated ROCK activity was associated with an increased incidence of CVAEs in Ph+ leukemia patients. In endothelial cells in vitro, we found that dasatinib and ponatinib treatment led to changes in actin intensity and endothelial permeability, which can be reversed by pharmacologic inhibition of ROCK. Ponatinib led to decreased cell proliferation, but this was not accompanied by senescence. Dasatinib and ponatinib treatment led to phosphor-inhibition of endothelial nitric oxide synthase and decreased nitric oxide production. ROCK inhibition reversed endothelial permeability and endothelial nitric oxide synthase-related endothelial dysfunction. Imatinib and nilotinib induce phosphorylation of p190RhoGAP. Conclusions Our findings suggest ROCK activity may be a prognostic indicator of CVAEs in patients receiving BCR-ABL1 TKIs. With further study, ROCK inhibition may be a promising approach to reduce the incidence of CVAEs associated with second- and third-generation BCR-ABL1 TKIs.
Collapse
|
46
|
Rong D, Chen X, Xiao J, Liu D, Ni X, Tong X, Wang H. Histone methylation modification patterns and relevant M-RiskScore in acute myeloid leukemia. Heliyon 2022; 8:e10610. [PMID: 36164519 PMCID: PMC9508520 DOI: 10.1016/j.heliyon.2022.e10610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/13/2022] [Accepted: 09/07/2022] [Indexed: 12/05/2022] Open
Abstract
Objective We tried to identify novel molecular subtypes of acute myeloid leukemia (AML) associated with histone methylation and established a relevant scoring system to predict treatment response and prognosis of AML. Methods Gene expression data and clinical characteristics of patients with AML were obtained from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database. Molecular subtyping was carried out by consensus clustering analysis, based on the expression of 24 histone methylation modification regulators (HMMRs). The clinical and biological features of each clustered pattern were taken into account. The scoring system was constructed by using differential expression analysis, Cox regression method and lasso regression analysis. Subsequently, the scoring system in the roles of prognostic and chemotherapeutic prediction of AML were explored. Finally, an independent GSE dataset was used for validating the established clustering system. Results Two distinct subtypes of AML were identified based on the expression of the 24 HMMRs, which exhibited remarkable differences in several clinical and biological characteristics, including HMMRs expression, AML-M0 distribution, NPM1 mutation, tumor mutation burden, somatic mutations, pathway activation, immune cell infiltration and patient survival. The scoring system, M-RiskScore, was established. Integrated analysis demonstrated that patients with the low M-RiskScore displayed a prominent survival advantage and a good response to decitabine treatment, while patients with high M-RiskScore have resistance to decitabine, but they could benefit from IA regimen therapy. Conclusion Detection of HMMRs expression would be a potential strategy for AML subtyping. Meanwhile, targeting histone methylation would be a preferred strategy for either AML-M0 or NPM1 mutant patients. M-RiskScore was a useful prognostic biomarker and a guide for the choice of appropriate chemotherapy strategy.
Collapse
Affiliation(s)
- Dade Rong
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China.,Faculty of Health Sciences, University of Macau, Macau, China
| | - Xiaomin Chen
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.,GenePlus, Beijing, China
| | - Jing Xiao
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Department of Clinical Laboratory, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, China
| | - Daiyuan Liu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Xiangna Ni
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China
| | - Xiuzhen Tong
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| |
Collapse
|
47
|
Dong EE, Bruno MA, Kim JJ, Bernhardt MB, Brown AL, Gramatges MM. Distribution and frequency of tyrosine kinase inhibitor-associated long-term complications in children with chronic myeloid leukemia. Pediatr Blood Cancer 2022; 69:e29786. [PMID: 35593027 DOI: 10.1002/pbc.29786] [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] [Received: 03/11/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) improve outcomes for pediatric malignancies characterized by specific gene rearrangements and mutations; however, little is known about the long-term impact of TKI exposure. Our objective was to assess the incidence and type of late-onset TKI-related toxicities in children with chronic myeloid leukemia (CML). METHODS We reviewed medical records from patients diagnosed with CML between 2006 and 2019 at <21 years of age and prescribed one or more TKIs. Patients treated with stem cell transplant were excluded. Outcomes were captured beginning at 1 year after CML diagnosis. Outcome incidence was described overall and stratified by TKI exposure during the data-capture period. RESULTS Twenty-two eligible TKI-exposed patients with CML were identified. The median follow-up was 6.0 years (range: 2.2-14.3). All pericardial (n = 3) or pleural (n = 3) effusion outcomes occurred in patients treated with TKIs during the data-capture period. Other outcomes included hypertension (n = 2), ectopy on electrocardiogram (n = 2), and gastrointestinal bleed (n = 1). All outcomes were graded as mild to moderate: some resulted in a temporary discontinuation of TKI, but none led to a change in TKI. No differences were noted in outcome incidence by type of TKI exposure. CONCLUSIONS TKIs have substantially improved prognosis for subsets of childhood leukemia, but there are limited long-term data to inform exposure-based risk for late-onset complications and screening. Our results suggest that TKI-exposed survivors may be at risk for long-term outcomes that extend well into survivorship.
Collapse
Affiliation(s)
| | - Michael A Bruno
- Division of Cardiology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Jeffrey J Kim
- Division of Cardiology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Melanie B Bernhardt
- Division of Cancer and Hematology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Austin L Brown
- Division of Cancer and Hematology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Maria M Gramatges
- Division of Cancer and Hematology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
48
|
Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, Boriani G, Cardinale D, Cordoba R, Cosyns B, Cutter DJ, de Azambuja E, de Boer RA, Dent SF, Farmakis D, Gevaert SA, Gorog DA, Herrmann J, Lenihan D, Moslehi J, Moura B, Salinger SS, Stephens R, Suter TM, Szmit S, Tamargo J, Thavendiranathan P, Tocchetti CG, van der Meer P, van der Pal HJH. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J Cardiovasc Imaging 2022; 23:e333-e465. [PMID: 36017575 DOI: 10.1093/ehjci/jeac106] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
49
|
Pandrala M, Bruyneel AAN, Hnatiuk AP, Mercola M, Malhotra SV. Designing Novel BCR-ABL Inhibitors for Chronic Myeloid Leukemia with Improved Cardiac Safety. J Med Chem 2022; 65:10898-10919. [PMID: 35944901 PMCID: PMC9421657 DOI: 10.1021/acs.jmedchem.1c01853] [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] [Indexed: 02/08/2023]
Abstract
Development of tyrosine kinase inhibitors (TKIs) targeting the BCR-ABL oncogene constitutes an effective approach for the treatment of chronic myeloid leukemia (CML) and/or acute lymphoblastic leukemia. However, currently available inhibitors are limited by drug resistance and toxicity. Ponatinib, a third-generation inhibitor, has demonstrated excellent efficacy against both wild type and mutant BCR-ABL kinase, including the "gatekeeper" T315I mutation that is resistant to all other currently available TKIs. However, it is one of the most cardiotoxic of the FDA-approved TKIs. Herein, we report the structure-guided design of a novel series of potent BCR-ABL inhibitors, particularly for the T315I mutation. Our drug design paradigm was coupled to iPSC-cardiomyocyte models. Systematic structure-activity relationship studies identified two compounds, 33a and 36a, that significantly inhibit the kinase activity of both native BCR-ABL and the T315I mutant. We have identified the most cardiac-safe TKIs reported to date, and they may be used to effectively treat CML patients with the T315I mutation.
Collapse
Affiliation(s)
- Mallesh Pandrala
- Department
of Cell, Developmental and Cancer Biology, Center for Experimental
Therapeutics, Knight Cancer Institute, Oregon
Health and Science University, Portland, Oregon 97201, United States
| | - Arne Antoon N. Bruyneel
- Cardiovascular
Institute and Department of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Anna P. Hnatiuk
- Cardiovascular
Institute and Department of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Mark Mercola
- Cardiovascular
Institute and Department of Medicine, Stanford
University, Stanford, California 94305, United States,
| | - Sanjay V. Malhotra
- Department
of Cell, Developmental and Cancer Biology, Center for Experimental
Therapeutics, Knight Cancer Institute, Oregon
Health and Science University, Portland, Oregon 97201, United States,
| |
Collapse
|
50
|
Perpinia AS, Kadoglou N, Vardaka M, Gkortzolidis G, Karavidas A, Marinakis T, Papachrysostomou C, Makaronis P, Vlachou C, Mantzourani M, Farmakis D, Konstantopoulos K. Pharmaceutical Prevention and Management of Cardiotoxicity in Hematological Malignancies. Pharmaceuticals (Basel) 2022; 15:ph15081007. [PMID: 36015155 PMCID: PMC9412591 DOI: 10.3390/ph15081007] [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: 06/14/2022] [Revised: 07/26/2022] [Accepted: 07/30/2022] [Indexed: 11/16/2022] Open
Abstract
Modern treatment modalities in hematology have improved clinical outcomes of patients with hematological malignancies. Nevertheless, many new or conventional anticancer drugs affect the cardiovascular system, resulting in various cardiac disorders, including left ventricular dysfunction, heart failure, arterial hypertension, myocardial ischemia, cardiac rhythm disturbances, and QTc prolongation on electrocardiograms. As these complications may jeopardize the significantly improved outcome of modern anticancer therapies, it is crucial to become familiar with all aspects of cardiotoxicity and provide appropriate care promptly to these patients. In addition, established and new drugs contribute to primary and secondary cardiovascular diseases prevention. This review focuses on the clinical manifestations, preventive strategies, and pharmaceutical management of cardiotoxicity in patients with hematologic malignancies undergoing anticancer drug therapy or hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
| | | | - Maria Vardaka
- Department of Hematology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | | | - Apostolos Karavidas
- Department of Cardiology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | - Theodoros Marinakis
- Department of Hematology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | | | - Panagiotis Makaronis
- Department of Cardiology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | - Charikleia Vlachou
- Department of Hematology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | - Marina Mantzourani
- First Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, “Laiko” General Hospital, 11527 Athens, Greece
| | | | - Konstantinos Konstantopoulos
- Department of Hematology, Medical School, National and Kapodistrian University of Athens, “Laiko” General Hospital, 11527 Athens, Greece
| |
Collapse
|