1
|
Sanagawa A, Takase H. Increased Mitochondrial Superoxide Level Is Partially Associated With Vemurafenib-Induced Renal Tubular Toxicity. Basic Clin Pharmacol Toxicol 2025; 136:e70015. [PMID: 40018909 PMCID: PMC11869354 DOI: 10.1111/bcpt.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Vemurafenib (VEM) reportedly inhibits the mitochondrial respiratory chain and reduces ferrochelatase (FECH) activity, thereby causing VEM-induced renal tubular toxicity. However, the exact mechanisms underlying VEM-induced renal tubular toxicity remain unclear. In this study, we treated human renal proximal tubular epithelial cells with VEM to elucidate these mechanisms. VEM treatment for 24 h resulted in cell damage, reduced cell viability, increased lactate dehydrogenase release and elevated the production of inflammatory cytokines. Transmission electron microscopy (TEM) and fluorescence microscopy revealed accumulation and enlargement of lysosome-derived vacuoles and mitochondrial superoxide production. Although MitoTracker showed no change in the total mitochondrial volume, TEM indicated mitochondrial damage, including smaller and less visible mitochondria. Enhanced superoxide production was confirmed using mtSOX. The mitochondria-specific antioxidant XJB-5-131 partially alleviated VEM-induced superoxide production and improved cell viability, indicating the role of superoxide in VEM-induced renal tubular toxicity. The inhibition of lysosomal acidification by bafilomycin A1 did not mitigate VEM-induced cytotoxicity, suggesting potential autophagy impairment. These findings highlight that mitochondrial dysfunction and lysosomal abnormalities are significant factors in VEM-induced renal tubular toxicity, warranting further investigation into the relationship between their mechanisms, reduced FECH activity and potential renoprotective targets.
Collapse
Affiliation(s)
- Akimasa Sanagawa
- Department of Clinical PharmaceuticsNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Hiroshi Takase
- Research Equipment Sharing CenterNagoya City University Graduate School of Medical SciencesNagoyaJapan
| |
Collapse
|
2
|
Onyiba CI, Kumar NK, Scarlett CJ, Weidenhofer J. Cell Progression and Survival Functions of Enzymes Secreted in Extracellular Vesicles Associated with Breast and Prostate Cancers. Cells 2025; 14:468. [PMID: 40214422 PMCID: PMC11988166 DOI: 10.3390/cells14070468] [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: 02/25/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound cargoes secreted by normal and pathological cells. Through their protein, nucleic acid, and lipid cargoes, EVs mediate several cellular processes, such as cell-cell communication, cell development, immune response, and tissue repair. Most importantly, through their enzyme cargo, EVs mediate pathophysiological processes, including the pathogenesis of cancer. In this review, we enumerate several enzymes secreted in EVs (EV enzyme cargo) from cells and patient clinical samples of breast and prostate cancers and detail their contributions to the progression and survival of both cancers. Findings in this review reveal that the EV enzyme cargo could exert cell progression functions via adhesion, proliferation, migration, invasion, and metastasis. The EV enzyme cargo might also influence cell survival functions of chemoresistance, radioresistance, angiogenesis, cell death inhibition, cell colony formation, and immune evasion. While the current literature provides evidence of the possible contributions of the EV enzyme cargo to the progression and survival mechanisms of breast and prostate cancers, future studies are required to validate that these effects are modified by EVs and provide insights into the clinical applications of the EV enzyme cargo in breast and prostate cancer.
Collapse
Affiliation(s)
- Cosmos Ifeanyi Onyiba
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Niwasini Krishna Kumar
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Christopher J. Scarlett
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Ourimbah, NSW 2258, Australia
| | - Judith Weidenhofer
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
3
|
Munch PV, Nørgaard M, Jensen SK, Birn H, Schmidt H, Christiansen CF. Risk of and Mortality After Acute Kidney Injury Following Cancer Treatment: A Cohort Study. Cancer Med 2025; 14:e70646. [PMID: 39927749 PMCID: PMC11808929 DOI: 10.1002/cam4.70646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) can be a severe complication in cancer patients. However, uncertainty remains regarding the risk of and prognosis after AKI following cancer treatments. We therefore aimed to examine the risk of and mortality after AKI following a wide range of specific cancer treatments, including surgical procedures, anticancer drugs, and hematopoietic stem cell transplantations (HSCTs). METHODS We conducted a nationwide population-based cohort study. We included adult patients receiving cancer treatment in Denmark from 2010 to 2024. We calculated the risk of AKI within 7 days after surgeries, 1 year after initiation of anticancer drugs, and 100 days after HSCTs. Furthermore, we examined the 1-year mortality in patients with and without AKI following cancer treatment. RESULTS We identified 357,870 cancer patients. The 7-day risk of AKI after surgery ranged from 0.3% (breast cancer surgery) to 68.9% (radical nephrectomy in kidney cancer) while the 1-year risk following anticancer drug treatment ranged from 3.5% (cyclophosphamide in breast cancer) to 79.3% (all drugs in acute lymphatic leukemia). The 100-day AKI risk following HSCT ranged from 20.7% (multiple myeloma) to 81.8% (leukemia). For most treatments, AKI was associated with a higher 1-year hazard ratio and risk of death, with exceptions including radical nephrectomy in kidney cancer. CONCLUSION In conclusion, several cancer treatments were associated with a high risk of AKI, and AKI was associated with increased mortality in most treatments. These findings highlight the prognostic value of assessing kidney function following specific cancer treatments in clinical practice.
Collapse
Affiliation(s)
- Philip Vestergaard Munch
- Department of Clinical EpidemiologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Mette Nørgaard
- Department of Clinical EpidemiologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Simon Kok Jensen
- Department of Clinical EpidemiologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Henrik Birn
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Renal MedicineAarhus University HospitalAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Henrik Schmidt
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of OncologyAarhus University HospitalAarhusDenmark
| | - Christian Fynbo Christiansen
- Department of Clinical EpidemiologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
4
|
Dong Y, Wang G, Yan X, Ye W, Qiao X, Deng X, Ren P, Jia C, Chen G, Zheng K, Jiang C, Li X. Ponatinib exacerbate renal injury in systemic lupus erythematosus mouse model through PDGFR-PI3K/AKT pathway. Biochem Pharmacol 2024; 230:116578. [PMID: 39427918 DOI: 10.1016/j.bcp.2024.116578] [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: 05/03/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Lupus nephritis (LN) is a common clinical complication of systemic lupus erythematosus (SLE). Proliferative lupus nephritis represents the gravest form of LN, and since effective drugs for its treatment are still lacking, tyrosine kinase inhibitors (TKIs) find extensive clinical utility due to their notable impact on suppressing cell proliferation and may serve as potential drugs for LN treatment. However, previous studies on the effects of TKI on LN have been controversial. Ponatinib, a third-generation TKI, lacks studies on its role in LN. This study aimed to investigate the impact of the ponatinib on LN. MRL/lpr mice were evaluated for renal function, autoimmune markers and histopathological changes after oral administration of ponatinib. RNA-seq analysis was performed to explore the molecular pathways involved in ponatinib-induced kidney injury. Ponatinib uniquely exacerbated renal damage in MRL/lpr mice, evidenced by a decline in renal function and acute pathological changes, without affecting lupus-related autoimmune markers. Differential expressed genes analysis and functional enrichment implicate ponatinib-induced renal damage in MRL/lpr mice associated with adiponectin. Furthermore, we verified ponatinib signaling the PI3K/AKT pathway through PDGFRα, potentially influencing high molecular weight adiponectin (HMW ADIPOQ) expression and exacerbating renal damage. In conclusion, this study demonstrates that ponatinib can up-regulate HMW ADIPOQ expression via the PI3K/AKT pathway by inhibiting PDGFRα phosphorylation, highlighting the potential nephrotoxic effects of ponatinib in lupus-prone mice, and underscoring the importance of monitoring renal function in systemic autoimmune diseases patients receiving ponatinib.
Collapse
Affiliation(s)
- Yixin Dong
- State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Gangan Wang
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiwei Yan
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wenling Ye
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiangyu Qiao
- State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xingyu Deng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Pengju Ren
- State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chunyu Jia
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Gang Chen
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ke Zheng
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chengyu Jiang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xuemei Li
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Kala J, Joseph T, Pirovano M, Fenoglio R, Cosmai L. Acute Kidney Injury Associated with Anticancer Therapies: Small Molecules and Targeted Therapies. KIDNEY360 2024; 5:1750-1762. [PMID: 39186376 DOI: 10.34067/kid.0000000566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
Molecular targeted therapy has revolutionized cancer treatment by significantly improving patient survival compared with standard conventional chemotherapies. The use of these drugs targets specific molecules or targets, which block growth and spread of cancer cells. Many of these therapies have been approved for use with remarkable success in breast, blood, colorectal, lung, and ovarian cancers. The advantage over conventional chemotherapy is its ability to deliver drugs effectively with high specificity while being less toxic. Although known as "targeted," many of these agents lack specificity and selectivity, and they tend to inhibit multiple targets, including those in the kidneys. The side effects usually arise because of dysregulation of targets of the inhibited molecule in normal tissue. The off-target effects are caused by drug binding to unintended targets. The on-target effects are associated with inhibition toward the pathway reflecting inappropriate inhibition or activation of the intended drug target. Early detection and correct management of kidney toxicities is crucial to preserve kidney functions. The knowledge of these toxicities helps guide optimal and continued utilization of these potent therapies. This review summarizes the different types of molecular targeted therapies used in the treatment of cancer and the incidence, severity, and pattern of nephrotoxicity caused by them, with their plausible mechanism and proposed treatment recommendations.
Collapse
Affiliation(s)
- Jaya Kala
- Division of Nephrology, Department of Internal Medicine, University of Texas Health Science Center-McGovern Medical School, Houston, Texas
| | - Teresa Joseph
- Division of Nephrology, Department of Internal Medicine, University of Texas Health Science Center-McGovern Medical School, Houston, Texas
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Marta Pirovano
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Roberta Fenoglio
- University Center of Excellence on Nephrological, Rheumatological and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) including Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Turin, Italy
- Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley (North-West Italy), San Giovanni Bosco Hub Hospital, ASL Cittàdi Torino, Turin, Italy
- Department of Clinical and Biological Sciences of the University of Turin, Turin, Italy
| | - Laura Cosmai
- Onconephrology Outpatient Clinic, Nephrology and Dialysis Unit, ASST Fatebenefratelli Sacco, Milan, Italy
| |
Collapse
|
6
|
Li Q, Lin J, Hao G, Xie A, Liu S, Tang B. Nephrotoxicity of targeted therapy used to treat lung cancer. Front Immunol 2024; 15:1369118. [PMID: 39026680 PMCID: PMC11254629 DOI: 10.3389/fimmu.2024.1369118] [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: 01/11/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide, especially non-small cell lung cancer. Early diagnosis and better treatment choices have already provided a more promising prognosis for cancer patients. In targeted therapy, antagonists target specific genes supporting cancer growth, proliferation and metastasis. With the incorporation of targeted therapies in routine cancer therapy, it is imperative that the array of toxicities associated with these agents must be well-recognized and managed, especially since these toxicities are distinct from those seen with conventional cytotoxic agents. Drug-related nephrotoxicity has attracted attention when initiating cancer therapy. Our review aims to summarize the adverse renal effects caused by targeted therapy during lung cancer treatment, mainly focusing on EGFR and ALK tyrosine kinase inhibitors. Also, we discuss the possible mechanism of the side effect and provide managements to help improve the renal function in clinical practice.
Collapse
Affiliation(s)
- Qiuling Li
- Department of Nephrology, Blood Purification Center, Zhongshan People’s Hospital, Zhongshan, China
| | - Jieshan Lin
- Department of Nephrology, Blood Purification Center, Zhongshan People’s Hospital, Zhongshan, China
- Department of Nephrology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guojun Hao
- Department of Nephrology, Blood Purification Center, Zhongshan People’s Hospital, Zhongshan, China
| | - Aihua Xie
- Department of Nephrology, Blood Purification Center, Zhongshan People’s Hospital, Zhongshan, China
| | - Shuangxin Liu
- Department of Nephrology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Bin Tang
- Department of Nephrology, Blood Purification Center, Zhongshan People’s Hospital, Zhongshan, China
| |
Collapse
|
7
|
Zhang P, Xu J, Wu Q, Qian J, Wang S. Development of crizotinib-associated renal cyst in a non-small cell lung cancer patient with ALK fusion: a case report and review of the literature. Diagn Pathol 2024; 19:58. [PMID: 38616252 PMCID: PMC11016210 DOI: 10.1186/s13000-024-01480-7] [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: 09/14/2023] [Accepted: 03/21/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Crizotinib, an oral first-generation tyrosine kinase inhibitor (TKI), is superior to systemic chemotherapy for the treatment of non-small cell lung cancer (NSCLC) with positive rearrangement of anaplastic lymphoma kinase (ALK). However, an increased incidence of renal and hepatic cysts has been reported in the patients on crizotinib treatment. CASE PRESENTATION Here, we describe a case of a 71-year-old Chinese women developed multiple cystic lesions in kidney and liver during crizotinib treatment for the primary and metastatic NSCLC. The renal and hepatic cysts were noted by CT scan 3 months after crizotinib treatment, which were spontaneously and significantly regressed after stopping crizotinib. CONCLUSIONS Based on literature review and our experience in this case report, we concluded that crizotinib-associated renal cyst (CARCs) has features of malignancy and abscess in radiographic imaging, and thus, pathological confirmation is necessary to avoid inappropriate treatment decision. In addition, to benefit the patients with progress-free survival (PFS), switching from crizotinib to alectinib is recommended for the treatment of NSCLC patients who developed CARCs.
Collapse
Affiliation(s)
- Peng Zhang
- Six Departments of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - JiaHua Xu
- Seven Departments of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Wu
- Seven Departments of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianxin Qian
- Seven Departments of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Song Wang
- Department of Radiology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
8
|
Czogalla J, Schliffke S, Lu S, Schwerk M, Petereit H, Zhang T, Liu S, Dumoulin B, Gies S, Wu G, Hänzelmann S, Bode M, Grahammer F, Gödel M, Voigtländer M, Butt L, Bokemeyer C, Bergmann C, Benzing T, Wiech T, Puelles VG, Huber TB. Ibrutinib-associated focal segmental glomerulosclerosis and the impact of podocin mutations in chronic lymphocytic leukemia. Kidney Int 2024; 105:877-881. [PMID: 38336226 DOI: 10.1016/j.kint.2024.02.001] [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: 03/29/2023] [Revised: 01/28/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024]
Affiliation(s)
- Jan Czogalla
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany.
| | - Simon Schliffke
- II. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Shun Lu
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Maria Schwerk
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Helena Petereit
- Department II of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Tianran Zhang
- Institute of Medical Systems Biology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Shuya Liu
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Bernhard Dumoulin
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Sydney Gies
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Guochao Wu
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Sonja Hänzelmann
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Institute of Medical Systems Biology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Center for Biomedical AI, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Marlies Bode
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Florian Grahammer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Markus Gödel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Minna Voigtländer
- II. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Linus Butt
- Department II of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Carsten Bokemeyer
- II. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | | | - Thomas Benzing
- Department II of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Thorsten Wiech
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Institute of Pathology, Section for Renal Pathology, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Department of Clinical Medicine, Division of Pathology, Aarhus University, Aarhus, Denmark
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf | UKE, Hamburg, Hamburg, Germany
| |
Collapse
|
9
|
Selamet U, Ahdoot RS, Salasnek R, Abdelnour L, Hanna RM. Onconephrology: mitigation of renal injury in chemotherapy administration. Curr Opin Nephrol Hypertens 2024; 33:257-266. [PMID: 38095483 DOI: 10.1097/mnh.0000000000000960] [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: 01/23/2024]
Abstract
PURPOSE OF REVIEW Onconephrology was first coined as a name for the intersection of cancer medicine and nephrology in the early 2010s. It was recognized then that beyond and understanding of kidney physiology, a new generation of nephrologists skilled in both molecular biology and precision medicine were needed to deal with the challenges of emerging cancer therapies. Stem cell transplants, biologic agents, adjuvants blocking basic cellular signaling pathways, immunotherapy were found to promote novel anticancer outcomes, but also to pose new risks to the kidneys. The field rapidly overlapped with emerging expertise in vascular glomerular disease, glomerular disease, and the same biologic agents now applied to auto immune systemic and kidney diseases. RECENT FINDINGS Many categories of chemotherapeutic agents have been discovered to have adverse renal side effects. In this review, we address classic chemotherapeutic nephrotoxicity and oncologic clinical situations leading to acute kidney injury. We also review the frontiers of nephrotoxicity reported with cell cycle inhibitors, diverse classes of tyrosine kinase inhibitors, immune checkpoint inhibitors, chimeric antigen receptor T-cell therapy, anticancer vaccines, and thrombotic microangiopathies triggered by malignancy and chemotherapy. The aim will be to focus on published strategies to mitigate nephrotoxicity. SUMMARY As onconephrology expands into its own field, it gives birth to new subdisciplines. An understanding that patient populations want the benefits of chemotherapy without the renal (and other) systemic toxicities is emerging. A need to develop a new class of molecular and genetic experts in onconephrology to mitigate nephrotoxicity from chemotherapy is apparent and urgent.
Collapse
Affiliation(s)
- Umut Selamet
- Department of Medical Oncology of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Rebecca S Ahdoot
- Department of Medicine- Division of Nephrology, University of California-Irvine, Irvine
| | - Reed Salasnek
- Department of Medicine- Division of Nephrology, University of California-Irvine, Irvine
| | - Lama Abdelnour
- Department of Medicine-Division of Nephrology, University of California Los Angeles, Los Angeles, CA, USA
| | - Ramy M Hanna
- Department of Medicine- Division of Nephrology, University of California-Irvine, Irvine
| |
Collapse
|
10
|
Silvestris N, Franchina T, Gallo M, Argentiero A, Avogaro A, Cirino G, Colao A, Danesi R, Di Cianni G, D'Oronzo S, Faggiano A, Fogli S, Giuffrida D, Gori S, Marrano N, Mazzilli R, Monami M, Montagnani M, Morviducci L, Natalicchio A, Ragni A, Renzelli V, Russo A, Sciacca L, Tuveri E, Zatelli MC, Giorgino F, Cinieri S. Diabetes management in cancer patients. An Italian Association of Medical Oncology, Italian Association of Medical Diabetologists, Italian Society of Diabetology, Italian Society of Endocrinology and Italian Society of Pharmacology multidisciplinary consensus position paper. ESMO Open 2023; 8:102062. [PMID: 38070434 PMCID: PMC10714217 DOI: 10.1016/j.esmoop.2023.102062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 12/31/2023] Open
Abstract
Cancer management has significantly evolved in recent years, focusing on a multidisciplinary team approach to provide the best possible patient care and address the various comorbidities, toxicities, and complications that may arise during the patient's treatment journey. The co-occurrence of diabetes and cancer presents a significant challenge for health care professionals worldwide. Management of these conditions requires a holistic approach to improve patients' overall health, treatment outcomes, and quality of life, preventing diabetes complications and cancer treatment side-effects. In this article, a multidisciplinary panel of experts from different Italian scientific societies provide a critical overview of the co-management of cancer and diabetes, with an increasing focus on identifying a novel specialty field, 'diabeto-oncology', and suggest new co-management models of cancer patients with diabetes to improve their care. To better support cancer patients with diabetes and ensure high levels of coordinated care between oncologists and diabetologists, 'diabeto-oncology' could represent a new specialized field that combines specific expertise, skills, and training.
Collapse
Affiliation(s)
- N Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina.
| | - T Franchina
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina
| | - M Gallo
- Endocrinology and Metabolic Diseases Unit, AO SS Antonio e Biagio e Cesare Arrigo of Alessandria, Alessandria
| | - A Argentiero
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari
| | - A Avogaro
- Department of Medicine, University of Padova, Padua
| | - G Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples
| | - A Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples; UNESCO Chair, Education for Health and Sustainable Development, Federico II University, Naples
| | - R Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa
| | | | - S D'Oronzo
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro, Bari
| | - A Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome
| | - S Fogli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa
| | - D Giuffrida
- Department of Oncology, Istituto Oncologico del Mediterraneo, Viagrande, Catania
| | - S Gori
- Oncologia Medica, IRCCS Ospedale Don Calabria-Sacro Cuore di Negrar, Verona
| | - N Marrano
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari
| | - R Mazzilli
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome
| | - M Monami
- Diabetology, Careggi Hospital and University of Florence, Firenze
| | - M Montagnani
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, Medical School, University of Bari Aldo Moro, Bari
| | - L Morviducci
- Diabetology and Nutrition Unit, Department of Medical Specialties, ASL Roma 1 - S, Spirito Hospital, Rome
| | - A Natalicchio
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari
| | - A Ragni
- Endocrinology and Metabolic Diseases Unit, AO SS Antonio e Biagio e Cesare Arrigo of Alessandria, Alessandria
| | - V Renzelli
- Diabetologist and Endocrinologist, Italian Association of Medical Diabetologists, Rome
| | - A Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo
| | - L Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania, Catania
| | - E Tuveri
- Diabetology, Endocrinology and Metabolic Diseases Service, ASL-Sulcis, Carbonia
| | - M C Zatelli
- Section of Endocrinology, Geriatrics, and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara
| | - F Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari
| | - S Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| |
Collapse
|
11
|
Bharati J, Shah N, Desai A, Gladstone D, Krushna Das C, Nieto MJ, Jhaveri KD, Izzedine H. Kidney and urinary tract involvement in systemic mastocytosis. Nephrol Dial Transplant 2023; 38:2456-2463. [PMID: 37113073 DOI: 10.1093/ndt/gfad081] [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: 02/18/2023] [Indexed: 04/29/2023] Open
Abstract
Systemic mastocytosis (SM) is a disorder of excessive mast cell accumulation in tissues due to a somatic gain-of-function mutation, commonly in the KIT gene, which prevents apoptosis of mast cells. Whereas bone marrow, skin, lymph nodes, spleen and gastrointestinal tract are commonly involved, kidneys are rarely involved directly by SM. However, there are increasing reports of indirect kidney involvement in patients with SM. Novel anti-neoplastic agents to treat advanced forms of SM include non-specific tyrosine kinase inhibitors, which are reported to be associated with kidney dysfunction in some patients. SM is also associated with immune-mediated glomerulonephritis (GN) such as mesangioproliferative GN, membranous nephropathy and diffuse proliferative GN. Kidney injury, in the form of monoclonal deposition disease and primary light chain amyloidosis, is reported in SM associated with plasma cell dyscrasia. In this narrative review we discuss the various ways kidneys (and the urinary tract) are involved in patients with SM.
Collapse
Affiliation(s)
- Joyita Bharati
- Division of Kidney Diseases and Hypertension, Glomerular Center at Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | | | - Ankuri Desai
- Department of Dermatology, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | - Douglas Gladstone
- Division of Hematology and Oncology, R J Zuckerberg Cancer Institute at Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | - Chandan Krushna Das
- Department of Clinical Hematology and Medical Oncology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Maria Jacqueline Nieto
- Division of Hematology and Oncology, R J Zuckerberg Cancer Institute at Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | - Kenar D Jhaveri
- Division of Kidney Diseases and Hypertension, Glomerular Center at Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | - Hassan Izzedine
- Department of Nephrology, Peupliers Private Hospital, Paris, France
| |
Collapse
|
12
|
Schnuelle P. Renal Biopsy for Diagnosis in Kidney Disease: Indication, Technique, and Safety. J Clin Med 2023; 12:6424. [PMID: 37835066 PMCID: PMC10573674 DOI: 10.3390/jcm12196424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Renal biopsies are the gold standard for diagnosis, staging, and prognosis of underlying parenchymal kidney disease. This article provides an overview of the current indications and highlights ways to reduce bleeding complications in order to achieve optimal diagnostic yield with minimal risk to the patient. Novel indications have emerged from the increasing use of new molecularly targeted oncologic therapies in recent years, which often induce immune-mediated renal disease. On the other hand, the detection of specific antibodies against target antigens on podocytes in the sera of patients with new-onset nephrotic syndrome has now relativized the indication for biopsy in membranous nephropathy. The use of semi-automatic spring-loaded biopsy devices and real-time ultrasound considerably declined the complication rate and is the current standard. Percutaneous renal biopsies are overall a safe procedure if contraindications are considered. A coagulation disorder needs to be excluded beforehand, and an elevated blood pressure must be reduced to the normotensive range with medications. A laparoscopic approach or a radiology interventional procedure through the internal jugular vein may be considered for obtaining a kidney tissue sample if there is an urgent indication and a bleeding tendency cannot be adequately corrected. Major bleeding after a percutaneous renal biopsy can usually be managed with selective arterial embolization of the injured renal vessel. The use of a 16-gauge needle is the most reasonable compromise between diagnostic benefit and risk of complication. In the routine diagnostic, the biopsy specimen is examined with light microscopy, immunohistochemistry, and electron microscopy. Combination with modern molecular pathology techniques will contribute to more precise insights into the development and progression of kidney disease, which will likely refine future treatments in nephrology.
Collapse
Affiliation(s)
- Peter Schnuelle
- Center for Renal Diseases Weinheim, Academic Teaching Practice of the University Medical Center Mannheim, University of Heidelberg, D-69469 Weinheim, Germany
| |
Collapse
|
13
|
Arai H, Yamamoto S, Matsubara T, Miyake T, Tochio A, Mii A, Shimizu A, Minamiguchi S, Muso E, Yanagita M. Focal Segmental Sclerosis Associated with the Novel Multi-tyrosine Kinase Inhibitor Ponatinib. Intern Med 2023; 62:2693-2698. [PMID: 36642524 PMCID: PMC10569927 DOI: 10.2169/internalmedicine.1283-22] [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: 11/14/2022] [Accepted: 12/07/2022] [Indexed: 01/15/2023] Open
Abstract
Ponatinib is a novel multi-tyrosine kinase inhibitor (TKI) with potent inhibitory activity against refractory chronic myeloid leukemia (CML). Despite its high clinical efficacy, ponatinib induces various adverse events due to its multi-target characteristic. However, renal complications associated with ponatinib are rare. A 76-year-old woman had a history of chronic myeloid leukemia (CML) resistant to imatinib and nilotinib. Our patient developed proteinuria and renal function deterioration during treatment with ponatinib but not with imatinib or nilotinib. We herein report the first case of a patient with secondary focal segmental glomerulosclerosis (FSGS) with partial glomerular collapse induced by ponatinib treatment.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
| | - Shinya Yamamoto
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
| | - Takeshi Matsubara
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
| | - Takafumi Miyake
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
| | - Akira Tochio
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
- Department of Nephrology, Japanese Red Cross Society Wakayama Medical Center, Japan
| | - Akiko Mii
- Department of Nephrology, Nippon Medical School, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Japan
| | | | - Eri Muso
- Division of Nephrology and Dialysis, Kitano Hospital, Japan
- Department of Food and Nutrition, Faculty of Contemporary Home Economics, Kyoto Kacho University, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Japan
| |
Collapse
|
14
|
Adegbite BO, Abramson MH, Gutgarts V, Musteata FM, Chauhan K, Muwonge AN, Meliambro KA, Salvatore SP, El Ghaity-Beckley S, Kremyanskaya M, Marcellino B, Mascarenhas JO, Campbell KN, Chan L, Coca SG, Berman EM, Jaimes EA, Azeloglu EU. Patient-Specific Pharmacokinetics and Dasatinib Nephrotoxicity. Clin J Am Soc Nephrol 2023; 18:1175-1185. [PMID: 37382967 PMCID: PMC10564352 DOI: 10.2215/cjn.0000000000000219] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Dasatinib has been associated with nephrotoxicity. We sought to examine the incidence of proteinuria on dasatinib and determine potential risk factors that may increase dasatinib-associated glomerular injury. METHODS We examined glomerular injury through urine albumin-creatinine ratio (UACR) in 82 patients with chronic myelogenous leukemia who were on tyrosine-kinase inhibitor therapy for at least 90 days. t tests were used to compare mean differences in UACR, while regression analysis was used to assess the effects of drug parameters on proteinuria development while on dasatinib. We assayed plasma dasatinib pharmacokinetics using tandem mass spectroscopy and further described a case study of a patient who experienced nephrotic-range proteinuria while on dasatinib. RESULTS Participants treated with dasatinib ( n =32) had significantly higher UACR levels (median 28.0 mg/g; interquartile range, 11.5-119.5) than participants treated with other tyrosine-kinase inhibitors ( n =50; median 15.0 mg/g; interquartile range, 8.0-35.0; P < 0.001). In total, 10% of dasatinib users exhibited severely increased albuminuria (UACR >300 mg/g) versus zero in other tyrosine-kinase inhibitors. Average steady-state concentrations of dasatinib were positively correlated with UACR ( ρ =0.54, P = 0.03) and duration of treatment ( P = 0.003). There were no associations with elevated BP or other confounding factors. In the case study, kidney biopsy revealed global glomerular damage with diffuse foot process effacement that recovered on termination of dasatinib treatment. CONCLUSIONS Exposure to dasatinib was associated with a significant chance of developing proteinuria compared with other similar tyrosine-kinase inhibitors. Dasatinib plasma concentration significantly correlated with higher risk of developing proteinuria while receiving dasatinib. PODCAST This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/CJASN/2023_09_08_CJN0000000000000219.mp3.
Collapse
Affiliation(s)
- Benjamin O. Adegbite
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Internal Medicine, Mount Sinai Morningside/West, New York, New York
| | - Matthew H. Abramson
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Victoria Gutgarts
- Renal Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Florin M. Musteata
- Department of Pharmaceutical Sciences, Albany College of Pharmacy & Health Sciences, Albany, New York
| | - Kinsuk Chauhan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alecia N. Muwonge
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kristin A. Meliambro
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Steven P. Salvatore
- Clinical Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Sebastian El Ghaity-Beckley
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marina Kremyanskaya
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bridget Marcellino
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John O. Mascarenhas
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kirk N. Campbell
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lili Chan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Steven G. Coca
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ellin M. Berman
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Edgar A. Jaimes
- Renal Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Evren U. Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
15
|
Russo G, Barbieri MA, Sorbara EE, Cicala G, Franchina T, Santarpia M, Silvestris N, Spina E. Renal Disorders with Oral Tyrosine Kinase Inhibitors in Metastatic Colorectal Cancer: An Analysis from the FDA Adverse Event Reporting System Database. Biomedicines 2023; 11:2311. [PMID: 37626807 PMCID: PMC10452753 DOI: 10.3390/biomedicines11082311] [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: 07/25/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND this study assessed the nephrotoxicity of regorafenib (REG) and encorafenib (ENC) in metastatic colorectal cancer (mCRC) through an analysis of reports from the US Food and Drug Administration's Adverse Event Reporting System (FAERS) database. METHODS descriptive and disproportional analyses were performed for all reports using ENC and REG as the primary suspect. RESULTS A total of 379 reports had at least one renal adverse drug reaction (ADR), and these ADRs were mainly related to REG (93.1%). Potential safety signals for REG included chromaturia (n = 44; ROR = 12.00, CI 95% = 8.92-16.16; IC = 2.36, IC025-IC075 = 2.06-2.66), hydronephrosis (10; 8.70, 4.67-16.19; 1.85, 1.23-2.47), nephrotic syndrome (7; 5.73, 2.73-12.03; 1.47, 0.73-2.21), renal impairment (53; 4.16, 3.17-5.45; 1.39, 1.12-1.66), dysuria (19; 3.06, 1.95-4.81; 1.06, 0.61-1.52), renal failure (38; 1.66, 1.20-2.28; 0.49, 0.17-0.81), and acute kidney injury (AKI) (43; 1.46, 1.08-1.97; 0.37, 0.07-0.67). For ENC, consistent disproportionalities were observed for AKI (n = 11; ROR = 3.79, CI 95% = 2.09-6.90; IC = 1.32, IC025-IC075 = 0.72-1.91) and dysuria (4; 6.50, 2.43-17.39; 1.86, 0.88-2.85). CONCLUSIONS these findings highlight some not extensively reported renal ADRs that require further investigations to better characterize the safety profiles of REG and ENC in patients with mCRC.
Collapse
Affiliation(s)
- Giulia Russo
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (G.R.); (M.A.B.); (E.E.S.); (G.C.)
| | - Maria Antonietta Barbieri
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (G.R.); (M.A.B.); (E.E.S.); (G.C.)
| | - Emanuela Elisa Sorbara
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (G.R.); (M.A.B.); (E.E.S.); (G.C.)
| | - Giuseppe Cicala
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (G.R.); (M.A.B.); (E.E.S.); (G.C.)
| | - Tindara Franchina
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, 98125 Messina, Italy; (T.F.); (M.S.); (N.S.)
| | - Mariacarmela Santarpia
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, 98125 Messina, Italy; (T.F.); (M.S.); (N.S.)
| | - Nicola Silvestris
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, 98125 Messina, Italy; (T.F.); (M.S.); (N.S.)
| | - Edoardo Spina
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (G.R.); (M.A.B.); (E.E.S.); (G.C.)
| |
Collapse
|
16
|
Vincenti F, Angeletti A, Ghiggeri GM. State of the art in childhood nephrotic syndrome: concrete discoveries and unmet needs. Front Immunol 2023; 14:1167741. [PMID: 37503337 PMCID: PMC10368981 DOI: 10.3389/fimmu.2023.1167741] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Nephrotic syndrome (NS) is a clinical entity characterized by proteinuria, hypoalbuminemia, and peripheral edema. NS affects about 2-7 per 100,000 children aged below 18 years old yearly and is classified, based on the response to drugs, into steroid sensitive (SSNS), steroid dependent, (SDNS), multidrug dependent (MDNS), and multidrug resistant (MRNS). Forms of NS that are more difficult to treat are associated with a worse outcome with respect to renal function. In particular, MRNS commonly progresses to end stage renal failure requiring renal transplantation, with recurrence of the original disease in half of the cases. Histological presentations of NS may vary from minimal glomerular lesions (MCD) to focal segmental glomerulosclerosis (FSGS) and, of relevance, the histological patterns do not correlate with the response to treatments. Moreover, around half of MRNS cases are secondary to causative pathogenic variants in genes involved in maintaining the glomerular structure. The pathogenesis of NS is still poorly understood and therapeutic approaches are mostly based on clinical experience. Understanding of pathogenetic mechanisms of NS is one of the 'unmet needs' in nephrology and represents a significant challenge for the scientific community. The scope of the present review includes exploring relevant findings, identifying unmet needs, and reviewing therapeutic developments that characterize NS in the last decades. The main aim is to provide a basis for new perspectives and mechanistic studies in NS.
Collapse
Affiliation(s)
- Flavio Vincenti
- Division of Nephrology, Department of Medicine and Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Andrea Angeletti
- Nephrology Dialysis and Transplantation, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Gian Marco Ghiggeri
- Nephrology Dialysis and Transplantation, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
17
|
Rahnea-Nita RA, Stoian AR, Anghel RM, Rebegea LF, Ciuhu AN, Bacinschi XE, Zgura AF, Trifanescu OG, Toma RV, Constantin GB, Rahnea-Nita G. The Efficacy of Immunotherapy in Long-Term Survival in Non-Small Cell Lung Cancer (NSCLC) Associated with the Syndrome of Inappropriate Antidiuretic Hormone Secretion (SIADH). Life (Basel) 2023; 13:1279. [PMID: 37374062 DOI: 10.3390/life13061279] [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: 05/08/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION The syndrome of inappropriate antidiuretic hormone secretion (SIADH) is the most common cause of hyponatremia in cancer patients, occurring most frequently in patients with small cell lung cancer. However, this syndrome occurs extremely rarely in patients with non-small cell lung cancer. The results of the clinical trials have revealed that immuno-oncological therapies are effective for long periods of time, providing hope for long survival and with a good quality of life. CASE PRESENTATION We present the case of a female patient who was 62 years old at the time of diagnosis in 2016 who underwent surgery for a right pulmonary tumor (pulmonary adenocarcinoma) and subsequently underwent adjuvant chemotherapy. The patient had a left inoperable mediastinohilar relapse in 2018, which was treated using polychemotherapy The patient also had an occurrence of progressive metastasis and a syndrome of inappropriate antidiuretic hormone secretion (SIADH) in 2019 for which immunotherapy was initiated. The patient has continued with immunotherapy until the time this study began to be written (April 2023), the results being the remission of hyponatremia, the clinical benefits and long-term survival. DISCUSSION The main therapeutic option for SIADH in cancer patients is the treatment of the underlying disease, and its correction depends almost exclusively on a good response to oncological therapy. The initiation of immunotherapy at the time of severe hyponatremia occurrence led to its remission as well as the remission of the other two episodes of hyponatremia, which the patient presented throughout the evolution of the disease, demonstrating an obvious causal relationship between SIADH and the favorable response to immunotherapy. CONCLUSIONS Each patient must be approached individually, taking into account the various particular aspects. Immunotherapy proves to be the innovative treatment that contributes to increasing the survival of patients with metastatic non-small cell lung cancer and to increasing their quality of life.
Collapse
Affiliation(s)
- Roxana-Andreea Rahnea-Nita
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy "Carol Davila", 050474 Bucharest, Romania
- "Sf. Luca" Chronic Disease Hospital, 041915 Bucharest, Romania
| | - Alexandru-Rares Stoian
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy "Carol Davila", 050474 Bucharest, Romania
- "Bagdasar-Arseni" Emergency Clinical Hospital, 041915 Bucharest, Romania
| | - Rodica-Maricela Anghel
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy "Carol Davila", 050474 Bucharest, Romania
- The Oncological Institute "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Laura-Florentina Rebegea
- The Radiotherapy Department, "Sf. Ap. Andrei" County Emergency Clinical Hospital, 800579 Galati, Romania
- The Clinical Department, The Faculty of Medicine and Pharmacy "Dunarea de Jos" University in Galati, 800008 Galati, Romania
- The Research Center in the Field of Medical and Pharmaceutical Sciences, ReFORM-UDJ, 800010 Galati, Romania
| | | | - Xenia-Elena Bacinschi
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy "Carol Davila", 050474 Bucharest, Romania
- The Oncological Institute "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Anca-Florina Zgura
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy "Carol Davila", 050474 Bucharest, Romania
- The Oncological Institute "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Oana-Gabriela Trifanescu
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy "Carol Davila", 050474 Bucharest, Romania
- The Oncological Institute "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Radu-Valeriu Toma
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy "Carol Davila", 050474 Bucharest, Romania
- The Oncological Institute "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Georgiana Bianca Constantin
- The Morphological and Functional Sciences Department, The Faculty of Medicine and Pharmacy, "Dunarea de Jos" University in Galati, 800008 Galati, Romania
| | - Gabriela Rahnea-Nita
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy "Carol Davila", 050474 Bucharest, Romania
- "Sf. Luca" Chronic Disease Hospital, 041915 Bucharest, Romania
| |
Collapse
|
18
|
Adegbite BO, Abramson MH, Gutgarts V, Musteata MF, Chauhan K, Muwonge AN, Meliambro KA, Salvatore SP, Ghaity-Beckley SE, Kremyanskaya M, Marcellino B, Mascarenhas JO, Campbell KN, Chan L, Coca SG, Berman EM, Jaimes EA, Azeloglu EU. Dasatinib nephrotoxicity correlates with patient-specific pharmacokinetics. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.09.23288333. [PMID: 37131844 PMCID: PMC10153335 DOI: 10.1101/2023.04.09.23288333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Introduction Dasatinib has been associated with nephrotoxicity. We sought to examine the incidence of proteinuria on dasatinib and determine potential risk factors that may increase dasatinib-associated glomerular injury. Methods We examine glomerular injury via urine albumin-to-creatinine ratio (UACR) in 101 chronic myelogenous leukemia patients who were on tyrosine-kinase inhibitor (TKI) therapy for at least 90 days. We assay plasma dasatinib pharmacokinetics using tandem mass spectroscopy, and further describe a case study of a patient who experienced nephrotic-range proteinuria while on dasatinib. Results Patients treated with dasatinib (n= 32) had significantly higher UACR levels (median 28.0 mg/g, IQR 11.5 - 119.5) than patients treated with other TKIs (n=50; median 15.0 mg/g, IQR 8.0 - 35.0; p < 0.001). In total, 10% of dasatinib users exhibited severely increased albuminuria (UACR > 300 mg/g) versus zero in other TKIs. Average steady state concentrations of dasatinib were positively correlated with UACR (ρ = 0.54, p = 0.03) as well as duration of treatment ( p =0.003). There were no associations with elevated blood pressure or other confounding factors. In the case study, kidney biopsy revealed global glomerular damage with diffuse foot process effacement that recovered upon termination of dasatinib treatment. Conclusions Exposure to dasatinib is associated a significant chance of developing proteinuria compared to other similar TKIs. Dasatinib plasma concentration significantly correlates with increased risk of developing proteinuria while receiving dasatinib. Screening for renal dysfunction and proteinuria is strongly advised for all dasatinib patients.
Collapse
|
19
|
Vanhoutte T, Sprangers B. Pseudo-AKI associated with targeted anti-cancer agents-the truth is in the eye of the filtration marker. Clin Kidney J 2023; 16:603-610. [PMID: 37007700 PMCID: PMC10061433 DOI: 10.1093/ckj/sfad011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Indexed: 01/18/2023] Open
Abstract
Besides true acute kidney injury (AKI), the occurrence of pseudo-AKI has been associated with several targeted agents. To improve the management of cancer patients treated with targeted agents, we need to be aware of this and use diagnostic approaches to differentiate between pseudo-AKI and AKI. In an article by Wijtvliet et al. in this issue of CKJ, tepotinib is added to the list of targeted agents associated with pseudo-AKI. In this editorial we discuss the current literature regarding pseudo-AKI and true AKI associated with targeted agents, and subsequently propose a management strategy to monitor kidney function in patients treated with targeted agents.
Collapse
Affiliation(s)
- Thomas Vanhoutte
- Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Ben Sprangers
- Ziekenhuis Oost-Limburg, Genk, Belgium
- Biomedical Research Institute, Department of Immunology and Infection, UHasselt, Diepenbeek, Belgium
| |
Collapse
|
20
|
Cheng F, Xu Q, Li Q, Cui Z, Li W, Zeng F. Adverse reactions after treatment with dasatinib in chronic myeloid leukemia: Characteristics, potential mechanisms, and clinical management strategies. Front Oncol 2023; 13:1113462. [PMID: 36814818 PMCID: PMC9939513 DOI: 10.3389/fonc.2023.1113462] [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: 12/01/2022] [Accepted: 01/23/2023] [Indexed: 02/09/2023] Open
Abstract
Dasatinib, a second-generation tyrosine kinase inhibitor, is recommended as first-line treatment for patients newly diagnosed with chronic myeloid leukemia (CML) and second-line treatment for those who are resistant or intolerant to therapy with imatinib. Dasatinib is superior to imatinib in terms of clinical response; however, the potential pulmonary toxicities associated with dasatinib, such as pulmonary arterial hypertension and pleural effusion, may limit its clinical use. Appropriate management of dasatinib-related severe events is important for improving the quality of life and prognosis of patients with CML. This review summarizes current knowledge regarding the characteristics, potential mechanisms, and clinical management of adverse reactions occurring after treatment of CML with dasatinib.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Qiling Xu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Qiang Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Zheng Cui
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Weiming Li
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Weiming Li, ; Fang Zeng,
| | - Fang Zeng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China,*Correspondence: Weiming Li, ; Fang Zeng,
| |
Collapse
|
21
|
Eymael J, van den Broek M, Miesen L, Monge VV, van den Berge BT, Mooren F, Velez VL, Dijkstra J, Hermsen M, Bándi P, Vermeulen M, de Wildt S, Willemsen B, Florquin S, Wetzels R, Steenbergen E, Kramann R, Moeller M, Schreuder MF, Wetzels JF, van der Vlag J, Jansen J, Smeets B. Human scattered tubular cells represent a heterogeneous population of glycolytic dedifferentiated proximal tubule cells. J Pathol 2023; 259:149-162. [PMID: 36373978 PMCID: PMC10107692 DOI: 10.1002/path.6029] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/22/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
Scattered tubular cells (STCs) are a phenotypically distinct cell population in the proximal tubule that increase in number after acute kidney injury. We aimed to characterize the human STC population. Three-dimensional human tissue analysis revealed that STCs are preferentially located within inner bends of the tubule and are barely present in young kidney tissue (<2 years), and their number increases with age. Increased STC numbers were associated with acute tubular injury (kidney injury molecule 1) and interstitial fibrosis (alpha smooth muscle actin). Isolated CD13+ CD24- CD133- proximal tubule epithelial cells (PTECs) and CD13+ CD24+ and CD13+ CD133+ STCs were analyzed using RNA sequencing. Transcriptome analysis revealed an upregulation of nuclear factor κB, tumor necrosis factor alpha, and inflammatory pathways in STCs, whereas metabolism, especially the tricarboxylic acid cycle and oxidative phosphorylation, was downregulated, without showing signs of cellular senescence. Using immunostaining and a publicly available single-cell sequencing database of human kidneys, we demonstrate that STCs represent a heterogeneous population in a transient state. In conclusion, STCs are dedifferentiated PTECs showing a metabolic shift toward glycolysis, which could facilitate cellular survival after kidney injury. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jennifer Eymael
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martijn van den Broek
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pediatric Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Laura Miesen
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Valerie Villacorta Monge
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bartholomeus T van den Berge
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fieke Mooren
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vicky Luna Velez
- Department of Molecular Biology, Radboud Institute for Molecular Life Science, Nijmegen, The Netherlands
| | - Jelmer Dijkstra
- Department of Molecular Biology, Radboud Institute for Molecular Life Science, Nijmegen, The Netherlands
| | - Meyke Hermsen
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Péter Bándi
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Radboud Institute for Molecular Life Science, Nijmegen, The Netherlands
| | - Saskia de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Science, Nijmegen, The Netherlands
| | - Brigith Willemsen
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sandrine Florquin
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Amsterdam Institute for Infection and Immunology, Amsterdam, The Netherlands
| | - Roy Wetzels
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eric Steenbergen
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany.,Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marcus Moeller
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Jack Fm Wetzels
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Nephrology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jitske Jansen
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pediatric Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Bart Smeets
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
22
|
Ishii T, Watanabe T, Higashi T. Differences in the performance of adjuvant chemotherapy between hemodialysis and nonhemodialysis patients. Cancer Med 2023; 12:4033-4041. [PMID: 36128898 PMCID: PMC9972071 DOI: 10.1002/cam4.5258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The survival of hemodialysis (HD) patients with cancer is poor, which may be caused by undertreatment due to renal dysfunction. Particularly, adjuvant chemotherapy after surgery may be considered optional because of its preventive nature. This study investigated the current frequency of administration of adjuvant chemotherapy to HD patients compared with non-HD patients in Japan. METHODS We used data from the Hospital-Based Cancer Registries national database linked to health services utilization data to analyze cases of newly diagnosed colon cancer, gastric cancer, breast cancer, and non-small cell lung cancer (NSCLC) at the stages where adjuvant chemotherapy is generally required. We compared the performance rate of adjuvant chemotherapy and the adjuvant chemotherapy regimens between HD and non-HD patients from October 2011 to December 2017. RESULTS Of the 99,761 patients who underwent curative surgery, 1207 (1%) were HD patients. HD patients received adjuvant chemotherapy less frequently than non-HD patients (24% vs. 63%, p < 0.001). After adjusting for potential confounders, HD remained negatively related to adjuvant chemotherapy administration for all four cancer types. Among all patients who received adjuvant chemotherapy 0(N = 61,873), HD patients were less likely to receive standard regimens and chemotherapy requiring dose adjustment than non-HD patients (88% vs. 95%, p < 0.001 and 92% vs. 98%, p < 0.001, respectively). This trend was particularly pronounced among patients with gastric cancer. CONCLUSIONS HD patients were less likely to receive adjuvant chemotherapy with standard regimens than non-HD patients.
Collapse
Affiliation(s)
- Taisuke Ishii
- Division of Health Services Research, National Cancer Center, Tokyo, Japan
| | - Tomone Watanabe
- Division of Health Services Research, National Cancer Center, Tokyo, Japan
| | - Takahiro Higashi
- Division of Health Services Research, National Cancer Center, Tokyo, Japan
| |
Collapse
|
23
|
Roberto M, Panebianco M, Aschelter AM, Buccilli D, Cantisani C, Caponnetto S, Cortesi E, d’Amuri S, Fofi C, Ierinò D, Maestrini V, Marchetti P, Marignani M, Stigliano A, Vivona L, Santini D, Tomao S. The value of the multidisciplinary team in metastatic renal cell carcinoma: Paving the way for precision medicine in toxicities management. Front Oncol 2023; 12:1026978. [PMID: 36713496 PMCID: PMC9879059 DOI: 10.3389/fonc.2022.1026978] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/07/2022] [Indexed: 01/14/2023] Open
Abstract
The new landscape of treatments for metastatic clear cell renal carcinoma (mRCC) is constantly expanding, but it is associated with the emergence of novel toxicities, adding to up to those observed in the tyrosine-kinase inhibitor (TKI) era. Indeed, the introduction of immune checkpoint inhibitors (ICIs) alone or in combination has been associated with the development of immune-related adverse events (irAEs) involving multiple-organ systems which, even if rarely, had led to fatal outcomes. Moreover, due to the relatively recent addition of ICIs to the previously available treatments, the potential additive adverse effects of these combinations are still unknown. A prompt recognition and management of these toxicities currently represents a fundamental issue in oncology, since it correlates with the outcome of cancer patients. Even if clinical guidelines provide indications for the management of irAEs, no specific protocol to evaluate the individual risk of developing an adverse event during therapy is currently available. A multidisciplinary approach addressing appropriate interventions aimed at reducing the risk of any insidious, severe, and/or dose-limiting toxicity might represent the most efficacious strategy to timely prevent and manage severe irAEs, allowing indirectly to improve both patients' cancer-specific survival and quality of life. In this review, we reported a five-case series of toxicity events that occurred at our center during treatment for mRCC followed by the remarks of physicians from different specialties, pinpointing the relevant role of an integrated and extended multidisciplinary team in a modern model of mRCC patient management.
Collapse
Affiliation(s)
- Michela Roberto
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Martina Panebianco
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, Rome, Italy,*Correspondence: Martina Panebianco,
| | - Anna Maria Aschelter
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Dorelsa Buccilli
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Carmen Cantisani
- Department of Dermatology, Complex Operative Unit (UOC) of Dermatology, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Salvatore Caponnetto
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit B, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Enrico Cortesi
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit B, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Sara d’Amuri
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Claudia Fofi
- Department of Clinical and Molecular Medicine, Nephrology and Dialysis Unit, Sant’ Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Debora Ierinò
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Viviana Maestrini
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Paolo Marchetti
- Scientific Direction, Istituto Dermopatico dell’Immacolata (IDI-IRCCS), Rome, Italy
| | - Massimo Marignani
- Head Liver Disease Section, Digestive and Liver Diseases Department, Sant’ Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Antonio Stigliano
- Department of Clinical and Molecular Medicine, Endocrinology Unit, Sant ‘Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Luca Vivona
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Daniele Santini
- Complex Operative Unit (UOC) Oncologia Medica, Sapienza University, Polo Pontino, Latina, Italy
| | - Silverio Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
24
|
García-Carro C, Draibe J, Soler MJ. Onconephrology: Update in Anticancer Drug-Related Nephrotoxicity. Nephron Clin Pract 2023; 147:65-77. [PMID: 35717937 DOI: 10.1159/000525029] [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: 01/18/2022] [Accepted: 04/24/2022] [Indexed: 12/24/2022] Open
Abstract
The relation that connects cancer and renal damage is bidirectional and this renal damage worsens quality of life and increases morbidity in high-complexity patients such as patients with cancer and kidney injury. Strikingly, in the last decade, the treatment of advanced cancer has clearly advanced in terms of new therapeutic strategies with the ability to transform the advanced metastatic cancer in a chronic condition. In this new era of cancer therapies, cancer treatment including conventional chemotherapy, targeted cancer agents and immunotherapies among others are significantly associated with kidney injury. Renal toxicity that is currently seen in onconephrology departments is in part related to the new therapies such as immunotherapy, and to the prolonged survival achieved at the expense of increasing therapy lines, and a combination of different drugs. In this review, we will discuss in a practical way, nephrotoxicity caused by the main oncospecific treatments such as classical chemotherapy agents, targeted therapies, and immunotherapy. In addition, strategies for prevention and management recommendations in patients with malignancies and kidney disease will also be addressed.
Collapse
Affiliation(s)
- Clara García-Carro
- Nephrology Department, San Carlos Clinical University Hospital, Madrid, Spain
| | - Juliana Draibe
- Nephrology Department, Bellvitge Hospital, Hospitalet Llobregat, Barcelona, Spain
| | - María José Soler
- Nephrology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Research, CSUR National unit of Expertise for Complex Glomerular diseases of Spain, Barcelona, Spain
| |
Collapse
|
25
|
Muacevic A, Adler JR, Deshpande S, Buragamadagu BC, Khanam A, Paravathaneni M, Mulla S, Bedi V, Thota V, Baralo R, Jain A, Choi E, Thirumaran R. Review of Hematology-Oncology Emergencies for Internal Medicine Residents. Cureus 2023; 15:e33563. [PMID: 36779153 PMCID: PMC9908426 DOI: 10.7759/cureus.33563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2023] [Indexed: 01/11/2023] Open
Abstract
The prevalence of cancer continues to grow globally every year. With therapeutic advances over the recent decades, the prevalence of individuals living with cancer continues to increase. Internal medicine residents can see patients admitted to the hospital for cancer-related emergencies. Early identification and appropriate management of these emergencies have been shown to improve mortality and morbidity. In this article, we aim to review the recent updates in the management of commonly encountered oncologic emergencies in the practice of internal medicine residents. This review will cover spinal cord compression, superior vena cava syndrome, tumor lysis syndrome, hypercalcemia, pericardial tamponade, hypoglycemia, hyponatremia, bowel obstruction, increased intracranial pressure, leukostasis, hyperviscosity syndrome, neutropenic fever, and hypersensitivity reactions.
Collapse
|
26
|
Hałka J, Spaleniak S, Kade G, Antosiewicz S, Sigorski D. The Nephrotoxicity of Drugs Used in Causal Oncological Therapies. Curr Oncol 2022; 29:9681-9694. [PMID: 36547174 PMCID: PMC9776938 DOI: 10.3390/curroncol29120760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
In recent years, a dynamic development of oncology has been observed, resulting from the increasingly frequent occurrence of neoplasms and therefore, increasing population of patients. The most effective form of therapy for cancer patients is complex multidisciplinary specialized disease management, including nephro-oncology care. Different forms of renal function impairment are frequently diagnosed in cancer patients. They are caused by different co-morbidities existing before starting the oncologic treatment as well as the direct undesirable effects of this therapy which may cause temporary or irreversible damage of the urinary system-especially kidneys. According to different therapeutic programs, in such cases the degree of renal damage is often crucial for the possibility of further anti-cancer treatment. Medical personnel responsible for delivering care to oncology patients should be properly educated on current methods of prevention and treatment of renal complications resulting from anti-cancer therapy. The development of oncologic medicines design, including especially immuno-oncological agents, obliges us to learn new patomechanisms determining potential adverse effects, including renal complications. This publication is focused on the most important undesirable nephrotoxic effects of the frequently used anti-cancer drugs.
Collapse
Affiliation(s)
- Janusz Hałka
- Department of Clinical Hematology, Warmian-Masurian Cancer Center of the Ministry of the Interior and Administration’s Hospital, Wojska Polskiego 37, 10-228 Olsztyn, Poland
| | - Sebastian Spaleniak
- Department of Internal Diseases and Nephrodiabetology, Medical University of Lodz, Żeromskiego 113, 90-549 Lodz, Poland
- Correspondence:
| | - Grzegorz Kade
- Warmian-Masurian Cancer Center of the Ministry of the Interior and Administration’s Hospital, Wojska Polskiego 37, 10-228 Olsztyn, Poland
| | - Stefan Antosiewicz
- Military Institute of Aviation Medicine, Center of Aeromedical Examination and Occupational Medicine, Zygmunta Krasińskiego 54/56, 01-755 Warsaw, Poland
| | - Dawid Sigorski
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, Aleja Warszawska 30, 11-082 Olsztyn, Poland
- Department of Oncology and Immuno-Oncology, Warmian-Masurian Cancer Center of the Ministry of the Interior and Administration’s Hospital, Wojska Polskiego 37, 10-228 Olsztyn, Poland
| |
Collapse
|
27
|
Abstract
Viruses are intracellular pathogen that exploit host cellular machinery for their propagation. Extensive research on virus-host interaction have shed light on an alternative antiviral strategy that targets host cell factors. Epidermal growth factor receptor (EGFR) is a versatile signal transducer that is involved in a range of cellular processes. Numerous studies have revealed how viruses exploit the function of EGFR in different stages of viral life cycle. In general, viruses attach onto the host cell surface and interacts with EGFR to facilitate viral entry, viral replication and spread as well as evasion from host immunosurveillance. Moreover, virus-induced activation of EGFR signalling is associated with mucin expression, tissue damage and carcinogenesis that contribute to serious complications. Herein, we review our current understanding of roles of EGFR in viral infection and its potential as therapeutic target in managing viral infection. We also discuss the available EGFR-targeted therapies and their limitations.
Collapse
Affiliation(s)
- Kah Man Lai
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Wai Leng Lee
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
28
|
Frazier KS. Kidney Effects by Alternative Classes of Medicines in Patients and Relationship to Effects in Nonclinical Toxicity Studies. Toxicol Pathol 2022; 50:408-414. [PMID: 35608030 DOI: 10.1177/01926233221100414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Drug-induced kidney injury has historically been associated with renal tubule injury related to small molecule pharmaceuticals such as nonsteroidal anti-inflammatory drugs, antineoplastic agents, or antibiotics, but as a greater number of alternative classes of medicines such as biotherapeutics, molecular-targeted antineoplastic drugs, chimeric antigen receptor T-cell therapies, antibody-drug conjugates, oligonucleotide therapies, or other immunomodulatory drugs come to market, the presentation of drug-induced nephrotoxicity is changing. This review article describes the potential rare clinical events in drug-induced kidney injury that might be noted with these new therapies and their potential impact on patients. Potential pathogenic mechanisms related to immunogenicity, immune complex formation, and stimulation of downstream proinflammatory pathways with some of these alternative medicine classes have resulted in the potential for glomerulonephritis, acute interstitial nephritis, renal vasculitis, and other immune-mediated renal disorders in humans. This contrasts with nonclinical toxicity studies, where biologic therapies more often result in vasculitis and glomerulonephritis associated with antidrug antibodies and immunomodulatory pharmacology, and which are not always predictive of clinical effects. While nonclinical antidrug antibody-related renal disease is generally not clinically relevant, other immune-mediated nephrotoxicities associated with immunomodulatory drugs may be predictive of clinical adverse events. Fortunately, these conditions are still rare and account for a small percentage of serious adverse events in kidneys of patients.
Collapse
|
29
|
Thrombotic microangiopathy (TMA) in adult patients with solid tumors: a challenging complication in the era of emerging anticancer therapies. Support Care Cancer 2022; 30:8599-8609. [PMID: 35545722 PMCID: PMC9095052 DOI: 10.1007/s00520-022-06935-5] [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: 10/26/2021] [Accepted: 02/23/2022] [Indexed: 11/02/2022]
Abstract
Thrombotic microangiopathy (TMA) is a syndrome that encompasses a group of disorders defined by the presence of endothelial damage leading to abnormal activation of coagulation, microangiopathic hemolytic anemia and thrombocytopenia, occlusive (micro)vascular dysfunction, and organ damage. TMA may occur in patients with malignancy as a manifestation of cancer-related coagulopathy itself or tumor-induced TMA (Ti-TMA) as a paraneoplastic uncommon manifestation of Trousseau syndrome. TMA can also be triggered by other overlapping conditions such as infections or more frequently as an adverse effect of anticancer drugs (drug-induced TMA or Di-TMA) due to direct dose-dependent toxicity or a drug-dependent antibody reaction. The clinical spectrum of TMA may vary widely from asymptomatic abnormal laboratory tests to acute severe potentially life-threatening forms due to massive microvascular occlusion. While TMA is a rare condition, its incidence may progressively increase within the context of the great development of anticancer drugs and the emerging scenarios in supportive care in cancer. The objective of the present narrative review is to provide a general perspective of the main causes, the key work-up clues that allow clinicians to diagnose and manage TMA in patients with solid tumors who develop anemia and thrombocytopenia due to frequent overlapping causes.
Collapse
|
30
|
Abstract
It has been estimated that nearly 80% of anticancer drug-treated patients receive potentially nephrotoxic drugs, while the kidneys play a central role in the excretion of anticancer drugs. Nephrotoxicity has long been a serious complication that hampers the effectiveness of cancer treatment and continues to influence both mortality and length of hospitalization among cancer patients exposed to either conventional cytotoxic agents or targeted therapies. Kidney injury arising from anticancer drugs tends to be associated with preexisting comorbidities, advanced cancer stage, and the use of concomitant non-chemotherapeutic nephrotoxic drugs. Despite the prevalence and impact of kidney injury on therapeutic outcomes, the field is sorely lacking in an understanding of the mechanisms driving cancer drug-induced renal pathophysiology, resulting in quite limited and largely ineffective management of anticancer drug-induced nephrotoxicity. Consequently, there is a clear imperative for understanding the basis for nephrotoxic manifestations of anticancer agents for the successful management of kidney injury by these drugs. This article provides an overview of current preclinical research on the nephrotoxicity of cancer treatments and highlights prospective approaches to mitigate cancer therapy-related renal toxicity.
Collapse
Affiliation(s)
- Chaoling Chen
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Dengpiao Xie
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Ningjun Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.
| |
Collapse
|
31
|
Vaddepally R, Doddamani R, Sodavarapu S, Madam NR, Katkar R, Kutadi AP, Mathew N, Garje R, Chandra AB. Review of Immune-Related Adverse Events (irAEs) in Non-Small-Cell Lung Cancer (NSCLC)—Their Incidence, Management, Multiorgan irAEs, and Rechallenge. Biomedicines 2022; 10:biomedicines10040790. [PMID: 35453540 PMCID: PMC9027181 DOI: 10.3390/biomedicines10040790] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/08/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of advanced malignancies, including non-small cell lung cancer (NSCLC). These agents have improved clinical outcomes and have become quite an attractive alternative alone or combined with other treatments. Although ICIs are tolerated better, they also lead to unique toxicities, termed immune-related adverse events (irAEs). A reconstituted immune system may lead to dysregulation in normal immune self-tolerance and cause inflammatory side effects (irAEs). Although any organ system can be affected, immune-related adverse events most commonly involve the gastrointestinal tract, endocrine glands, skin, and liver. They can occur anytime during the treatment course and rarely even after completion. Owen and colleagues showed that approximately 30% of patients with NSCLC treated with ICIs develop irAEs. Kichenadasse et al. conducted a thorough evaluation of multiorgan irAEs, which is of particular interest because information regarding these types of irAEs is currently sparse. It is important to delineate between infectious etiologies and symptom progression during the management of irAEs. Close consultation with disease-specific subspecialties is encouraged. Corticosteroids are the mainstay of treatment of most irAEs. Early intervention with corticosteroids is crucial in the general management of immune-mediated toxicity. Grade 1–2 irAEs can be closely monitored; hypothyroidism and other endocrine irAEs may be treated with hormone supplementation without the need for corticosteroid therapy. Moderate- to high-dose steroids and other additional immunosuppressants such as tocilizumab and cyclophosphamide might be required in severe, grade 3–4 cases. Recently, increasing research on irAEs after immunotherapy rechallenge has garnered much attention. Dolladille and colleagues assessed the safety in patients with cancer who resumed therapy with the same ICIs and found that rechallenge was associated with about 25–30% of the same irAEs experienced previously (4). However, such data should be carefully considered. Further pooled analyses may be required before we conclude about ICIs’ safety in rechallenge.
Collapse
Affiliation(s)
- Raju Vaddepally
- Yuma Regional Medical Center, 2400 S Avenue A, Yuma, AZ 85364, USA; (N.R.M.); (R.K.); (A.P.K.); (N.M.); (A.B.C.)
- Correspondence:
| | - Rajiv Doddamani
- Slidell Memorial Hospital, 1001 Gause Blvd, Slidell, LA 70458, USA;
| | - Soujanya Sodavarapu
- San Joaquin General Hospital, 500 W Hospital Road, French Camp, CA 95231, USA;
| | - Narasa Raju Madam
- Yuma Regional Medical Center, 2400 S Avenue A, Yuma, AZ 85364, USA; (N.R.M.); (R.K.); (A.P.K.); (N.M.); (A.B.C.)
| | - Rujuta Katkar
- Yuma Regional Medical Center, 2400 S Avenue A, Yuma, AZ 85364, USA; (N.R.M.); (R.K.); (A.P.K.); (N.M.); (A.B.C.)
| | - Anupama P. Kutadi
- Yuma Regional Medical Center, 2400 S Avenue A, Yuma, AZ 85364, USA; (N.R.M.); (R.K.); (A.P.K.); (N.M.); (A.B.C.)
| | - Nibu Mathew
- Yuma Regional Medical Center, 2400 S Avenue A, Yuma, AZ 85364, USA; (N.R.M.); (R.K.); (A.P.K.); (N.M.); (A.B.C.)
| | - Rohan Garje
- Department of Internal Medicine-Hematology/Oncology, University of Iowa, Iowa, IA 52242, USA;
| | - Abhinav B. Chandra
- Yuma Regional Medical Center, 2400 S Avenue A, Yuma, AZ 85364, USA; (N.R.M.); (R.K.); (A.P.K.); (N.M.); (A.B.C.)
| |
Collapse
|
32
|
Shirali AC, Sprangers B. Cancer Drug Dosing in Chronic Kidney Disease and Dialysis. Adv Chronic Kidney Dis 2022; 29:208-216.e1. [PMID: 35817528 DOI: 10.1053/j.ackd.2021.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/20/2021] [Accepted: 12/08/2021] [Indexed: 11/11/2022]
Abstract
Patients with malignancies have a high prevalence of kidney disease and are often treated with antineoplastic agents that undergo kidney metabolism or excretion or clearance via renal replacement therapies. Thus, the dosing of these agents, including classic chemotherapeutic drugs, targeted therapies, and immunotherapy, must take into account patients' kidney function. In this review, we will discuss the pitfalls of accurate measurement of kidney function and how kidney disease affects both pharmacodynamic and pharmacokinetic properties of drugs. Lastly, we will discuss specific agents and summarize current dosing strategies for use in patients with chronic kidney disease and end-stage kidney disease.
Collapse
Affiliation(s)
- Anushree C Shirali
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ben Sprangers
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium; Division of Nephrology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
33
|
Bonilla M, Jhaveri KD, Izzedine H. Anaplastic lymphoma kinase inhibitors and their effect on the kidney. Clin Kidney J 2022; 15:1475-1482. [PMID: 35892021 PMCID: PMC9308093 DOI: 10.1093/ckj/sfac062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Indexed: 11/30/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality and approximately 5% of non–small-cell lung cancer (NSCLC) patients are positive for anaplastic lymphoma kinase (ALK) gene rearrangement or fusion with echinoderm microtubule-associated protein-like 4. ALK inhibitors are the mainstay treatment for patients with NSCLC harboring a rearrangement of the ALK gene or the ROS1 oncogenes. With the recent publication of pivotal trials leading to the approval of these compounds in different indications, their toxicity profile warrants an update. Several ALK-1 inhibitors are used in clinical practice, including crizotinib, ceritinib and alectinib. According to the package insert and published literature, treatment with several ALK-1 inhibitors appears to be associated with the development of peripheral edema and rare electrolyte disorders, kidney failure, proteinuria and an increased risk for the development and progression of renal cysts. This review introduces the different types of ALK inhibitors, focusing on their detailed kidney-related side effects in clinical practice.
Collapse
Affiliation(s)
- Marco Bonilla
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra Northwell, 100 Community Drive, Great Neck, NY, USA
| | - Kenar D Jhaveri
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra Northwell, 100 Community Drive, Great Neck, NY, USA
| | - Hassan Izzedine
- Department of Nephrology, Peupliers Private Hospital, Ramsay Générale de Santé, Paris, France
| |
Collapse
|
34
|
Korkmaz M, Hendem E, Karakurt Eryılmaz M, Demirkıran A, Karaağaç M, Artaç M. Nephrotic syndrome induced by cetuximab in a patient with metastatic colorectal cancer. J Oncol Pharm Pract 2022; 28:998-1002. [PMID: 35023411 DOI: 10.1177/10781552211073790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Cetuximab, an anti-EGFR monoclonal antibody, often cause skin toxicity, most commonly acneiform rash. We present a rare case of glomerulonephritis associated with cetuximab therapy. CASE REPORT A 58-year-old male patient recently completed cetuximab-based chemotherapy for metastatic colorectal adenocarcinoma. He presented with acute renal failure, anasarca edema and nephrotic proteinuria. The amount of protein in the 24-h urine test was over 15.6 grams. MANAGEMENT & OUTCOME The patient showed a dramatic improvement in renal function shortly after terminated of cetuximab therapy without immunosuppressive therapy. DISCUSSION Therefore, drugs targeting epidermal growth factor receptor (EGFR) monoclonal antibody were thought to trigger nephrotic syndrome by causing glomerular damage. As a result, physicians using EGFR monoclonal inhibitors should be very careful about renal functions and proteinuria in patients.
Collapse
Affiliation(s)
- Mustafa Korkmaz
- Department of Medical Oncology, 64222Necmettin Erbakan University School of Medicine, Konya, Turkey
| | - Engin Hendem
- Department of Medical Oncology, 64222Necmettin Erbakan University School of Medicine, Konya, Turkey
| | - Melek Karakurt Eryılmaz
- Department of Medical Oncology, 64222Necmettin Erbakan University School of Medicine, Konya, Turkey
| | - Aykut Demirkıran
- Department of Medical Oncology, 64222Necmettin Erbakan University School of Medicine, Konya, Turkey
| | - Mustafa Karaağaç
- Department of Medical Oncology, 64222Necmettin Erbakan University School of Medicine, Konya, Turkey
| | - Mehmet Artaç
- Department of Medical Oncology, 64222Necmettin Erbakan University School of Medicine, Konya, Turkey
| |
Collapse
|
35
|
Chen YW, Yang M, Wang MX, Jiang JH, Jiang DY, Chen ZL, Yang L. Refractory hypokalemia caused by cetuximab with advanced colorectal cancer patients: the case series and literature review. Anticancer Drugs 2022; 33:e789-e794. [PMID: 34419963 PMCID: PMC8670339 DOI: 10.1097/cad.0000000000001212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 07/19/2021] [Indexed: 01/20/2023]
Abstract
Cetuximab is the first-line treatment for advanced metastatic colon cancer. But cetuximab can cause electrolyte disturbances, including hypomagnesemia and hypokalemia. Among them, hypokalemia is often caused by hypomagnesemia, not directly caused by cetuximab. This article reports two cases of refractory hypokalemia caused by cetuximab without hypomagnesemia. The two patients had no abnormalities in serum potassium before cetuximab treatment. The occurrence of hypokalemia was clearly correlated with the cetuximab, and they were significantly improved after stopping or reducing the dose. At the same time, the appearance of hypokalemia is significantly related to the efficacy of cetuximab. They have received 37 and 35 cycles of cetuximab-related therapy, with condition stable periods of 12.8 and 15.1 months, respectively. Obviously, our report refutes the above view. In our opinion, hypokalemia, a side effect of cetuximab, may be directly caused by it, rather than secondary to hypomagnesemia. Similar to hypomagnesemia, the appearance of hypokalemia often indicates a better curative effect of cetuximab.
Collapse
Affiliation(s)
- Yun-Wang Chen
- The Qingdao University Medical College, Qingdao, Shandong
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
| | - Min Yang
- The Qingdao University Medical College, Qingdao, Shandong
| | - Ming-Xing Wang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
- Graduate School of Clinical Medicine, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Jia-Hong Jiang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
| | - Ding-Yi Jiang
- The Qingdao University Medical College, Qingdao, Shandong
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
| | - Zhe-Ling Chen
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
| | - Liu Yang
- The Qingdao University Medical College, Qingdao, Shandong
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
| |
Collapse
|
36
|
Kim MJ. Nilotinib-Induced Acute Interstitial Nephritis during the Treatment of Chronic Myeloid Leukemia. KOSIN MEDICAL JOURNAL 2021. [DOI: 10.7180/kmj.2021.36.2.153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are targeted therapy drugs that selectively inhibit protein kinases. Nephrotoxicity associated with TKIs is uncommon. We report a case of a 39-year-old man with acute kidney injury that developed after nilotinib treatment for chronic myeloid leukemia (CML). The renal function of the patient decreased during treatment with nilotinib but improved when treatment was discontinued due to neutropenia. However, the renal function of the patient deteriorated again with the reintroduction of nilotinib for treatment. A renal biopsy revealed acute interstitial nephritis (AIN). The patient had no history of comorbidities and medication causing renal injury. Finally, we diagnosed the patient with nilotinib-induced AIN. After switching to imatinib mesylate, the renal function of the patient stabilized without further deterioration. Our case indicates that nilotinib can be a potential cause of renal dysfunction by inducing AIN when renal function deteriorates in patients treated with nilotinib.
Collapse
|
37
|
Crosnier A, Abbara C, Cellier M, Lagarce L, Babin M, Bourneau-Martin D, Briet M. Renal Safety Profile of EGFR Targeted Therapies: A Study from VigiBase ® the WHO Global Database of Individual Case Safety Reports. Cancers (Basel) 2021; 13:5907. [PMID: 34885014 PMCID: PMC8657199 DOI: 10.3390/cancers13235907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/12/2021] [Accepted: 11/19/2021] [Indexed: 01/22/2023] Open
Abstract
Kidney EGFR expression together with reported cases of glomerular diseases in the context of anti-EGFR drug administration raise concerns about the renal safety profile of these drugs. This issue is addressed in a case/non-case study carried out on VigiBase®, the WHO global database of individual case safety reports (ICRS). Disproportionality analysis of renal adverse effects related to the selected anti-EGFR drugs, erlotinib, gefitinib, afatinib, osimertinib, cetuximab and panitumumab, was assessed using the reporting odds ratio (ROR). Nine hundred and eighty-nine ICRSs were included. A signal of disproportionate reporting (SDR) was found for afatinib (ROR = 2.70; 95% CI [2.22-3.29]) and erlotinib (ROR = 1.73; 95% CI [1.46-2.04]) with acute kidney injury, and for afatinib (ROR = 2.41; 95% CI [1.78-3.27]), cetuximab (ROR = 1.42; 95% CI [1.14-1.78]) and erlotinib (ROR = 2.23; 95% CI [1.80-2.77]) with renal failure. The preferred term "diarrhoea" was frequently reported in the included cases. An SDR was found for erlotinib with haemolytic and uremic syndrome (ROR = 4.01; 95% CI [1.80-8.94]) and thrombotic microangiopathy (ROR = 4.94; 95% CI [2.80-8.72]). No SDR was seen for glomerular or tubule-interstitial diseases. This study showed that the anti-EGFR drug renal toxicity is mainly related to renal failure in the context of digestive toxicity.
Collapse
Affiliation(s)
- Alexandre Crosnier
- Department of Pharmacology-Toxicology and Pharmacovigilance, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France; (A.C.); (C.A.); (M.C.); (L.L.); (M.B.); (D.B.-M.)
- Département de Médecine, Faculté de Médecine d’Angers, Université d’Angers, 49035 Angers, France
| | - Chadi Abbara
- Department of Pharmacology-Toxicology and Pharmacovigilance, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France; (A.C.); (C.A.); (M.C.); (L.L.); (M.B.); (D.B.-M.)
| | - Morgane Cellier
- Department of Pharmacology-Toxicology and Pharmacovigilance, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France; (A.C.); (C.A.); (M.C.); (L.L.); (M.B.); (D.B.-M.)
| | - Laurence Lagarce
- Department of Pharmacology-Toxicology and Pharmacovigilance, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France; (A.C.); (C.A.); (M.C.); (L.L.); (M.B.); (D.B.-M.)
| | - Marina Babin
- Department of Pharmacology-Toxicology and Pharmacovigilance, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France; (A.C.); (C.A.); (M.C.); (L.L.); (M.B.); (D.B.-M.)
| | - Delphine Bourneau-Martin
- Department of Pharmacology-Toxicology and Pharmacovigilance, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France; (A.C.); (C.A.); (M.C.); (L.L.); (M.B.); (D.B.-M.)
| | - Marie Briet
- Department of Pharmacology-Toxicology and Pharmacovigilance, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France; (A.C.); (C.A.); (M.C.); (L.L.); (M.B.); (D.B.-M.)
- Département de Médecine, Faculté de Médecine d’Angers, Université d’Angers, 49035 Angers, France
- Research Institute MitoVasc, UMR CNRS 6214 INSERM 1083, University of Angers, 49100 Angers, France
| |
Collapse
|
38
|
Niitsu T, Hayashi T, Uchida J, Yanase T, Tanaka S, Kuroyama M, Ueno K. Drug-Induced Kidney Injury Caused by Osimertinib: Report of a Rare Case. Nephron Clin Pract 2021; 146:58-63. [PMID: 34569520 DOI: 10.1159/000518774] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/28/2021] [Indexed: 11/19/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) that target the epidermal growth factor receptor (EGFR) have shown highly favourable outcomes in patients with advanced-stage non-small-cell lung cancer (NSCLC). The adverse effects of EGFR-TKIs are generally less severe than those of conventional cytotoxic therapies. We report a patient with NSCLC who presented with acute kidney injury associated with biopsy-proven acute tubular injury during osimertinib treatment and whose renal function recovered after reducing the osimertinib dose. A 61-year-old male smoker complained of dyspnoea on exertion for 1 month before his visit to the medical centre. He was diagnosed with lung adenocarcinoma of the left lower lobe (cT4N3M1a, stage IVA) and was positive for an EGFR mutation (exon 19 deletion). Osimertinib was initiated at 80 mg/day. At treatment initiation, the patient's serum creatinine level was 0.64 mg/dL, with microscopic haematuria; by day 83, this level had increased to 1.33 mg/dL, with proteinuria. On day 83, we reduced the osimertinib dose to 40 mg/day and performed a kidney biopsy on day 98. The histological diagnosis was tubular injury with IgA deposition. Based on the clinical course and histological findings, we speculated that the kidney injury was associated with osimertinib. After dose reduction, the patient's serum creatinine level decreased to 1.07 mg/dL, and proteinuria disappeared. He maintained a partial response for >6 months after osimertinib administration. We report the first case of biopsy-proven mild IgA deposition, crescent formation, and tubular injury probably caused by osimertinib and demonstrate how reducing the osimertinib dose could strike a balance between its anti-cancer efficacy and adverse effects.
Collapse
Affiliation(s)
- Takayuki Niitsu
- Department of Respiratory Medicine, Osaka General Medical Center, Osaka, Japan
| | - Terumasa Hayashi
- Department of Kidney Disease and Hypertension, Osaka General Medical Center, Osaka, Japan
| | - Junji Uchida
- Department of Respiratory Medicine, Osaka General Medical Center, Osaka, Japan
| | - Takafumi Yanase
- Department of Respiratory Medicine, Osaka General Medical Center, Osaka, Japan
| | - Satoshi Tanaka
- Department of Respiratory Medicine, Osaka General Medical Center, Osaka, Japan
| | - Munenori Kuroyama
- Department of Respiratory Medicine, Osaka General Medical Center, Osaka, Japan
| | - Kiyonobu Ueno
- Department of Respiratory Medicine, Osaka General Medical Center, Osaka, Japan
| |
Collapse
|
39
|
Pullen RL. Renal cell carcinoma, part 3. Nursing 2021; 51:30-38. [PMID: 34463651 DOI: 10.1097/01.nurse.0000769804.33935.a1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Renal cell carcinoma (RCC) accounts for most renal malignancies. This article, the last in a three-part series, presents treatment options for RCC using the American Joint Committee on Cancer Tumor, Node, and Metastasis staging system as a framework, as well as nursing-care options for patients undergoing partial or radical nephrectomy.
Collapse
Affiliation(s)
- Richard L Pullen
- Richard L. Pullen is a professor of nursing at Texas Tech University Health Sciences Center School of Nursing in Lubbock, Tex., and a member of the Nursing2021 editorial board
| |
Collapse
|
40
|
Kala J, Salman LA, Geara AS, Izzedine H. Nephrotoxicity From Molecularly Targeted Chemotherapeutic Agents. Adv Chronic Kidney Dis 2021; 28:415-428.e1. [PMID: 35190108 DOI: 10.1053/j.ackd.2021.09.003] [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: 05/26/2021] [Revised: 08/16/2021] [Accepted: 09/01/2021] [Indexed: 11/11/2022]
Abstract
The introduction of novel molecularly targeted therapies in the last 2 decades has significantly improved the patient survival compared to standard conventional chemotherapies. However, this improvement has been accompanied by a whole new spectrum of kidney adverse events. Although known as "targeted," many of these agents lack specificity and selectivity, and they have a tendency to inhibit multiple targets including those in the kidneys. Early detection and correct management of kidney toxicities is crucial to preserve kidney functions. The knowledge of these toxicities helps guide optimal and continued utilization of these potent therapies. The incidence, severity, and pattern of nephrotoxicity may vary depending on the respective target of the drug. Here, we review the mechanism of action, clinical findings of kidney adverse events, and their proposed management strategies.
Collapse
|
41
|
BRAF/MEK inhibitor-associated nephrotoxicity in a real-world setting and human kidney cells. Anticancer Drugs 2021; 32:1076-1083. [PMID: 34232935 DOI: 10.1097/cad.0000000000001106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Acute kidney injury (AKI) associated with cancer chemotherapy can be life-threatening. Inhibitors of rapidly accelerated fibrosarcoma kinase B (BRAF)-mutants and mitogen-activated extracellular signal-regulated kinase (MEK) administered as combination therapy are effective against BRAF-mutant melanoma, but drug-associated AKI events were reported after marketing. Here, we examined the nephrotoxicity of two BRAF inhibitors, vemurafenib and dabrafenib, and two MEK inhibitors, cobimetinib and trametinib, in a real-world setting and human kidney cells. Target drug-associated AKI signals were detected by reporting odds ratio (ROR) derived from report data in the Food and Drug Administration Adverse Events Reporting System database. In-vitro cytotoxicity was evaluated in proximal renal tubular epithelial cells (RPTEC), glomerular endothelial cells (GEnC), and glomerular epithelial cells (GEpC). RESULTS AKI RORs associated with vemurafenib [ROR, 3.28; confidence interval (CI), 2.91-3.69] and cobimetinib (ROR, 4.40; CI, 3.55-5.45) were higher than those associated with dabrafenib (ROR, 1.35; CI, 1.15-1.60) and trametinib (ROR, 1.32; CI, 1.11-1.56). Vemurafenib reduced cell viability and increased cell death in RPTEC and GEpC at 10 μM, which was below the mean maximum concentration in blood under steady-state condition [115.7 μM (56.7 μg/mL)]. No vemurafenib-associated cytotoxicity was detected in GEnC. Mean maximum concentrations of cobimetinib, dabrafenib and trametinib did not induce cell death. CONCLUSION This work revealed that vemurafenib had stronger cytotoxic effects on tubular and glomerular epithelial cells than the other BRAF and MEK inhibitors. Hence, we recommend careful monitoring for clinical signs of kidney injury in patients treated with vemurafenib.
Collapse
|
42
|
Valério P, Barreto JP, Ferreira H, Chuva T, Paiva A, Costa JM. Thrombotic microangiopathy in oncology - a review. Transl Oncol 2021; 14:101081. [PMID: 33862523 PMCID: PMC8065296 DOI: 10.1016/j.tranon.2021.101081] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/31/2022] Open
Abstract
Thrombotic microangiopathy is a syndrome triggered by a wide spectrum of situations, some of which are specific to the Oncology setting. It is characterized by a Coombs-negative microangiopathic haemolytic anemia, thrombocytopenia and organ injury, with characteristic pathological features, resulting from platelet microvascular occlusion. TMA is rare and its cancer-related subset even more so. TMA triggered by drugs is the most common within this group, including classic chemotherapy and the latest targeted therapies. The neoplastic disease itself and hematopoietic stem-cell transplantation could also be potential triggers. Evidence-based medical guidance in the management of cancer-related TMA is scarce and the previous knowledge about primary TMA is valuable to understand the disease mechanisms and the potential treatments. Given the wide spectrum of potential causes for TMA in cancer patients, the aim of this review is to gather the vast information available. For each entity, pathophysiology, clinical features, therapeutic approaches and prognosis will be covered.
Collapse
Affiliation(s)
- Patrícia Valério
- Nephrology Department, Setúbal Hospital Center, Portugal Rua Camilo Castelo Branco 175, 2910-549 Setúbal, Portugal.
| | - João Pedro Barreto
- Laboratory Diagnosis Department, Portuguese Oncology Institute of Porto, Portugal Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Hugo Ferreira
- Nephrology Department, Portuguese Oncology Institute of Porto, Portugal Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Teresa Chuva
- Nephrology Department, Portuguese Oncology Institute of Porto, Portugal Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Ana Paiva
- Nephrology Department, Portuguese Oncology Institute of Porto, Portugal Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - José Maximino Costa
- Nephrology Department, Portuguese Oncology Institute of Porto, Portugal Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| |
Collapse
|
43
|
Manzari MT, Shamay Y, Kiguchi H, Rosen N, Scaltriti M, Heller DA. Targeted drug delivery strategies for precision medicines. NATURE REVIEWS. MATERIALS 2021; 6:351-370. [PMID: 34950512 PMCID: PMC8691416 DOI: 10.1038/s41578-020-00269-6] [Citation(s) in RCA: 460] [Impact Index Per Article: 115.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/24/2020] [Indexed: 05/05/2023]
Abstract
Progress in the field of precision medicine has changed the landscape of cancer therapy. Precision medicine is propelled by technologies that enable molecular profiling, genomic analysis, and optimized drug design to tailor treatments for individual patients. Although precision medicines have resulted in some clinical successes, the use of many potential therapeutics has been hindered by pharmacological issues, including toxicities and drug resistance. Drug delivery materials and approaches have now advanced to a point where they can enable the modulation of a drug's pharmacological parameters without compromising the desired effect on molecular targets. Specifically, they can modulate a drug's pharmacokinetics, stability, absorption, and exposure to tumours and healthy tissues, and facilitate the administration of synergistic drug combinations. This Review highlights recent progress in precision therapeutics and drug delivery, and identifies opportunities for strategies to improve the therapeutic index of cancer drugs, and consequently, clinical outcomes.
Collapse
Affiliation(s)
- Mandana T. Manzari
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- These authors have contributed equally to this work
| | - Yosi Shamay
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- These authors have contributed equally to this work
| | - Hiroto Kiguchi
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- These authors have contributed equally to this work
| | - Neal Rosen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Maurizio Scaltriti
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel A. Heller
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
44
|
Bridoux F, Cockwell P, Glezerman I, Gutgarts V, Hogan JJ, Jhaveri KD, Joly F, Nasr SH, Sawinski D, Leung N. Kidney injury and disease in patients with haematological malignancies. Nat Rev Nephrol 2021; 17:386-401. [PMID: 33785910 DOI: 10.1038/s41581-021-00405-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) is common in patients with cancer, especially in those with haematological malignancies. Kidney injury might be a direct consequence of the underlying haematological condition. For example, in the case of lymphoma infiltration or extramedullary haematopoiesis, it might be caused by a tumour product; in the case of cast nephropathy it might be due to the presence of monoclonal immunoglobulin; or it might result from tumour complications, such as hypercalcaemia. Kidney injury might also be caused by cancer treatment, as many chemotherapeutic agents are nephrotoxic. High-intensity treatments, such as high-dose chemotherapy followed by haematopoietic stem cell transplantation, not only increase the risk of infection but can also cause AKI through various mechanisms, including viral nephropathies, engraftment syndrome and sinusoidal obstruction syndrome. Some conditions, such as thrombotic microangiopathy, might also result directly from the haematological condition or the treatment. Novel immunotherapies, such as immune checkpoint inhibitors and chimeric antigen receptor T cell therapy, can also be nephrotoxic. As new therapies for haematological malignancies with increased anti-tumour efficacy and reduced toxicity are developed, the number of patients receiving these treatments will increase. Clinicians must gain a good understanding of the different mechanisms of kidney injury associated with cancer to better care for these patients.
Collapse
Affiliation(s)
- Frank Bridoux
- Department of Nephrology, and Centre d'Investigation Clinique (CIC INSERM 1402), Centre Hospitalier Universitaire et Université de Poitiers, Poitiers, France.,CNRS, UMR7276, Limoges, France.,Centre de référence Amylose AL et autres maladies par dépôt d'immunoglobulines monoclonales, Poitiers, France
| | - Paul Cockwell
- Department of Nephrology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Ilya Glezerman
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Victoria Gutgarts
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Jonathan J Hogan
- Renal, Electrolyte and Hypertension Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kenar D Jhaveri
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, NY, USA
| | - Florent Joly
- Department of Nephrology, and Centre d'Investigation Clinique (CIC INSERM 1402), Centre Hospitalier Universitaire et Université de Poitiers, Poitiers, France
| | - Samih H Nasr
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Deirdre Sawinski
- Renal, Electrolyte and Hypertension Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nelson Leung
- Division of Nephrology and Hypertension, Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
45
|
McSweeney KR, Gadanec LK, Qaradakhi T, Ali BA, Zulli A, Apostolopoulos V. Mechanisms of Cisplatin-Induced Acute Kidney Injury: Pathological Mechanisms, Pharmacological Interventions, and Genetic Mitigations. Cancers (Basel) 2021; 13:1572. [PMID: 33805488 PMCID: PMC8036620 DOI: 10.3390/cancers13071572] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Administration of the chemotherapeutic agent cisplatin leads to acute kidney injury (AKI). Cisplatin-induced AKI (CIAKI) has a complex pathophysiological map, which has been linked to cellular uptake and efflux, apoptosis, vascular injury, oxidative and endoplasmic reticulum stress, and inflammation. Despite research efforts, pharmaceutical interventions, and clinical trials spanning over several decades, a consistent and stable pharmacological treatment option to reduce AKI in patients receiving cisplatin remains unavailable. This has been predominately linked to the incomplete understanding of CIAKI pathophysiology and molecular mechanisms involved. Herein, we detail the extensively known pathophysiology of cisplatin-induced nephrotoxicity that manifests and the variety of pharmacological and genetic alteration studies that target them.
Collapse
|
46
|
Alonso F, Auñón P, Cavero T, Salgueira M, Praga M, Quiroga B, de Francisco ÁLM, Macía M. Monographic consultation of onconephrology. Rationale and implementation. Nefrologia 2021; 41:154-164. [PMID: 36165376 DOI: 10.1016/j.nefroe.2021.04.006] [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: 01/11/2020] [Accepted: 08/06/2020] [Indexed: 06/16/2023] Open
Abstract
The increase in demand for medical care for renal complications associated with neoplastic diseases is a reality in most nephrology departments. In response to this overall situation, the creation of healthcare models such as monographic consultations and develop training programs in Onconephrology could improve the care of these patients. Through an exploratory and descriptive study, we identified current situation of kidney involvement in cancer patients. The objective of the present study is to establish the criteria for specific assistance in the field of Onconephrology. For this, we have reviewed key aspects and analyzed the current situation in our country, through a survey addressed to all nephrologists through the Spanish Society of Nephrology., together with the experience of two Spanish centers. From this information, we have established some requirements and recommendations for the start-up of these consultations.
Collapse
Affiliation(s)
| | - Pilar Auñón
- Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Teresa Cavero
- Hospital Universitario Doce de Octubre, Madrid, Spain
| | | | - Manuel Praga
- Hospital Universitario Doce de Octubre, Madrid, Spain
| | | | - Ángel L M de Francisco
- Hospital Universitario Marqués de Valdecilla, Universidad de Cantabria, Santander, Cantabria, Spain
| | - Manuel Macía
- Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| |
Collapse
|
47
|
Workeneh BT, Uppal NN, Jhaveri KD, Rondon-Berrios H. Hypomagnesemia in the Cancer Patient. KIDNEY360 2020; 2:154-166. [PMID: 35368816 PMCID: PMC8785729 DOI: 10.34067/kid.0005622020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/11/2020] [Indexed: 02/04/2023]
Abstract
Hypomagnesemia is a common medical problem that contributes to the morbidity and mortality of patients with cancer. This review summarizes magnesium physiology and highlights the mechanisms underlying magnesium disturbances due to cancer and cancer treatment. The causes of hypomagnesemia can be categorized according to the pathophysiologic mechanism: decreased intake, transcellular shift, gastrointestinal losses, and kidney losses. Patients with cancer are at risk for opportunistic infections, frequently experience cardiovascular complications, and often receive classes of medications that cause or exacerbate hypomagnesemia. Also, cancer-specific therapies are responsible for hypomagnesemia, including platinum-based chemotherapy, anti-EGF receptor mAbs, human EGF receptor-2 target inhibitors (HER2), and calcineurin inhibitors. Urinary indices, such as the fractional excretion of magnesium, can provide useful information about the etiology. The management of hypomagnesemia depends on the magnitude of hypomagnesemia and the underlying cause. We recommended checking serum magnesium at the beginning of treatment and as part of routine monitoring throughout cancer treatment. Opportunities exist for potential research and practice improvement, including further characterization of hypomagnesemia regarding the clinical effect on cancer outcomes, preventing hypomagnesemia in patients receiving high-risk anticancer agents, and developing effective therapeutic strategies.
Collapse
Affiliation(s)
- Biruh T. Workeneh
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nupur N. Uppal
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York
| | - Kenar D. Jhaveri
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York
| | - Helbert Rondon-Berrios
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
48
|
Krishnamurthy A, Bhattacharya S, Lathia T, Kantroo V, Kalra S, Dutta D. Anticancer Medications and Sodium Dysmetabolism. EUROPEAN ENDOCRINOLOGY 2020; 16:122-130. [PMID: 33117443 DOI: 10.17925/ee.2020.16.2.122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
Abstract
Therapeutic advances have revolutionised cancer treatment over the last two decades, but despite improved survival and outcomes, adverse effects to anticancer therapy such as dyselectrolytaemias do occur and need to be managed appropriately. This review explores essential aspects of sodium homeostasis in cancer with a focus on alterations arising from anticancer medications. Sodium and water balance are tightly regulated by close interplay of stimuli arising from hypothalamic osmoreceptors, arterial and atrial baroreceptors and the renal juxtaglomerular apparatus. This delicate balance can be disrupted by cancer itself, as well as the medications used to treat it. Some of the conventional chemotherapeutics, such as alkylating agents and platinum-based drugs, can cause hyponatraemia and, on rare occasions, hypernatraemia. Other conventional agents such as vinca alkaloids, as well as newer targeted cancer therapies including small molecule inhibitors and monoclonal antibodies, can cause hyponatraemia, usually as a result of inappropriate antidiuretic hormone secretion. Hyponatraemia can also sometimes occur secondarily to drug-induced hypocortisolism or salt-wasting syndromes. Another atypical but distinct mechanism for hyponatraemia is via pituitary dysfunction induced by immune checkpoint inhibitors. Hypernatraemia is uncommon and occasionally ensues as a result of drug-induced nephrogenic diabetes insipidus. Identification of the aetiology and appropriate management of these conditions, in addition to averting treatment-related problems, can be lifesaving in critical situations.
Collapse
Affiliation(s)
- Aishwarya Krishnamurthy
- Endocrinology Department, Max Super Speciality Hospital, Patparganj, New Delhi, Delhi, India
| | - Saptarshi Bhattacharya
- Endocrinology Department, Max Super Speciality Hospital, Patparganj, New Delhi, Delhi, India
| | - Tejal Lathia
- Endocrinology Department, Fortis Hospital, Vashi, Navi Mumbai, Maharashtra, India
| | - Viny Kantroo
- Respiratory Department, Critical Care and Sleep Medicine, Apollo Hospitals, Sarita Vihar, New Delhi, Delhi, India
| | - Sanjay Kalra
- Endocrinology Department, Bharti Hospital, Karnal, Haryana, India
| | - Deep Dutta
- CEDAR Superspeciality Clinics, Dwarka, New Delhi, Delhi, India
| |
Collapse
|
49
|
Milluzzo A, Vigneri P, Martorana F, Vigneri R, Sciacca L. Type 2 diabetes and cancer: problems and suggestions for best patient management. EXPLORATION OF MEDICINE 2020. [DOI: 10.37349/emed.2020.00013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Diabetes and cancer are widespread worldwide and the number of subjects presenting both diseases increased over the years. The management of cancer patients having diabetes represents a challenge not only because of the complexity and heterogeneity of these pathologies but also for the lack of standardised clinical guidelines. The diagnosis of cancer is traumatizing and monopolizes the attention of both patients and caregivers. Thus, pre-existent or new-onset diabetes can be overshadowed thus increasing the risk for short- and long-term adverse events. Moreover, drugs used for each disease can interfere with the clinical course of the concomitant disease, making challenging the management of these patients. Over the years, this issue has become more relevant because of the increased patients’ life expectancy due to the improved efficacy of diabetes and cancer therapies.
The purpose of this review is to highlight what is known and what should be taken into consideration to optimise the clinical management of patients with diabetes and cancer. Due to the complexity of these diseases, a multidisciplinary, shared approach, including all the protagonists involved, is necessary to improve patients’ quality of life and lifespan.
Collapse
Affiliation(s)
- Agostino Milluzzo
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, 95122 Catania, Italy
| | - Paolo Vigneri
- Center of Experimental Oncology and Hematology, Department of Clinical and Experimental Medicine, University of Catania, A.O.U. Policlinico-Vittorio Emanuele, 95124 Catania, Italy
| | - Federica Martorana
- Center of Experimental Oncology and Hematology, Department of Clinical and Experimental Medicine, University of Catania, A.O.U. Policlinico-Vittorio Emanuele, 95124 Catania, Italy
| | - Riccardo Vigneri
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, 95122 Catania, Italy; Institute of Crystallography, Catania Section, National Research Council, CNR, 95126 Catania, Italy
| | - Laura Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, 95122 Catania, Italy
| |
Collapse
|
50
|
Mossoba ME, Sprando RL. In Vitro to In Vivo Concordance of Toxicity Using the Human Proximal Tubule Cell Line HK-2. Int J Toxicol 2020; 39:452-464. [PMID: 32723106 DOI: 10.1177/1091581820942534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The renal proximal tubule cell line, human kidney 2 (HK-2), recapitulates many of the functional cellular and molecular characteristics of differentiated primary proximal tubule cells. These features include anchorage dependence, gluconeogenesis capability, and sodium-dependent sugar transport. In order to ascertain how well HK-2 cells can reliably reveal the toxicological profile of compounds having a potential to cause proximal tubule injury in vivo, we sought to evaluate the effects of known proximal tubule toxicants using the HK-2 cell line. We selected 20 pure nephrotoxic compounds that included chemotherapeutic drugs, antibiotics, and heavy metal-containing compounds and evaluated their ability to induce HK-2 cell injury relative to 10 innocuous pure compounds or cell culture media alone. We performed a comprehensive set of in vitro cellular toxicological assays to evaluate cell viability, oxidative stress, mitochondrial integrity, and a specific biomarker of renal injury, Kidney Injury Molecule 1. For each of our selected compounds, we were able to establish a reproducible profile of toxicological outcomes. We compared our results to those described in peer-reviewed publications to understand how well the HK-2 cellular model agrees with overall in vivo rat or human toxicological outcomes. This study begins to address the question of how well in vitro data generated with HK-2 cells can mirror in vivo animal and human outcomes.
Collapse
Affiliation(s)
- Miriam E Mossoba
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, 4137US Food and Drug Administration, Laurel, MD, USA
| | - Robert L Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, 4137US Food and Drug Administration, Laurel, MD, USA
| |
Collapse
|