1
|
Guan C, Chen R, Wang Y. Inflammatory markers mediate association of AIP with kidney failure risk: data from National Health and Nutrition Examination Survey (NHANES) 2005-2018. Ren Fail 2025; 47:2486565. [PMID: 40230193 PMCID: PMC12001854 DOI: 10.1080/0886022x.2025.2486565] [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: 07/25/2024] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/16/2025] Open
Abstract
Dyslipidemia and inflammation often coexist in the progression of kidney failure, with the atherosclerosis index of plasma (AIP) serving as a valuable marker for monitoring dyslipidemia. This cross-sectional study analyzed data from the National Health and Nutrition Examination Survey (NHANES) spanning 2005 to 2018, involving a total of 10,358 participants. AIP was calculated as the logarithmic ratio (base 10) of triglycerides to high-density lipoprotein cholesterol (log10[TG/HDL-C]), while kidney failure was assessed through self-reported physician diagnosis. Logistic regression models and restricted cubic splines (RCS) were utilized to examine the association between AIP and the risk of kidney failure, with additional subgroup analyses performed to explore potential interactions. Mediation analyses were conducted to investigate whether inflammatory markers mediated the relationship between AIP and kidney failure. In logistic regression, after adjusting for all covariates, AIP was found to be positively associated with the risk of kidney failure [OR = 1.74 (95% CI: 1.04-2.92)], and a linear relationship between AIP and kidney failure risk was observed (P-non-linear = 0.4050). Mediation analysis revealed that segmented neutrophils, eosinophils, and monocytes partially mediated the association between AIP and kidney failure, with mediation proportions of 19.65%, 2.44%, and 7.25%, respectively. These findings suggest that Higher AIP was associated with an increased risk of kidney failure, with segmented neutrophils, eosinophils, and monocytes serving as partial mediators. The results provide valuable insights into the role of inflammation in kidney failure and highlight potential avenues for its prevention.
Collapse
Affiliation(s)
- Chengjing Guan
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ruixue Chen
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yu Wang
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
2
|
Kommer A, Stortz M, Kraus D, Weinmann-Menke J. Immune Checkpoint Inhibitor-Associated Acute Kidney Injury: A Single-Center Experience of Biopsy-Proven Cases. J Clin Med 2025; 14:3231. [PMID: 40364262 PMCID: PMC12072658 DOI: 10.3390/jcm14093231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/26/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Immune checkpoint inhibitor therapy (ICI) has greatly changed cancer therapy in recent years. The main side effects are immune-related adverse events (irAEs) that can affect any organ system. With the widespread use of ICIs, even rare irAEs, like acute kidney injury due to ICI-induced nephritis (ICI-AKI), have become a more common complication. Methods: All ICI-treated patients who underwent a kidney biopsy for AKI at a single academic center between January 2020 and December 2023 were analyzed. Results: We identified twelve cases of biopsy-proven ICI-AKI. The median follow up was 11.5 months. All cases showed acute interstitial nephritis (AIN) on the biopsy. Melanoma was the most common cancer, and dual-checkpoint inhibition with Ipilimumab and Nivolumab was the most common regimen. Extrarenal irAEs were present in only 25% of cases. Two-thirds had concomitant medication with proton pump inhibitors (PPIs). Only four patients completely recovered their kidney function, and one patient remained on kidney replacement therapy. Conclusions: AIN is a common cause of AKI in ICI-treated cancer patients. Although they respond well to steroid treatment, full restitution of kidney function occurs in less than half of the subjects. As ICIs are increasingly used in cancer management, more research on the prevention and treatment of ICI-associated AKI is needed.
Collapse
Affiliation(s)
- Andreas Kommer
- Department of Internal Medicine I, University Medical Center of Johannes Gutenberg University, 55131 Mainz, Germany; (M.S.); (D.K.); (J.W.-M.)
| | | | | | | |
Collapse
|
3
|
Moledina DG, Shelton K, Menez S, Aklilu AM, Yamamoto Y, Kadhim BA, Shaw M, Kent C, Makhijani A, Hu D, Simonov M, O’Connor K, Bitzel J, Thiessen-Philbrook H, Wilson FP, Parikh CR. External Validation of an Electronic Health Record-Based Diagnostic Model for Histological Acute Tubulointerstitial Nephritis. J Am Soc Nephrol 2025; 36:859-868. [PMID: 39500309 PMCID: PMC12059105 DOI: 10.1681/asn.0000000556] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/29/2024] [Indexed: 05/02/2025] Open
Abstract
Key Points Individual noninvasive diagnostic tests lack accuracy for diagnosing histological acute tubulointerstitial nephritis. A validated diagnostic model combining four clinical tests accurately predicted acute tubulointerstitial nephritis on biopsy in two separate populations. Background Accurate diagnosis of acute tubulointerstitial nephritis (AIN) often requires a kidney biopsy. We previously developed a diagnostic statistical model for predicting biopsy-confirmed AIN by combining four laboratory tests after evaluating over 150 potential predictors from the electronic health record. In this study, we validate this diagnostic model in two biopsy-based cohorts at Johns Hopkins Hospital (JHH) and Yale University, which were geographically and temporally distinct from the development cohort, respectively. Methods We analyzed patients who underwent kidney biopsy at JHH and Yale University (2019–2023). We assessed discrimination (area under receiver-operating characteristics curve [AUC]) and calibration using previously derived model coefficients and recalibrated the model using an intercept correction factor that accounted for differences in baseline prevalence of AIN between development and validation cohorts. Results We included 1982 participants: 1454 at JHH and 528 at Yale. JHH (5%) and Yale (17%) had lower proportions of biopsies with AIN than the development set (23%). The AUC was 0.73 (95% confidence interval [CI], 0.66 to 0.79) at JHH and 0.73 (95% CI, 0.67 to 0.78) at Yale, similar to the development set (0.73 [95% CI, 0.64 to 0.81]). Calibration was imperfect in validation cohorts, particularly at JHH, but improved with the application of an intercept correction factor. The model increased AUC of clinicians’ prebiopsy suspicion for AIN by 0.10 to 0.77 (95% CI, 0.71 to 0.82). Conclusions An AIN diagnostic model retained discrimination in two validation cohorts but needed recalibration to account for local AIN prevalence. The model improved clinicians’ ability to predict AIN.
Collapse
Affiliation(s)
- Dennis G. Moledina
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Kyra Shelton
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Steven Menez
- Division of Nephrology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Abinet M. Aklilu
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Yu Yamamoto
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Bashar A. Kadhim
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Melissa Shaw
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Candice Kent
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Amrita Makhijani
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - David Hu
- Division of Nephrology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Michael Simonov
- Department of Biostatistics (Health Informatics), Yale School of Public Health, New Haven, Connecticut
| | - Kyle O’Connor
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jack Bitzel
- Division of Nephrology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - F. Perry Wilson
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Chirag R. Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
4
|
Karam S, Ali A, Fung W, Mehta P, Nair S, Anandh U. Acute Kidney Injury Associated with Novel Anticancer Therapies: Immunotherapy. KIDNEY360 2025; 6:652-662. [PMID: 39992727 PMCID: PMC12045508 DOI: 10.34067/kid.0000000749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
The landscape of cancer survival has been positively affected by the introduction and dissemination of immunotherapy with the wide usage of immune checkpoint inhibitors and chimeric antigen receptors cell therapies. The success of these novel therapies can, however, be limited to a certain extent by systemic inflammatory toxicities affecting, directly or indirectly, the kidney. In the case of immune checkpoint inhibitors, severe acute interstitial nephritis is the main adverse event and can lead to permanent discontinuation of the therapy. In turn, chimeric antigen receptor cell therapy can cause cytokine release syndrome and immune effector cell-associated hemophagocytic lympho-histiocytosis, with kidney damage through various mechanisms, and be life threatening. Prompt diagnosis and management of these entities is essential to preserve kidney function and ensure the best possible kidney and overall outcomes to patients with cancer.
Collapse
Affiliation(s)
- Sabine Karam
- Division of Nephrology and Hypertension, Department of Medicine, University of Minnesota, Minneapolis, Minneapolis
- Division of Nephrology and Hypertension, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Ala Ali
- Nephrology and Renal Transplantation Center, The Medical City, Baghdad, Iraq
| | - Winston Fung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Prashant Mehta
- Department of Hematology, Medical Oncology and Bone Marrow Transplantation, Amrita Institute of Medical Sciences and Research Centre, Faridabad, India
| | - Sanjeev Nair
- Department of Nephrology, Madras Medical Mission, Chennai, India
| | - Urmila Anandh
- Department of Nephrology, Amrita Institute of Medical Sciences and Research Centre, Faridabad, India
| |
Collapse
|
5
|
Baker ML, Cantley LG. Adding insult to injury: the spectrum of tubulointerstitial responses in acute kidney injury. J Clin Invest 2025; 135:e188358. [PMID: 40091836 PMCID: PMC11910233 DOI: 10.1172/jci188358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
Acute kidney injury (AKI) encompasses pathophysiology ranging from glomerular hypofiltration to tubular cell injury and outflow obstruction. This Review will focus on the tubulointerstitial processes that underlie most cases of AKI. Tubular epithelial cell (TEC) injury can occur via distinct insults, including ischemia, nephrotoxins, sepsis, and primary immune-mediated processes. Following these initial insults, tubular cells can activate survival and repair responses or they can develop mitochondrial dysfunction and metabolic reprogramming, cell-cycle arrest, and programmed cell death. Developing evidence suggests that the fate of individual tubular cells to survive and proliferate or undergo cell death or senescence is frequently determined by a biphasic immune response with initial proinflammatory macrophage, neutrophil, and lymphocyte infiltration exacerbating injury and activating programmed cell death, while alternatively activated macrophages and specific lymphocyte subsets subsequently modulate inflammation and promote repair. Functional recovery requires that this reparative phase supports proteolytic degradation of tubular casts, proliferation of surviving TECs, and restoration of TEC differentiation. Incomplete resolution or persistence of inflammation can lead to failed tubular repair, fibrosis, and chronic kidney disease. Despite extensive research in animal models, translating preclinical findings to therapies remains challenging, emphasizing the need for integrated multiomic approaches to advance AKI understanding and treatment.
Collapse
|
6
|
Yang H, Chen Y, He J, Li Y, Feng Y. Advances in the diagnosis of early biomarkers for acute kidney injury: a literature review. BMC Nephrol 2025; 26:115. [PMID: 40045274 PMCID: PMC11884078 DOI: 10.1186/s12882-025-04040-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
Acute kidney injury (AKI) is a critical condition with diverse manifestations and variable outcomes. Its diagnosis traditionally relies on delayed indicators such as serum creatinine and urine output, making early detection challenging. Early identification is essential to improving patient outcomes, driving the need for novel biomarkers. Recent advancements have identified promising biomarkers across various biological processes. Tubular injury markers, including neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), N-acetyl-β-D-glucosaminidase (NAG), and liver-type fatty acid-binding protein (L-FABP), offer insights into early tubular damage. Inflammatory and repair-associated biomarkers, such as interleukin-18 (IL-18), monocyte chemotactic protein-1 (MCP-1), osteopontin (OPN), and C-C motif chemokine ligand 14 (CCL14), reflect ongoing injury and recovery processes. Additionally, stress and repair markers like tissue inhibitor of metalloproteinase-2 (TIMP-2) and insulin-like growth factor-binding protein-7 (IGFBP-7), alongside filtration markers such as cystatin C (CysC) and proenkephalin (PenKid®) e.tal, further enhance diagnostic precision. Oxidative stress-related markers, including Superoxide Dismutase 1 (SOD1), also contribute valuable information. Emerging candidates, such as microRNAs, soluble urokinase plasminogen activator receptor (SuPAR), and chitinase-3-like protein 1 (CHI3L1), hold substantial promise for AKI detection and prognosis. This review summarizes the progress in AKI biomarker research, highlighting their clinical utility and exploring their potential to refine early diagnosis and management strategies. These findings offer a new perspective for integrating novel biomarkers into routine clinical practice, ultimately improving AKI care.
Collapse
Affiliation(s)
- Hongsha Yang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yanqin Chen
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Jiajia He
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yi Li
- Department of Nephrology, Institute of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Centre for Kidney Diseases, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yunlin Feng
- Department of Nephrology, Institute of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Centre for Kidney Diseases, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
7
|
Long JP, Singh S, Dong Y, Yee C, Lin JS. Urine proteomics defines an immune checkpoint-associated nephritis signature. J Immunother Cancer 2025; 13:e010680. [PMID: 39863302 PMCID: PMC11784134 DOI: 10.1136/jitc-2024-010680] [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: 09/26/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy is a cornerstone treatment for many cancers, but it can induce severe immunotoxicity, including acute interstitial nephritis (AIN). Currently, kidney biopsy is required to differentiate ICI-AIN from other causes of acute kidney injury (AKI). However, this invasive approach can lead to morbidity, delayed glucocorticoid treatment for patients with AIN, and unnecessarily prolonged suspension of ICI therapy in non-AIN patients. Delayed or incorrect diagnosis of ICI-AIN is particularly detrimental, as over 50% of patients are at risk of permanent renal damage. Thus, there is an urgent need for non-invasive biomarkers that can rapidly and accurately distinguish ICI-AIN from other causes of AKI.The urine and plasma proteome contain actively secreted proteins that provide real-time insights into dynamic physiological processes. However, identification of effective biomarkers of disease using established technologies such as proximity ligation assays (PLA) and bead-based immunoassays is challenging due to their limited sensitivity and loss of precision in multiplex analysis.To address this, we employed cutting-edge NUcleic acid Linked Immuno-Sandwich Assay (NULISA) technology to measure protein expression in urine and plasma samples from AKI patients undergoing ICI therapy. NULISA offers 10,000-fold greater precision than PLA, enabling quantification of over 200 inflammatory proteins with unprecedented precision. Our analysis revealed that urine was more sensitive and specific than plasma in distinguishing ICI-AIN from non-AIN cases. Pathway analyses highlighted the involvement of JAK-STAT and tumor necrosis factor (TNF) signaling in ICI-AIN pathogenesis. We identified several novel urine biomarkers, including IL-5, Fas, TNFSF4, CD274, IL-20, TNFSF15, TSLP, TREM1 and CCL1 while confirming previously reported markers such as CXCL9 and TNF-α. Using statistical and machine learning methods, we constructed a novel urine biomarker signature-IL-5+Fas-that achieved an area under the curve of 0.94 for diagnosing ICI-AIN.By leveraging high-sensitivity proteomics, we developed a non-invasive strategy for diagnosing ICI-AIN. This approach will enable earlier intervention to mitigate immunotoxicity, preservation of antitumor efficacy of ICI therapy in non-AIN patients, and safe rechallenge of ICI therapy in patients previously treated for ICI-AIN.
Collapse
Affiliation(s)
- James P Long
- Department of Biostatistics, Division of Discovery Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shailbala Singh
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yanlan Dong
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cassian Yee
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jamie S Lin
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
8
|
Baeg SI, Lee K, Jeon J, Lee JE, Kwon GY, Huh W, Jang HR. Urinary RANTES and MCP-1 as noninvasive biomarkers for differential diagnosis and prediction of treatment response in acute interstitial nephritis. Clin Kidney J 2025; 18:sfae354. [PMID: 39781473 PMCID: PMC11704796 DOI: 10.1093/ckj/sfae354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Indexed: 01/12/2025] Open
Abstract
Background Although kidney biopsy is definitive for the diagnosis of acute interstitial nephritis (AIN) and acute tubular necrosis (ATN), its invasiveness limits its use. We aimed to identify urine biomarkers for differentiating AIN and ATN and to predict the response of patients with AIN to steroid treatment. Methods In this prospective cohort study, biopsy-proven ATN (n = 34) and AIN (n = 55) were included. Urinary cytokine/chemokine [interleukin-9, monocyte chemoattractant protein-1 (MCP-1), regulated on activation, normal T cell expressed and secreted (RANTES), tumor necrosis factor-α, tumor growth factor-β and vascular endothelial growth factor] levels and the proportion of immune cells [expressing cluster of differentiation (CD)45, CD3, CD20] and proliferating tubular cells (expressing Ki-67) were analyzed by immunohistochemistry. Cytokine/chemokine levels and intrarenal immunohistochemistry data according to the response to steroid treatment in the AIN patients were also analyzed. Results The urinary RANTES/creatinine ratio and the percentages of intrarenal CD45-, CD3-, CD20- and Ki-67-positive cells were significantly higher in the AIN group than in the ATN group (P < .05 for all). Among steroid-administered patients with AIN, renal function improved significantly in the steroid responder group. These patients had higher urinary MCP-1/creatinine and intrarenal CD45 and Ki-67 levels than those in the non-responder group. Conclusions The potential of the urinary RANTES/creatinine ratio as a noninvasive biomarker for differentiating AIN from ATN is highlighted. Urinary MCP-1/creatinine levels and the proportion of total intrarenal leukocytes and proliferating tubular cells may serve as indicators for predicting the response of patients with AIN to steroid treatment.
Collapse
Affiliation(s)
- Song In Baeg
- Department of Internal Medicine, Division of Nephrology, Myongji Hospital, Hanyang University Medical Center, Goyang, Republic of Korea
| | - Kyungho Lee
- Department of Medicine, Division of Nephrology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Junseok Jeon
- Department of Medicine, Division of Nephrology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Eun Lee
- Department of Medicine, Division of Nephrology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ghee Young Kwon
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Wooseong Huh
- Department of Medicine, Division of Nephrology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hye Ryoun Jang
- Department of Medicine, Division of Nephrology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Dirim AB, Namazova N, Dirim MG, Oto OA, Artan AS, Hurdogan O, Ozluk Y, Yazici H. Baseline systemic inflammatory indices and clinicopathological features to predict the outcome of acute tubulointerstitial nephritis : A single-center retrospective study. Wien Klin Wochenschr 2025; 137:31-40. [PMID: 39190047 PMCID: PMC11739233 DOI: 10.1007/s00508-024-02417-2] [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: 01/21/2024] [Accepted: 07/16/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Acute tubulointerstitial nephritis (AIN) is an immune-mediated disorder that can cause acute kidney injury (AKI). We aimed to investigate the characteristics of patients with AIN and predictive factors for treatment response. MATERIAL AND METHODS In this study, thirty-one patients diagnosed with AIN on kidney biopsy between 2006 and 2021 were included. Baseline clinical, histopathological, and laboratory findings, including complete blood count (CBC), creatinine, erythrocyte sedimentation rate, C‑reactive protein, C3, C4, systemic immune inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and urinalysis were evaluated. Treatment response, mortality, and creatinine levels at the time of last follow-up were also noted. RESULTS The median age was 46 years and 80.6% were female. Median baseline creatinine and proteinuria levels were 4.1 mg/dL and 0.84 gram/day. The median follow-up period was 14 months and 93.5% received immunosuppressives. End-stage kidney disease (ESKD) developed in five patients (16.1%). Renal recovery (creatinine < 1.4 mg/dL) was observed in 17 patients (54.8%). Higher degrees of interstitial fibrosis, tubular atrophy, granuloma formation, global glomerulosclerosis, and higher baseline hemoglobin levels, in addition to a longer interval between first symptom to initiation of immunosuppressives were associated with renal nonrecovery, statistically. Also, patients who progressed to ESKD had higher baseline hemoglobin (p = 0.033) and lymphocyte (p = 0.044) and lower PLR levels (p = 0.016), as well as higher degrees of global glomerulosclerosis (p = 0.014), interstitial fibrosis (p = 0.042), and tubular atrophy (p = 0.030). CONCLUSION Treatment response rates are low for AIN, which may lead to ESKD. Besides chronicity in histopathology specimens, higher baseline hemoglobin levels and lower platelet-to-lymphocyte ratio might be prognostic. Further studies should be conducted on new markers for AIN.
Collapse
Affiliation(s)
- Ahmet Burak Dirim
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Istanbul University, 34104, Istanbul, Turkey.
| | - Nazrin Namazova
- Department of Internal Medicine, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Merve Guzel Dirim
- Department of Internal Medicine, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ozgur Akin Oto
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Istanbul University, 34104, Istanbul, Turkey
| | - Ayse Serra Artan
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Istanbul University, 34104, Istanbul, Turkey
| | - Ozge Hurdogan
- Department of Pathology, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yasemin Ozluk
- Department of Pathology, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Halil Yazici
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Istanbul University, 34104, Istanbul, Turkey
| |
Collapse
|
10
|
Herrmann SM, Abudayyeh A, Gupta S, Gudsoorkar P, Klomjit N, Motwani SS, Karam S, Costa E Silva VT, Khalid SB, Anand S, Kala J, Leaf DE, Murakami N, Rashidi A, Wanchoo R, Kitchlu A. Diagnosis and management of immune checkpoint inhibitor-associated nephrotoxicity: a position statement from the American Society of Onco-nephrology. Kidney Int 2025; 107:21-32. [PMID: 39455026 DOI: 10.1016/j.kint.2024.09.017] [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: 06/07/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer and are now the backbone of therapy for several malignancies. However, ICIs can cause a spectrum of kidney immune-related adverse events including acute kidney injury (AKI), most commonly manifesting as acute interstitial nephritis (AIN), although glomerular disease and electrolyte disturbances have also been reported. In this position statement by the American Society of Onco-nephrology (ASON), we summarize the incidence and risk factors for ICI-AKI, pathophysiological mechanisms, and clinicopathologic features of ICI-AKI. We also discuss novel diagnostic approaches and promising biomarkers for ICI-AKI. From expert panel consensus, we provide clinical practice points for the initial assessment and diagnosis of ICI-AKI, management and immunosuppressive therapy, and consideration for rechallenge with ICI following AKI episodes. In addition, we explore ICI use in special populations, such as kidney transplant recipients, and propose key areas of focus for future research and clinical investigation.
Collapse
Affiliation(s)
- Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.
| | - Ala Abudayyeh
- Section of Nephrology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Shruti Gupta
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA; Department of Medical Oncology, Dana-Farber Cancer Institute. Harvard Medical School, Boston, Massachusetts, USA
| | | | - Nattawat Klomjit
- Department of Medicine, Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shveta S Motwani
- Department of Medical Oncology, Dana-Farber Cancer Institute. Harvard Medical School, Boston, Massachusetts, USA
| | - Sabine Karam
- Department of Medicine, Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota, USA; Department of Internal Medicine, Division of Nephrology and Hypertension, American University of Beirut, Beirut, Lebanon
| | - Verônica T Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Laboratório de Investigação Médica (LIM) 16, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sheikh B Khalid
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Shuchi Anand
- Department of Medicine (Nephrology), Stanford University, Stanford, California, USA
| | - Jaya Kala
- Division of Renal Diseases and Hypertension, University of Texas Health Science Center at Houston-McGovern Medical School, Houston, Texas, USA
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Naoka Murakami
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Arash Rashidi
- Division of Nephrology and Hypertension, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Rimda Wanchoo
- Division of Kidney Diseases and Hypertension, Glomerular Center at Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York, USA
| | - Abhijat Kitchlu
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Mistry K, Sadarangani S, Moreno D, Mejia SM, Moledina DG, Sise ME. Novel Biomarkers and Imaging Tests for Acute Kidney Injury Diagnosis in Patients with Cancer. KIDNEY360 2025; 6:167-174. [PMID: 39575585 PMCID: PMC11793192 DOI: 10.34067/kid.0000000660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025]
Abstract
The lack of noninvasive urine and blood-based biomarkers for the diagnosis of AKI in patients with cancer is an area of significant unmet clinical need. Traditional noninvasive diagnostic tools that are currently used in the clinic, such as creatinine and cystatin C-based eGFR measurements, urinalysis, urine sediment examination, urine protein quantification, and urine electrolyte measurement, lack the sensitivity and specificity to distinguish between the various underlying etiologies of AKI in patients with cancer. Imaging-based diagnostics can be helpful to rule out urinary obstruction, but also lack sensitivity and specificity to diagnose the etiology of AKI. Kidney biopsy is often required for definitive diagnosis. As our scientific understanding of the biological pathways that are dysregulated in AKI has advanced, there has been considerable interest in developing new biomarkers for AKI. For example, the diagnosis of acute interstitial nephritis, which can occur in patients treated with immune checkpoint inhibitors, promises to be revolutionized by the incorporation of urinary testing for inflammatory biomarkers, such as C-X-C motif ligand 9, TNF- α , and IL-9. In the case of cisplatin administration, biomarkers such as neutrophil gelatinase-associated lipocalin and kidney injury molecule-1 may improve prognostication, differentiating between persistent AKI resulting from acute tubular injury versus prerenal azotemia. The development and validation of blood, urine, and imaging biomarkers into widely used diagnostic tests will require a concerted effort, but could improve diagnosis, management, and prognostication for a growing group of patients who are at high risk of developing AKI during the course of their illness.
Collapse
Affiliation(s)
- Kavita Mistry
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Sagar Sadarangani
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Daiana Moreno
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Sherley M. Mejia
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Dennis G. Moledina
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Internal Medicine, Clinical and Translational Research Accelerator, New Haven, Connecticut
| | - Meghan E. Sise
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
12
|
Douville NJ, Mathis M, Kheterpal S, Heung M, Schaub J, Naik A, Kretzler M. Perioperative Acute Kidney Injury: Diagnosis, Prediction, Prevention, and Treatment. Anesthesiology 2025; 142:180-201. [PMID: 39527650 PMCID: PMC11620328 DOI: 10.1097/aln.0000000000005215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 08/20/2024] [Indexed: 11/16/2024]
Abstract
In this review, the authors define acute kidney injury in the perioperative setting, describe the epidemiologic burden, discuss procedure-specific risk factors, detail principles of management, and highlight areas of ongoing controversy and research.
Collapse
Affiliation(s)
- Nicholas J. Douville
- Department of Anesthesiology, Michigan Medicine, Ann Arbor, Michigan; Institute of Healthcare Policy & Innovation, University of Michigan, Ann Arbor, Michigan; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Michael Mathis
- Department of Anesthesiology, Michigan Medicine, Ann Arbor, Michigan; Institute of Healthcare Policy & Innovation, University of Michigan, Ann Arbor, Michigan; Department of Computational Medicine and Bioinformatics, Ann Arbor, Michigan
| | - Sachin Kheterpal
- Department of Anesthesiology, Michigan Medicine, Ann Arbor, Michigan
| | - Michael Heung
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jennifer Schaub
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Abhijit Naik
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Matthias Kretzler
- Department of Computational Medicine and Bioinformatics, Ann Arbor, Michigan; Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
13
|
Meersch M, Mayerhöfer T, Joannidis M. Acute kidney injury subphenotyping and personalized medicine. Curr Opin Crit Care 2024; 30:555-562. [PMID: 39503205 DOI: 10.1097/mcc.0000000000001212] [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: 11/08/2024]
Abstract
PURPOSE OF REVIEW This review discusses novel concepts of acute kidney injury (AKI), including subphenotyping, which may facilitate the development of target treatment strategies for specific subgroups of patients to achieve precision medicine. RECENT FINDINGS AKI is a multifaceted syndrome with a major impact on morbidity and mortality. As efforts to identify treatment strategies have largely failed, it is becoming increasingly apparent that there are different subphenotypes that require different treatment strategies. Various ways of subphenotyping AKI have been investigated, including the use of novel renal biomarkers, machine learning and artificial intelligence, some of which have already been implemented in the clinical setting. Thus, novel renal biomarkers have been recommended for inclusion in new definition criteria for AKI and for the use of biomarker bundled strategies for the prevention of AKI. Computational models have been explored and require future research. SUMMARY Subphenotyping of AKI may provide a new understanding of this syndrome and guide targeted treatment strategies in order to improve patient outcomes.
Collapse
Affiliation(s)
- Melanie Meersch
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Timo Mayerhöfer
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
14
|
Ren Y, Zhang J, Hu X, Yu R, Tu Q, Li Y, Lin B, Zhu B, Shao L, Wang M. Association of peripheral eosinophil count with chronic kidney disease progression risk: a retrospective cohort study in Chinese population. Ren Fail 2024; 46:2394164. [PMID: 39212259 PMCID: PMC11370690 DOI: 10.1080/0886022x.2024.2394164] [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: 06/16/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The role of peripheral eosinophils in chronic kidney disease (CKD) requires further evaluation. We aimed to determine whether an eosinophil count increase is related to the occurrence of end-stage renal disease (ESRD). METHODS This single-center, observational, retrospective cohort study was conducted between January 2016 and December 2018 in Hangzhou, China, and included 3163 patients, categorized into four groups according to peripheral eosinophil count (PEC) quartile values. The main outcome was ESRD development during follow-up. We evaluated the relationship between the serum eosinophil count, demographic and clinical information, and ESRD incidence. Cox proportional hazards models and Kaplan-Meier survival curves were used. RESULTS A total of 3163 patients with CKD were included in this cohort, of whom 1254 (39.6%) were females. The median (interquartile range [IQR]) age was 75 [64, 85] years, and the median (IQR) estimated glomerular filtration rate was 55.16 [45.19, 61.19] mL/min/1.73 m2. The median PEC was 0.1224 × 109/L (IQR, 0.0625-0.212). Among the 3163 patients with CKD, 273 (8.6%) developed ESRD during a median follow-up time of 443.8 [238.8, 764.9] days. Individuals in the highest PEC quartile had a 66.2% higher ESRD risk than those in the lowest quartile (hazard ratio, 1.662; 95% confidence interval, 1.165-2.372). The results from the Kaplan-Meier survival curves confirmed the conclusion. CONCLUSIONS Alongside traditional risk factors, patients with CKD and an elevated PEC are more likely to develop ESRD. Therefore, more attention should be paid to those patients with CKD who have a high PEC.
Collapse
Affiliation(s)
- Yan Ren
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jinshi Zhang
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiao Hu
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Rizhen Yu
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiudi Tu
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yiwen Li
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Bo Lin
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Bin Zhu
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lina Shao
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Minmin Wang
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Quickfall D, La AM, Koyner JL. 10 tips on how to use dynamic risk assessment and alerts for AKI. Clin Kidney J 2024; 17:sfae325. [PMID: 39588357 PMCID: PMC11586629 DOI: 10.1093/ckj/sfae325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Indexed: 11/27/2024] Open
Abstract
Acute kidney injury (AKI) is a common syndrome in hospitalized patients and is associated with increased morbidity and mortality. The focus of AKI care requires a shift away from strictly supportive management of established injury to the early identification and timely prevention of worsening renal injury. Identifying patients at risk for developing or progression of severe AKI is crucial for improving patient outcomes, reducing the length of hospitalization and minimizing resource utilization. Implementation of dynamic risk scores and incorporation of novel biomarkers show promise for early detection and minimizing progression of AKI. Like any risk assessment tools, these require further external validation in a variety of clinical settings prior to widespread implementation. Additionally, alerts that may minimize exposure to a variety of nephrotoxic medications or prompt early nephrology consultation are shown to reduce the incidence and progression of AKI severity and enhance renal recovery. While dynamic risk scores and alerts are valuable, implementation requires thoughtfulness and should be used in conjunction with the overall clinical picture in certain situations, particularly when considering the initiation of fluid and diuretic administration or renal replacement therapy. Despite the contemporary challenges encountered with alert fatigue, implementing an alert-based bundle to improve AKI care is associated with improved outcomes, even when implementation is incomplete. Lastly, all alert-based interventions should be validated at an institutional level and assessed for their ability to improve institutionally relevant and clinically meaningful outcomes, reduce resource utilization and provide cost-effective interventions.
Collapse
Affiliation(s)
- Danica Quickfall
- Committee on Clinical Pharmacology and Pharmacogenomics, Biological Science Division, University of Chicago, Chicago, IL, USA
| | - Ashley M La
- Section of Nephrology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jay L Koyner
- Committee on Clinical Pharmacology and Pharmacogenomics, Biological Science Division, University of Chicago, Chicago, IL, USA
- Section of Nephrology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
16
|
Ostermann M, Legrand M, Meersch M, Srisawat N, Zarbock A, Kellum JA. Biomarkers in acute kidney injury. Ann Intensive Care 2024; 14:145. [PMID: 39279017 PMCID: PMC11402890 DOI: 10.1186/s13613-024-01360-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/07/2024] [Indexed: 09/18/2024] Open
Abstract
Acute kidney injury (AKI) is a multifactorial syndrome with a high risk of short- and long-term complications as well as increased health care costs. The traditional biomarkers of AKI, serum creatinine and urine output, have important limitations. The discovery of new functional and damage/stress biomarkers has enabled a more precise delineation of the aetiology, pathophysiology, site, mechanisms, and severity of injury. This has allowed earlier diagnosis, better prognostication, and the identification of AKI sub-phenotypes. In this review, we summarize the roles and challenges of these new biomarkers in clinical practice and research.
Collapse
Affiliation(s)
- Marlies Ostermann
- Department of Critical Care, Guy's & St Thomas' NHS Foundation Hospital, London, SE1 7EH, UK.
| | - Matthieu Legrand
- Department of Anesthesia and Perioperative Care, Division of Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Melanie Meersch
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Nattachai Srisawat
- Division of Nephrology, Department of Medicine, Faculty of Medicine, and Center of Excellence in Critical Care Nephrology, Chulalongkorn University, Bangkok, Thailand
| | - Alexander Zarbock
- Department of Anesthesia and Perioperative Care, Division of Critical Care Medicine, University of California San Francisco, San Francisco, USA
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - John A Kellum
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
17
|
Zarbock A, Forni LG, Koyner JL, Bell S, Reis T, Meersch M, Bagshaw SM, Fuhmann DY, Liu KD, Pannu N, Arikan AA, Angus DC, Duquette D, Goldstein SL, Hoste E, Joannidis M, Jongs N, Legrand M, Mehta RL, Murray PT, Nadim MK, Ostermann M, Prowle J, See EJ, Selby NM, Shaw AD, Srisawat N, Ronco C, Kellum JA. Recommendations for clinical trial design in acute kidney injury from the 31st acute disease quality initiative consensus conference. A consensus statement. Intensive Care Med 2024; 50:1426-1437. [PMID: 39115567 PMCID: PMC11377501 DOI: 10.1007/s00134-024-07560-y] [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: 04/28/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE Novel interventions for the prevention or treatment of acute kidney injury (AKI) are currently lacking. To facilitate the evaluation and adoption of new treatments, the use of the most appropriate design and endpoints for clinical trials in AKI is critical and yet there is little consensus regarding these issues. We aimed to develop recommendations on endpoints and trial design for studies of AKI prevention and treatment interventions based on existing data and expert consensus. METHODS At the 31st Acute Disease Quality Initiative (ADQI) meeting, international experts in critical care, nephrology, involving adults and pediatrics, biostatistics and people with lived experience (PWLE) were assembled. We focused on four main areas: (1) patient enrichment strategies, (2) prevention and attenuation studies, (3) treatment studies, and (4) innovative trial designs of studies other than traditional (parallel arm or cluster) randomized controlled trials. Using a modified Delphi process, recommendations and consensus statements were developed based on existing data, with > 90% agreement among panel members required for final adoption. RESULTS The panel developed 12 consensus statements for clinical trial endpoints, application of enrichment strategies where appropriate, and inclusion of PWLE to inform trial designs. Innovative trial designs were also considered. CONCLUSION The current lack of specific therapy for prevention or treatment of AKI demands refinement of future clinical trial design. Here we report the consensus findings of the 31st ADQI group meeting which has attempted to address these issues including the use of predictive and prognostic enrichment strategies to enable appropriate patient selection.
Collapse
Affiliation(s)
- Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, Albert-Schweitzer Campus 1, Building A1, 48149, Münster, Germany.
- Outcomes Research Consortium, Cleveland, OH, USA.
| | - Lui G Forni
- Depatment of Critical Care, Royal Surrey Hospital Foundation Trust, Guildford, Surrey, UK
- School of Medicine, Kate Granger Building, University of Surrey, Guildford, Surrey, UK
| | - Jay L Koyner
- Section of Nephrology, University of Chicago, Chicago, IL, USA
| | - Samira Bell
- Division of Population Health and Genomics, University of Dundee, Dundee, UK
| | - Thiago Reis
- Hospital Sírio-Libanês, São Paulo, Brazil
- Fenix Nephrology, São Paulo, Brazil
- Laboratory of Molecular Pharmacology, University of Brasília, Brasília, Brazil
| | - Melanie Meersch
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, Albert-Schweitzer Campus 1, Building A1, 48149, Münster, Germany
| | - Sean M Bagshaw
- Department of Critical Care Medicine, Faculty of Medicine and Dentistry, University of Alberta and Alberta Health Services, Edmonton, AB, Canada
| | - Dana Y Fuhmann
- UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Suite 2000, Pittsburgh, PA, 15224, USA
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathleen D Liu
- Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, CA, USA
| | - Neesh Pannu
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Ayse Akcan Arikan
- Division of Nephrology and Critical Care Medicine, Department of Pediatric, Baylor College of Medicine, Houston, TX, USA
| | - Derek C Angus
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - D'Arcy Duquette
- Critical Care Strategic Clinical Network, Alberta Health Services, Calgary, Canada
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Eric Hoste
- Intensive Care Unit, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Niels Jongs
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Matthieu Legrand
- Department of Anesthesia and Perioperative Care, Division of Critical Care Medicine, UCSF, San Francisco, CA, USA
| | - Ravindra L Mehta
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | | | - Mitra K Nadim
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Marlies Ostermann
- Department of Intensive Care, King's College London, Thomas' Hospital, Guy's & St, London, UK
| | - John Prowle
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Emily J See
- Department of Critical Care, University of Melbourne, Parkville, Australia
- Department of Intensive Care, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Nephrology, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Nicholas M Selby
- Centre for Kidney Research and Innovation, University of Nottingham, Nottingham, UK
| | - Andrew D Shaw
- Department of Intensive Care and Resuscitation, The Cleveland Clinic, Cleveland, OH, USA
| | - Nattachai Srisawat
- Division of Nephrology, Department of Medicine, Faculty of Medicine, and Center of Excellence in Critical Care Nephrology, Chulalongkorn University, Bangkok, Thailand
| | - Claudio Ronco
- Department of Medicine, University of Padova, Padua, Italy
- International Renal Research Institute of Vicenza (IRRV), Vicenza, Italy
- Department of Nephrology, San Bortolo Hospital, Vicenza, Italy
| | - John A Kellum
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
18
|
Birkelo BC, Koyner JL, Ostermann M, Bhatraju PK. The Road to Precision Medicine for Acute Kidney Injury. Crit Care Med 2024; 52:1127-1137. [PMID: 38869385 PMCID: PMC11250999 DOI: 10.1097/ccm.0000000000006328] [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] [Indexed: 06/14/2024]
Abstract
OBJECTIVES Acute kidney injury (AKI) is a common form of organ dysfunction in the ICU. AKI is associated with adverse short- and long-term outcomes, including high mortality rates, which have not measurably improved over the past decade. This review summarizes the available literature examining the evidence of the need for precision medicine in AKI in critical illness, highlights the current evidence for heterogeneity in the field of AKI, discusses the progress made in advancing precision in AKI, and provides a roadmap for studying precision-guided care in AKI. DATA SOURCES Medical literature regarding topics relevant to precision medicine in AKI, including AKI definitions, epidemiology, and outcomes, novel AKI biomarkers, studies of electronic health records (EHRs), clinical trial design, and observational studies of kidney biopsies in patients with AKI. STUDY SELECTION English language observational studies, randomized clinical trials, reviews, professional society recommendations, and guidelines on areas related to precision medicine in AKI. DATA EXTRACTION Relevant study results, statements, and guidelines were qualitatively assessed and narratively synthesized. DATA SYNTHESIS We synthesized relevant study results, professional society recommendations, and guidelines in this discussion. CONCLUSIONS AKI is a syndrome that encompasses a wide range of underlying pathologies, and this heterogeneity has hindered the development of novel therapeutics for AKI. Wide-ranging efforts to improve precision in AKI have included the validation of novel biomarkers of AKI, leveraging EHRs for disease classification, and phenotyping of tubular secretory clearance. Ongoing efforts such as the Kidney Precision Medicine Project, identifying subphenotypes in AKI, and optimizing clinical trials and endpoints all have great promise in advancing precision medicine in AKI.
Collapse
Affiliation(s)
- Bethany C Birkelo
- Division of Nephrology, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Jay L Koyner
- Section of Nephrology, Department of Medicine, University of Chicago, Chicago, IL
| | - Marlies Ostermann
- Department of Critical Care and Nephrology, King's College London, Guy's and St. Thomas' Hospital, London, United Kingdom
| | - Pavan K Bhatraju
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA
- Kidney Research Institute, University of Washington, Seattle, WA
| |
Collapse
|
19
|
Palanisamy T, Basu S, Srinivas BH, Shamanna S. Urinary Biomarkers for Non-Invasive Diagnosis of Acute Interstitial Nephritis. Indian J Nephrol 2024; 34:409-410. [PMID: 39156852 PMCID: PMC11326792 DOI: 10.25259/ijn_38_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 08/20/2024] Open
Affiliation(s)
- Tamilselvan Palanisamy
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Sharbari Basu
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Bheemanathi Hanuman Srinivas
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Suryanarayana Shamanna
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
20
|
Lin KM, Su CC, Chen JY, Pan SY, Chuang MH, Lin CJ, Wu CJ, Pan HC, Wu VC. Biomarkers in pursuit of precision medicine for acute kidney injury: hard to get rid of customs. Kidney Res Clin Pract 2024; 43:393-405. [PMID: 38934040 PMCID: PMC11237332 DOI: 10.23876/j.krcp.23.284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/08/2024] [Accepted: 02/13/2024] [Indexed: 06/28/2024] Open
Abstract
Traditional acute kidney injury (AKI) classifications, which are centered around semi-anatomical lines, can no longer capture the complexity of AKI. By employing strategies to identify predictive and prognostic enrichment targets, experts could gain a deeper comprehension of AKI's pathophysiology, allowing for the development of treatment-specific targets and enhancing individualized care. Subphenotyping, which is enriched with AKI biomarkers, holds insights into distinct risk profiles and tailored treatment strategies that redefine AKI and contribute to improved clinical management. The utilization of biomarkers such as N-acetyl-β-D-glucosaminidase, tissue inhibitor of metalloprotease-2·insulin-like growth factor-binding protein 7, kidney injury molecule-1, and liver fatty acid-binding protein garnered significant attention as a means to predict subclinical AKI. Novel biomarkers offer promise in predicting persistent AKI, with urinary motif chemokine ligand 14 displaying significant sensitivity and specificity. Furthermore, they serve as predictive markers for weaning patients from acute dialysis and offer valuable insights into distinct AKI subgroups. The proposed management of AKI, which is encapsulated in a structured flowchart, bridges the gap between research and clinical practice. It streamlines the utilization of biomarkers and subphenotyping, promising a future in which AKI is swiftly identified and managed with unprecedented precision. Incorporating kidney biomarkers into strategies for early AKI detection and the initiation of AKI care bundles has proven to be more effective than using care bundles without these novel biomarkers. This comprehensive approach represents a significant stride toward precision medicine, enabling the identification of high-risk subphenotypes in patients with AKI.
Collapse
Grants
- MOST 107-2314-B-002-026-MY3, 108-2314B-002-058, 110-2314-B-002-241, 110-2314-B-002-239 Ministry of Science and Technology (MOST) of the Republic of China (Taiwan)
- NSTC 109-2314-B-002-174-MY3, 110-2314-B-002124-MY3, 111-2314-B-002-046, 111-2314-B-002-058 National Science and Technology Council
- PH-102-SP-09 National Health Research Institutes
- 109-S4634, PC-1246, PC-1309, VN109-09, UN109-041, UN110-030, 111-FTN0011 National Taiwan University Hospital
Collapse
Affiliation(s)
- Kun-Mo Lin
- Division of Nephrology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Ching-Chun Su
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jui-Yi Chen
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Szu-Yu Pan
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan
| | - Min-Hsiang Chuang
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Cheng-Jui Lin
- Division of Nephrology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chih-Jen Wu
- Division of Nephrology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Heng-Chih Pan
- Division of Nephrology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Taiwan
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Primary Aldosteronism Center of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- NSARF (National Taiwan University Hospital Study Group of ARF), Taipei, Taiwan
| |
Collapse
|
21
|
Barsotti GC, Luciano R, Kumar A, Meliambro K, Kakade V, Tokita J, Naik A, Fu J, Peck E, Pell J, Reghuvaran A, Tanvir E, Patel P, Zhang W, Li F, Moeckel G, Perincheri S, Cantley L, Moledina DG, Wilson FP, He JC, Menon MC. Rationale and Design of a Phase 2, Double-blind, Placebo-Controlled, Randomized Trial Evaluating AMP Kinase-Activation by Metformin in Focal Segmental Glomerulosclerosis. Kidney Int Rep 2024; 9:1354-1368. [PMID: 38707807 PMCID: PMC11068976 DOI: 10.1016/j.ekir.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction Focal segmental glomerulosclerosis (FSGS), the most common primary glomerular disease leading to end-stage kidney disease (ESKD), is characterized by podocyte injury and depletion, whereas minimal change disease (MCD) has better outcomes despite podocyte injury. Identifying mechanisms capable of preventing podocytopenia during injury could transform FSGS to an "MCD-like" state. Preclinical data have reported conversion of an MCD-like injury to one with podocytopenia and FSGS by inhibition of AMP-kinase (AMPK) in podocytes. Conversely, in FSGS, AMPK-activation using metformin (MF) mitigated podocytopenia and azotemia. Observational studies also support beneficial effects of MF on proteinuria and chronic kidney disease (CKD) outcomes in diabetes. A randomized controlled trial (RCT) to test MF in podocyte injury with FSGS has not yet been conducted. Methods We report the rationale and design of phase 2, double-blind, placebo-controlled RCT evaluating the efficacy and safety of MF as adjunctive therapy in FSGS. By randomizing 30 patients with biopsy-confirmed FSGS to MF or placebo (along with standard immunosuppression), we will study mechanistic biomarkers that correlate with podocyte injury or depletion and evaluate outcomes after 6 months. We specifically integrate novel urine, blood, and tissue markers as surrogates for FSGS progression along with unbiased profiling strategies. Results and Conclusion Our phase 2 trial will provide insight into the potential efficacy and safety of MF as adjunctive therapy in FSGS-a crucial step to developing a larger phase 3 study. The mechanistic assays here will guide the design of other FSGS trials and contribute to understanding AMPK activation as a potential therapeutic target in FSGS. By repurposing an inexpensive agent, our results will have implications for FSGS treatment in resource-poor settings.
Collapse
Affiliation(s)
- Gabriel C. Barsotti
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Randy Luciano
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ashwani Kumar
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kristin Meliambro
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vijayakumar Kakade
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joji Tokita
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Abhijit Naik
- Division of Nephrology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Jia Fu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Elizabeth Peck
- Clinical Research Coordinator, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John Pell
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anand Reghuvaran
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - E.M. Tanvir
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Prashant Patel
- Investigational Drug Service, Department of Pharmacy Services, Yale New Haven Hospital, Connecticut, USA
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fan Li
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
| | - Gilbert Moeckel
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sudhir Perincheri
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lloyd Cantley
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dennis G. Moledina
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - F. Perry Wilson
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - John C. He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Madhav C. Menon
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
22
|
D Souza S, Obeid W, Hernandez J, Hu D, Wen Y, Moledina DG, Albert A, Gregg A, Wheeler A, Philbrook HT, Parikh CR. The development of lateral flow devices for urinary biomarkers to assess kidney health. Sci Rep 2024; 14:8516. [PMID: 38609491 PMCID: PMC11014899 DOI: 10.1038/s41598-024-59104-5] [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: 10/31/2023] [Accepted: 04/08/2024] [Indexed: 04/14/2024] Open
Abstract
Serum creatinine levels are insensitive to real-time changes in kidney function or injury. There is a growing interest in assessing kidney injury by measuring biomarkers in body fluid. From our previous studies, we identified and reported three urinary biomarkers namely Uromodulin (UMOD), Osteopontin (OPN), and Interleukin-9 (IL-9) to be associated with kidney health. The availability of a rapid point-of-care test for these urinary biomarkers will potentially accelerate its applicability and accessibility. In this study, we aimed to develop novel lateral flow device (LFD) for UMOD, OPN and IL-9. We tested paired antibodies using Enzyme Linked Immunosorbent Assay wherein we observed functionality only for UMOD and OPN and not for IL-9. A conjugation buffer pH of 7.8 and 8.5 was found suitable at a detection antibody concentration of 15 µg/mL for LFD development. The developed LFDs were found to quantitatively measure UMOD standard (LLOD of 80,000 pg/mL) and OPN standard (LLOD of 8600 pg/mL) respectively. The LFD was also able to measure human urinary UMOD and OPN with a percent CV of 12.12 and 5.23 respectively.
Collapse
Affiliation(s)
- Serena D Souza
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, MD, 21287, USA
| | - Wassim Obeid
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, MD, 21287, USA
| | - Jeanine Hernandez
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, MD, 21287, USA
| | - David Hu
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, MD, 21287, USA
| | - Yumeng Wen
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, MD, 21287, USA
| | - Dennis G Moledina
- Section of Nephrology and Clinical and Translational Research Accelerator, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Andre Albert
- Mologic Inc (D/B/A Global Access Diagnostics), 83 Pineland Drive, Gray Hall Suite 202, New Gloucester, ME, USA
| | - Anya Gregg
- Mologic Ltd (D/B/A Global Access Diagnostics), Bedford Technology Park, Thurleigh, UK
| | - Andrew Wheeler
- Mologic Inc (D/B/A Global Access Diagnostics), 83 Pineland Drive, Gray Hall Suite 202, New Gloucester, ME, USA
| | - Heather Thiessen Philbrook
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, MD, 21287, USA
| | - Chirag R Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, MD, 21287, USA.
| |
Collapse
|
23
|
Parikh CR, Coca SG. Are biomarkers in acute kidney injury ready for prime time? The time is right for a second look. Kidney Int 2024; 105:675-678. [PMID: 38519236 DOI: 10.1016/j.kint.2024.01.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/18/2023] [Accepted: 01/03/2024] [Indexed: 03/24/2024]
Affiliation(s)
- Chirag R Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.
| | - Steven G Coca
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
24
|
Muhammad A, Zhang Y, Huang L, Yuan Q, Wang W, Pu J, Lin W, Tang R, Xiao X. The diagnosis of acute interstitial nephritis caused by infection versus antibiotic-induced interstitial nephritis: a narrative review. Clin Kidney J 2024; 17:sfae054. [PMID: 38572500 PMCID: PMC10986214 DOI: 10.1093/ckj/sfae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Indexed: 04/05/2024] Open
Abstract
Acute interstitial nephritis (AIN) is a significant contributor to acute kidney injury and can be attributed to a variety of factors, including but not limited to allergens or drugs, infections, autoimmune or systemic diseases, and idiopathic forms of the disease. In some cases, AIN requires a therapeutic action according to a single specific etiology by handling the offending agent and applying an immunosuppressant. Although AIN can be diagnosed through renal biopsy, it is not able to pinpoint the precise cause when multiple causes are suspected to be present simultaneously. Such situations arise when a patient suffering from infection develops AIN during antibiotic therapy, the exact causative factor of which becomes a challenge for the clinicians to determine. This is attributed to the different approaches employed in different etiologies, wherein clinicians are required to maintain the current antibiotic therapy or augment the dose in cases of infection as AIN etiology, without resorting to immunosuppressant therapy as the primary objective is infection killing. In contrast, antibiotics as an etiology for AIN require an alternative drug from the antibiotics group, along with an immunosuppressant. In the interim, delaying the identification of the precise cause may result in interstitial fibrosis and chronic kidney disease. This narrative review highlights certain findings that can be typical of infection-associated ATIN compared with antibiotic-associated ATIN based on clinical history and physical examination, clinical presentation of different antibiotic drug classes, histopathological features, classical and novel biomarkers, serum and urine cytokines and chemokines, cellular biomarkers, and genetic biomarkers. Although these findings cannot provide conclusive and clear recommendations that can be useful in the clinical practice, they can entice researchers to conduct original research on these features to discover clear recommendations.
Collapse
Affiliation(s)
- Amir Muhammad
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Yingli Zhang
- Department of Nephrology, Third Hospital of Changsha, Changsha, China
| | - Ling Huang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaxi Pu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Lin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Rong Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
25
|
Rossiter A, La A, Koyner JL, Forni LG. New biomarkers in acute kidney injury. Crit Rev Clin Lab Sci 2024; 61:23-44. [PMID: 37668397 DOI: 10.1080/10408363.2023.2242481] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/14/2023] [Accepted: 07/26/2023] [Indexed: 09/06/2023]
Abstract
Acute kidney injury (AKI) is a commonly encountered clinical syndrome. Although it often complicates community acquired illness, it is more common in hospitalized patients, particularly those who are critically ill or who have undergone major surgery. Approximately 20% of hospitalized adult patients develop an AKI during their hospital care, and this rises to nearly 60% in the critically ill, depending on the population being considered. In general, AKI is more common in older adults, in those with preexisting chronic kidney disease and in those with known risk factors for AKI (including diabetes and hypertension). The development of AKI is associated with an increase in both mortality and morbidity, including the development of post-AKI chronic kidney disease. Currently, AKI is defined by a rise in serum creatinine from either a known or derived baseline value and/or oliguria or anuria. However, clinicians may fail to recognize the initial development of AKI because of a delay in the rise of serum creatinine or because of inaccurate urine output monitoring. This, in turn, delays any putative measures to treat AKI or to limit its degree. Consequently, efforts have focused on new biomarkers associated with AKI that may allow early recognition of this syndrome with the intent that this will translate into improved patient outcomes. Here we outline current biomarkers associated with AKI and explore their potential in aiding diagnosis, understanding the pathophysiology and directing therapy.
Collapse
Affiliation(s)
- Adam Rossiter
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surry, UK
| | - Ashley La
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Jay L Koyner
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Lui G Forni
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surry, UK
- School of Medicine, Department of Clinical & Experimental Medicine, Faculty of Health Sciences, University of Surrey, Surry, UK
| |
Collapse
|
26
|
Nie Q, Zhang B, Li R, Yang Y, Ren J, Qiu L, Lu Y, Zhu L, Shen H, Liu Y, You R. Ultra-sensitive detection of tumor necrosis factor alpha based on silver-coated gold core shell and magnetically separated recognition of SERS aptamer sensors. Mikrochim Acta 2023; 191:41. [PMID: 38112843 DOI: 10.1007/s00604-023-06049-7] [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: 08/03/2023] [Accepted: 10/15/2023] [Indexed: 12/21/2023]
Abstract
A highly sensitive tumor necrosis factor α (TNF-α) detection method based on a surface-enhanced Raman scattering (SERS) magnetic patch sensor is reported. Magnetic beads (MNPs) and core shells were used as the capture matrix and signaling probe, respectively. For this purpose, antibodies were immobilized on the surface of magnetic beads, and then Au@4-MBN@Ag core-shell structures coupled with aptamers and TNF-α antigen were added sequentially to form a sandwich immune complex. Quantitative analysis was performed by monitoring changes in the characteristic SERS signal intensity of the Raman reporter molecule 4-MBN. The results showed that the limit of detection (LOD) of the proposed method was 4.37 × 10-15 mg·mL-1 with good linearity (R2 = 0.9918) over the concentration range 10-12 to 10-5 mg·mL-1. Excellent assay accuracy was also demonstrated, with recoveries in the range 102% to 114%. Since all reactions occur in solution and are separated by magnetic adsorption of magnetic beads, this SERS-based immunoassay technique solves the kinetic problems of limited diffusion and difficult separation on solid substrates. The method is therefore expected to be a good clinical tool for the diagnosis of the inflammatory biomarker THF-α and in vivo inflammation screening.
Collapse
Affiliation(s)
- Qingling Nie
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China
| | - Bohan Zhang
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China
| | - Rong Li
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China
| | - Yixuan Yang
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China
| | - Junjie Ren
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China
| | - Liting Qiu
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China
| | - Yudong Lu
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China
| | - Lanjin Zhu
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China
| | - Huiying Shen
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China
| | - Yunzhen Liu
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China.
| | - Ruiyun You
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Oriented Chemical Engineer, Fujian Key Laboratory of Polymer Materials, Engineering Research Center of Industrial Biocatalysis, Fujian Province Higher Education Institutes, Fujian Normal University, Fuzhou, 350007, Fujian, China.
| |
Collapse
|
27
|
Guo X, Blanc V, Davidson NO, Velazquez H, Chen TM, Moledina DG, Moeckel GW, Safirstein RL, Desir GV. APOBEC-1 deletion enhances cisplatin-induced acute kidney injury. Sci Rep 2023; 13:22255. [PMID: 38097707 PMCID: PMC10721635 DOI: 10.1038/s41598-023-49575-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023] Open
Abstract
Cisplatin (CP) induces acute kidney injury (AKI) whereby proximal tubules undergo regulated necrosis. Repair is almost complete after a single dose. We now demonstrate a role for Apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 (Apobec-1) that is prominently expressed at the interface between acute and chronic kidney injury (CKD), in the recovery from AKI. Apobec-1 knockout (KO) mice exhibited greater mortality than in wild type (WT) and more severe AKI in both CP- and unilateral ischemia reperfusion (IR) with nephrectomy. Specifically, plasma creatinine (pCr) 2.6 ± 0.70 mg/dL for KO, n = 10 and 0.16 ± 0.02 for WT, n = 6, p < 0.0001 in CP model and 1.34 ± 0.22 mg/dL vs 0.75 ± 0.06, n = 5, p < 0.05 in IR model. The kidneys of Apobec-1 KO mice showed increased necrosis, increased expression of KIM-1, NGAL, RIPK1, ASCL4 and increased lipid accumulation compared to WT kidneys (p < 0.01). Neutrophils and activated T cells were both increased, while macrophages were reduced in kidneys of Apobec-1 KO animals. Overexpression of Apobec-1 in mouse proximal tubule cells protected against CP-induced cytotoxicity. These findings suggest that Apobec-1 mediates critical pro-survival responses to renal injury and increasing Apobec-1 expression could be an effective strategy to mitigate AKI.
Collapse
Affiliation(s)
- Xiaojia Guo
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Valerie Blanc
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63105, USA
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63105, USA
| | - Heino Velazquez
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Veteran's Affair Medical Center, West Haven, CT, USA
| | - Tian-Min Chen
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Dennis G Moledina
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Robert L Safirstein
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Veteran's Affair Medical Center, West Haven, CT, USA.
| | - Gary V Desir
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Veteran's Affair Medical Center, West Haven, CT, USA.
| |
Collapse
|
28
|
Tang J, Yang N, Pan S, Ren P, Chen M, Jin J, He Q, Zeng Y. The renal damage and mechanisms relevant to antitumoral drugs. Front Oncol 2023; 13:1331671. [PMID: 38148845 PMCID: PMC10749913 DOI: 10.3389/fonc.2023.1331671] [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: 11/01/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Over the past few decades, significant progress has been made in the development of drugs to combat cancer. It is unfortunate that these drugs can also lead to various kidney injuries and imbalances in electrolyte levels. Nephrotoxicity caused by chemotherapy drugs can impact different parts of the kidneys, including the glomeruli, renal tubules, interstitium, or renal microvessels. Despite the existing knowledge, our understanding of the mechanisms underlying the renal damage caused by antitumoral drugs remains incomplete. In this review, we aim to provide a comprehensive overview of the specific types of kidney injury and the mechanisms responsible for the drug-mediated renal damage, and briefly discuss possible prevention and treatment measures. Sensitive blood and urine biomarkers can provide clinicians with more information about kidney injury detection and reference value for subsequent treatment options. In addition, we emphasize that both oncologists and nephrologists have a responsibility to remain vigilant against the potential nephrotoxicity of the drugs. It's crucial for experts in both fields to collaborate in early detection, monitoring and prevention of kidney damage.
Collapse
Affiliation(s)
- Jiyu Tang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | - Nan Yang
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
- Jinzhou Medical University, Graduate School of Clinical Medicine, Jinzhou, China
| | - Shujun Pan
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Peiyao Ren
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Maosheng Chen
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | - Juan Jin
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Qiang He
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Yuqun Zeng
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
29
|
Siew ED. Do Novel Biomarkers Have Utility in the Diagnosis and Prognosis of AKI?: Commentary. KIDNEY360 2023; 4:1670-1671. [PMID: 38153791 PMCID: PMC10917109 DOI: 10.34067/kid.0000000000000240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 07/31/2023] [Indexed: 12/30/2023]
Affiliation(s)
- Edward D Siew
- Division of Nephrology and Hypertension, Vanderbilt Center for Kidney Disease (VCKD) and Integrated Program for Acute Kidney Injury (AKI), Vanderbilt University Medical Center, Nashville, Tennessee and Tennessee Valley Health Systems (TVHS), Nashville Veterans Affairs Hospital, Tennessee
| |
Collapse
|
30
|
Coca SG. Do Novel Biomarkers Have Utility in the Diagnosis and Prognosis of AKI? CON. KIDNEY360 2023; 4:1667-1669. [PMID: 37291706 PMCID: PMC10758505 DOI: 10.34067/kid.0000000000000188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Affiliation(s)
- Steven G Coca
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
31
|
Miao J, Herrmann SM. Immune checkpoint inhibitors and their interaction with proton pump inhibitors-related interstitial nephritis. Clin Kidney J 2023; 16:1834-1844. [PMID: 37915905 PMCID: PMC10616479 DOI: 10.1093/ckj/sfad109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Indexed: 11/03/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy and outcomes, leading to an expanding use in millions of patients worldwide. However, they can cause a spectrum of immune-related adverse events (irAEs). Essentially, any organs can be affected by irAEs, which have emerged as therapy-limiting side effects. In the kidneys, ICI-associated acute interstitial nephritis (ICI-AIN) leads to acute kidney injury (AKI) in 2%-5% of patients on ICI therapy. AKI associated with ICI therapy pathologically presents with AIN in nearly 90% of the cases, but the pathophysiology of ICI-AIN remains to be defined. The generation of autoreactive T cells in patients receiving AIN-inducible drugs, such as proton pump inhibitors (PPIs), is one of the leading theories, supported by a higher incidence of ICI-AIN in patients on these AIN-inducible drugs. In this review, we will discuss our understanding of the incidence, potential pathophysiological mechanisms, clinical presentations, risk factors, diagnosis, and management of PPI-related AIN and its interaction with ICI therapy.
Collapse
Affiliation(s)
- Jing Miao
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
32
|
Seethapathy H, Mistry K, Sise ME. Immunological mechanisms underlying clinical phenotypes and noninvasive diagnosis of immune checkpoint inhibitor-induced kidney disease. Immunol Rev 2023; 318:61-69. [PMID: 37482912 PMCID: PMC10865966 DOI: 10.1111/imr.13243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/28/2023] [Indexed: 07/25/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have become a mainstay of cancer therapy, with over 80 FDA-approved indications. Used in a variety of settings and in combination with each other and with traditional chemotherapies, the hyperactive immune response induced by ICIs can often lead to immune-related adverse events in bystander normal tissues such as the kidneys, lungs, and the heart. In the kidneys, this immune-related adverse event manifests as acute interstitial nephritis (ICI-AIN). In the era of widespread ICI use, it becomes vital to understand the clinical manifestations of ICI-AIN and the importance of prompt diagnosis and management of these complications. In this review, we delve into the clinical phenotypes of ICI-AIN and how they differ from traditional drug-induced AIN. We also detail what is known about the mechanistic underpinnings of ICI-AIN and the important diagnostic and therapeutic implications behind harnessing those mechanisms to further our understanding of these events and to formulate effective treatment plans to manage ICI-AIN.
Collapse
Affiliation(s)
- Harish Seethapathy
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kavita Mistry
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Meghan E. Sise
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Stanski NL, Rodrigues CE, Strader M, Murray PT, Endre ZH, Bagshaw SM. Precision management of acute kidney injury in the intensive care unit: current state of the art. Intensive Care Med 2023; 49:1049-1061. [PMID: 37552332 DOI: 10.1007/s00134-023-07171-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/12/2023] [Indexed: 08/09/2023]
Abstract
Acute kidney injury (AKI) is a prototypical example of a common syndrome in critical illness defined by consensus. The consensus definition for AKI, traditionally defined using only serum creatinine and urine output, was needed to standardize the description for epidemiology and to harmonize eligibility for clinical trials. However, AKI is not a simple disease, but rather a complex and multi-factorial syndrome characterized by a wide spectrum of pathobiology. AKI is now recognized to be comprised of numerous sub-phenotypes that can be discriminated through shared features such as etiology, prognosis, or common pathobiological mechanisms of injury and damage. The characterization of sub-phenotypes can serve to enable prognostic enrichment (i.e., identify subsets of patients more likely to share an outcome of interest) and predictive enrichment (identify subsets of patients more likely to respond favorably to a given therapy). Existing and emerging biomarkers will aid in discriminating sub-phenotypes of AKI, facilitate expansion of diagnostic criteria, and be leveraged to realize personalized approaches to management, particularly for recognizing treatment-responsive mechanisms (i.e., endotypes) and targets for intervention (i.e., treatable traits). Specific biomarkers (e.g., serum renin; olfactomedin 4 (OLFM4); interleukin (IL)-9) may further enable identification of pathobiological mechanisms to serve as treatment targets. However, even non-specific biomarkers of kidney injury (e.g., neutrophil gelatinase-associated lipocalin, NGAL; [tissue inhibitor of metalloproteinases 2, TIMP2]·[insulin like growth factor binding protein 7, IGFBP7]; kidney injury molecule 1, KIM-1) can direct greater precision management for specific sub-phenotypes of AKI. This review will summarize these evolving concepts and recent innovations in precision medicine approaches to the syndrome of AKI in critical illness, along with providing examples of how they can be leveraged to guide patient care.
Collapse
Affiliation(s)
- Natalja L Stanski
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Camila E Rodrigues
- Department of Nephrology, Prince of Wales Clinical School, UNSW Medicine, Sydney, NSW, Australia
- Nephrology Department, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Michael Strader
- Department of Medicine, School of Medicine, University College Dublin, Dublin, Ireland
| | - Patrick T Murray
- Department of Medicine, School of Medicine, University College Dublin, Dublin, Ireland
| | - Zoltan H Endre
- Department of Nephrology, Prince of Wales Clinical School, UNSW Medicine, Sydney, NSW, Australia
| | - Sean M Bagshaw
- Department of Critical Care Medicine, Faculty of Medicine and Dentistry, University of Alberta and Alberta Health Services, 2-124 Clinical Sciences Building, 8440-112 ST NW, Edmonton, AB, T6G 2B7, Canada.
| |
Collapse
|
34
|
Canney M, Clark EG, Hiremath S. Biomarkers in acute kidney injury: On the cusp of a new era? J Clin Invest 2023; 133:e171431. [PMID: 37395275 PMCID: PMC10313371 DOI: 10.1172/jci171431] [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: 07/04/2023] Open
Abstract
The field of nephrology has been slow in moving beyond the utilization of creatinine as an indicator for chronic kidney disease and acute kidney injury (AKI). Early diagnosis and establishment of etiology, in particular, are important for treatment of AKI. In the setting of hospital-acquired AKI, tubular injury is more common, but acute interstitial nephritis (AIN) has a more treatable etiology. However, it is likely that AIN is under- or misdiagnosed due to current strategies that largely rely on clinical gestalt. In this issue of the JCI, Moledina and colleagues made an elegant case for the chemokine called C-X-C motif ligand 9 (CXCL9) as a biomarker of AIN. The authors used urine proteomics and tissue transcriptomics in patients with and without AIN to identify CXCL9 as a promising, noninvasive, diagnostic biomarker of AIN. These results have clinical implications that should catalyze future research and clinical trials in this space.
Collapse
Affiliation(s)
- Mark Canney
- Division of Nephrology, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Edward G. Clark
- Division of Nephrology, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Swapnil Hiremath
- Division of Nephrology, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
35
|
Moledina DG, Obeid W, Smith RN, Rosales I, Sise ME, Moeckel G, Kashgarian M, Kuperman M, Campbell KN, Lefferts S, Meliambro K, Bitzer M, Perazella MA, Luciano RL, Pober JS, Cantley LG, Colvin RB, Wilson FP, Parikh CR. Identification and validation of urinary CXCL9 as a biomarker for diagnosis of acute interstitial nephritis. J Clin Invest 2023; 133:e168950. [PMID: 37395276 PMCID: PMC10313360 DOI: 10.1172/jci168950] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/14/2023] [Indexed: 07/04/2023] Open
Abstract
BackgroundAcute tubulointerstitial nephritis (AIN) is one of the few causes of acute kidney injury with diagnosis-specific treatment options. However, due to the need to obtain a kidney biopsy for histological confirmation, AIN diagnosis can be delayed, missed, or incorrectly assumed. Here, we identify and validate urinary CXCL9, an IFN-γ-induced chemokine involved in lymphocyte chemotaxis, as a diagnostic biomarker for AIN.MethodsIn a prospectively enrolled cohort with pathologist-adjudicated histological diagnoses, termed the discovery cohort, we tested the association of 180 immune proteins measured by an aptamer-based assay with AIN and validated the top protein, CXCL9, using sandwich immunoassay. We externally validated these findings in 2 cohorts with biopsy-confirmed diagnoses, termed the validation cohorts, and examined mRNA expression differences in kidney tissue from patients with AIN and individuals in the control group.ResultsIn aptamer-based assay, urinary CXCL9 was 7.6-fold higher in patients with AIN than in individuals in the control group (P = 1.23 × 10-5). Urinary CXCL9 measured by sandwich immunoassay was associated with AIN in the discovery cohort (n = 204; 15% AIN) independently of currently available clinical tests for AIN (adjusted odds ratio for highest versus lowest quartile: 6.0 [1.8-20]). Similar findings were noted in external validation cohorts, where CXCL9 had an AUC of 0.94 (0.86-1.00) for AIN diagnosis. CXCL9 mRNA expression was 3.9-fold higher in kidney tissue from patients with AIN (n = 19) compared with individuals in the control group (n = 52; P = 5.8 × 10-6).ConclusionWe identified CXCL9 as a diagnostic biomarker for AIN using aptamer-based urine proteomics, confirmed this association using sandwich immunoassays in discovery and external validation cohorts, and observed higher expression of this protein in kidney biopsies from patients with AIN.FundingThis study was supported by National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) awards K23DK117065 (DGM), K08DK113281 (KM), R01DK128087 (DGM), R01DK126815 (DGM and LGC), R01DK126477 (KNC), UH3DK114866 (CRP, DGM, and FPW), R01DK130839 (MES), and P30DK079310 (the Yale O'Brien Center). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Collapse
Affiliation(s)
- Dennis G. Moledina
- Section of Nephrology, Department of Internal Medicine and
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Wassim Obeid
- Division of Nephrology, Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Rex N. Smith
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
- Immunopathology Research Laboratory and
| | - Ivy Rosales
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
- Immunopathology Research Laboratory and
| | - Meghan E. Sise
- Section of Nephrology, Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gilbert Moeckel
- Section of Renal Pathology, Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael Kashgarian
- Section of Renal Pathology, Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Kirk N. Campbell
- Division of Nephrology, Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sean Lefferts
- Division of Nephrology, Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristin Meliambro
- Division of Nephrology, Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Markus Bitzer
- Section of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Jordan S. Pober
- Department of Pathology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Robert B. Colvin
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
- Immunopathology Research Laboratory and
| | - F. Perry Wilson
- Section of Nephrology, Department of Internal Medicine and
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Chirag R. Parikh
- Division of Nephrology, Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Gevers-Montoro C, Puente-Tobares M, Monréal A, Conesa-Buendía FM, Piché M, Ortega-De Mues A. Urinary TNF-α as a potential biomarker for chronic primary low back pain. Front Integr Neurosci 2023; 17:1207666. [PMID: 37449008 PMCID: PMC10336221 DOI: 10.3389/fnint.2023.1207666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/06/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Over two thirds of individuals with low back pain (LBP) may experience recurrent or persistent symptoms in the long term. Yet, current data do not allow to predict who will develop chronic low back pain and who will recover from an acute episode. Elevated serum levels of the proinflammatory cytokine tumor necrosis factor-α (TNF-α) have been associated with poor recovery and persistent pain following an acute episode of LBP. Inflammatory cytokines may also mediate mechanisms involved in nociplastic pain, and thus, have significant implications in chronic primary low back pain (CPLBP). Methods This study aimed to investigate the potential of urinary TNF-α levels for predicting outcomes and characterizing clinical features of CPLBP patients. Twenty-four patients with CPLBP and 24 sex- and age-matched asymptomatic controls were recruited. Urinary TNF-α concentrations were measured at baseline and after 4 weeks, during which CPLBP patients underwent spinal manipulative therapy (SMT). Results Concentrations of TNF-α were found to be elevated in baseline urine samples of CPLBP patients compared to asymptomatic controls. Moreover, these values differed among patients depending on their pain trajectory. Patients with persistent pain showed higher levels of TNF-α, when compared to those with episodic CPLBP. Furthermore, baseline TNF-α concentrations and their changes after 4 weeks predicted alterations in pain intensity and disability following SMT in patients with CPLBP. Discussion These findings warrant further research on the potential use of urinary TNF-α concentrations as a prognostic biomarker for CPLBP.
Collapse
Affiliation(s)
- Carlos Gevers-Montoro
- Madrid College of Chiropractic – RCU María Cristina, Madrid, Spain
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
- CogNAC Research Group, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | | | - Aléxiane Monréal
- Madrid College of Chiropractic – RCU María Cristina, Madrid, Spain
| | | | - Mathieu Piché
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
- CogNAC Research Group, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | | |
Collapse
|
37
|
Zhou XJ, Su T, Xie J, Xie QH, Wang LZ, Hu Y, Chen G, Jia Y, Huang JW, Li G, Liu Y, Yu XJ, Nath SK, Tsoi LC, Patrick MT, Berthier CC, Liu G, Wang SX, Xu H, Chen N, Hao CM, Zhang H, Yang L. Genome-Wide Association Study in Acute Tubulointerstitial Nephritis. J Am Soc Nephrol 2023; 34:895-908. [PMID: 36749126 PMCID: PMC10125656 DOI: 10.1681/asn.0000000000000091] [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: 07/21/2022] [Accepted: 12/28/2022] [Indexed: 02/08/2023] Open
Abstract
SIGNIFICANCE STATEMENT Polymorphisms of HLA genes may confer susceptibility to acute tubulointerstitial nephritis (ATIN), but small sample sizes and candidate gene design have hindered their investigation. The first genome-wide association study of ATIN identified two significant loci, risk haplotype DRB1*14-DQA1*0101-DQB1*0503 (DR14 serotype) and protective haplotype DRB1*1501-DQA1*0102-DQB1*0602 (DR15 serotype), with amino acid position 60 in the peptide-binding groove P10 of HLA-DR β 1 key. Risk alleles were shared among different causes of ATIN and HLA genotypes associated with kidney injury and immune therapy response. HLA alleles showed the strongest association. The findings suggest that a genetically conferred risk of immune dysregulation is part of the pathogenesis of ATIN. BACKGROUND Acute tubulointerstitial nephritis (ATIN) is a rare immune-related disease, accounting for approximately 10% of patients with unexplained AKI. Previous elucidation of the relationship between genetic factors that contribute to its pathogenesis was hampered because of small sample sizes and candidate gene design. METHODS We undertook the first two-stage genome-wide association study and meta-analysis involving 544 kidney biopsy-defined patients with ATIN and 2346 controls of Chinese ancestry. We conducted statistical fine-mapping analysis, provided functional annotations of significant variants, estimated single nucleotide polymorphism (SNP)-based heritability, and checked genotype and subphenotype correlations. RESULTS Two genome-wide significant loci, rs35087390 of HLA-DQA1 ( P =3.01×10 -39 ) on 6p21.32 and rs2417771 of PLEKHA5 on 12p12.3 ( P =2.14×10 -8 ), emerged from the analysis. HLA imputation using two reference panels suggested that HLA-DRB1*14 mainly drives the HLA risk association . HLA-DRB1 residue 60 belonging to pocket P10 was the key amino acid position. The SNP-based heritability estimates with and without the HLA locus were 20.43% and 10.35%, respectively. Different clinical subphenotypes (drug-related or tubulointerstitial nephritis and uveitis syndrome) seemed to share the same risk alleles. However, the HLA risk genotype was associated with disease severity and response rate to immunosuppressive therapy. CONCLUSIONS We identified two candidate genome regions associated with susceptibility to ATIN. The findings suggest that a genetically conferred risk of immune dysregulation is involved in the pathogenesis of ATIN.
Collapse
Affiliation(s)
- Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Tao Su
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Jingyuan Xie
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiong-Hong Xie
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Li-Zhong Wang
- WeGene, Shenzhen Zaozhidao Technology Co., Ltd., Shenzhen, China
- Human Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China
| | - Yong Hu
- Beijing Institute of Biotechnology, Beijing, China
| | - Gang Chen
- WeGene, Shenzhen Zaozhidao Technology Co., Ltd., Shenzhen, China
- Human Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China
| | - Yan Jia
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Jun-Wen Huang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Gui Li
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Yang Liu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Xiao-Juan Yu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Swapan K. Nath
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Matthew T. Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Celine C. Berthier
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gang Liu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Su-Xia Wang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Nan Chen
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuan-Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Li Yang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
38
|
Njaanake KH, Omondi J, Mwangi I, Jaoko WG, Anzala O. Urinary interleukins (IL)-6 and IL-10 in schoolchildren from an area with low prevalence of Schistosoma haematobium infections in coastal Kenya. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001726. [PMID: 37018184 PMCID: PMC10075406 DOI: 10.1371/journal.pgph.0001726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/24/2023] [Indexed: 04/12/2023]
Abstract
Urinary cytokines are gaining traction as tools for assessing morbidity in infectious and non-infectious inflammatory diseases of the urogenital tract. However, little is known about the potential of these cytokines in assessing morbidity due to S. haematobium infections. Factors that may influence the urinary cytokine levels as morbidity markers also remain unknown. Therefore the objective of the present study was to assess how urinary interleukins (IL-) 6 and 10 are associated with gender, age, S. haematobium infections, haematuria and urinary tract pathology and; 2) to assess the effects of urine storage temperatures on the cytokines. This was a cross-sectional study in 2018 involving 245 children aged 5-12 years from a S. haematobium endemic area of coastal Kenya. The children were examined for S. haematobium infections, urinary tract morbidity, haematuria and urinary cytokines (IL-6 and IL-10). Urine specimens were also stored at -20°C, 4°C or 25°C for 14 days before being assayed for IL6 and IL-10 using ELISA. The overall prevalence of S. haematobium infections, urinary tract pathology, haematuria, urinary IL-6 and urinary IL-10 were 36.3%, 35.8%, 14.8%, 59.4% and 80.5%, respectively. There were significant associations between prevalence of urinary IL-6, but not IL-10, and age, S. haematobium infection and haematuria (p = 0.045, 0.011 and 0.005, respectively) but not sex or ultrasound-detectable pathology. There were significant differences in IL-6 and IL-10 levels between urine specimens stored at -20°C and those stored at 4°C (p<0.001) and, between those stored at 4°C and those stored at 25°C (p<0.001). Urinary IL-6, but not IL-10, was associated with children's age, S. haematobium infections and haematuria. However, both urinary IL-6 and IL-10 were not associated with urinary tract morbidity. Both IL-6 and IL-10 were sensitive to urine storage temperatures.
Collapse
Affiliation(s)
- Kariuki H. Njaanake
- Department of Medical Microbiology, College of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI- Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Job Omondi
- Department of Medical Microbiology, College of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - Irene Mwangi
- KAVI- Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Walter G. Jaoko
- Department of Medical Microbiology, College of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI- Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Omu Anzala
- Department of Medical Microbiology, College of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI- Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
39
|
Longhitano E, Muscolino P, Lo Re C, Ferrara SA, Cernaro V, Gembillo G, Tessitore D, Speranza D, Figura F, Santarpia M, Silvestris N, Santoro D, Franchina T. Immune Checkpoint Inhibitors and the Kidney: A Focus on Diagnosis and Management for Personalised Medicine. Cancers (Basel) 2023; 15:1891. [PMID: 36980777 PMCID: PMC10046877 DOI: 10.3390/cancers15061891] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Immunity plays a crucial role in fighting cancer, but tumours can evade the immune system and proliferate and metastasize. Enhancing immune responses is a new challenge in anticancer therapies. In this context, efficacy data are accumulating on immune checkpoint inhibitors and adjuvant therapies for various types of advanced-stage solid tumours. Unfortunately, immune-related adverse events are common. Although infrequent, renal toxicity may occur via several mechanisms and may require temporary or permanent drug suspension, renal biopsy, and/or immunosuppressive treatment. This short review aims to provide a practical approach to the multidisciplinary management of cancer patients with renal toxicity during treatment with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Elisa Longhitano
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. “G. Martino”, University of Messina, 98125 Messina, Italy
| | - Paola Muscolino
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Claudia Lo Re
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. “G. Martino”, University of Messina, 98125 Messina, Italy
| | - Serena Ausilia Ferrara
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. “G. Martino”, University of Messina, 98125 Messina, Italy
| | - Valeria Cernaro
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. “G. Martino”, University of Messina, 98125 Messina, Italy
| | - Guido Gembillo
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. “G. Martino”, University of Messina, 98125 Messina, Italy
| | - Dalila Tessitore
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Desirèe Speranza
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Francesco Figura
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Domenico Santoro
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. “G. Martino”, University of Messina, 98125 Messina, Italy
| | - Tindara Franchina
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| |
Collapse
|
40
|
Farooqui N, Zaidi M, Vaughan L, McKee TD, Ahsan E, Pavelko KD, Villasboas JC, Markovic S, Taner T, Leung N, Dong H, Alexander MP, Herrmann SM. Cytokines and Immune Cell Phenotype in Acute Kidney Injury Associated With Immune Checkpoint Inhibitors. Kidney Int Rep 2023; 8:628-641. [PMID: 36938084 PMCID: PMC10014345 DOI: 10.1016/j.ekir.2022.11.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/25/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction Immune checkpoint inhibitors (ICIs) induce impressive antitumor responses but may lead to acute kidney injury (AKI) associated with ICI therapy (AKI-ICI). Biomarkers distinguishing AKI-ICI from AKI because of other causes (AKI-other) are currently lacking. Because ICIs block immunoregulatory pathways, we hypothesized that biomarkers related to immune cell dysregulation, including tumor necrosis factor alpha (TNF-α) and other markers of B and T cell activation in the systemic circulation and kidney tissue, may aid with the diagnosis of AKI-ICI. Methods This is a prospective study consisting of 24 participants who presented with AKI during ICI therapy, adjudicated to either have AKI-ICI (n = 14) or AKI-other (n = 10). We compared markers of kidney inflammation and injury (neutrophil gelatinase-associated lipocalin, kidney injury molecule-1) as well as plasma and urine levels of T cell-associated cytokines (TNF-α, interferon-γ, interleukin (IL)-2, IL-4, IL-6, IL-8, IL-9, and IL-10) between groups. We also compared T-cell responses in the systemic circulation and in kidney tissue across groups, using mass cytometry systems. Results We observed increase in several specific immune cells, including CD4 memory, T helper cells, and dendritic cells in the kidney tissue, as well as in the urine cytokines IL-2, IL-10, and TNF-α, in patients who developed AKI-ICI compared to patients with AKI-other (P < 0.05 for all). The discriminatory ability of TNF-α on AKI cause was strong (area under the curve = 0.814, 95% confidence interval: 0.623-1.00. The CD4+ T cells with memory phenotype formed the dominant subset. Conclusion These results suggest that specific T-cell responses and their respective cytokines may be indicative of AKI associated with ICI therapy and may help to differentiate AKI-ICI from AKI-other. Urine TNF-α is a promising biomarker for AKI-ICI, which is most often caused by acute interstitial nephritis (AIN), and TNF-α pathway may serve as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Naba Farooqui
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark Zaidi
- Department of Medical Biophysics, University of Toronto, Ontario, Canada
| | - Lisa Vaughan
- Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Trevor D. McKee
- Department of Medical Biophysics, University of Toronto, Ontario, Canada
- Deciphex Inc., Chicago, Illinois, USA
| | - Eram Ahsan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin D. Pavelko
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | | | | | - Timucin Taner
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Haidong Dong
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Mariam P. Alexander
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sandra M. Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
41
|
Rodrigues CE, Endre ZH. Definitions, phenotypes, and subphenotypes in acute kidney injury-Moving towards precision medicine. Nephrology (Carlton) 2023; 28:83-96. [PMID: 36370326 PMCID: PMC10100386 DOI: 10.1111/nep.14132] [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: 06/24/2022] [Revised: 10/23/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022]
Abstract
The current definition of acute kidney injury (AKI) is generic and, based only on markers of function, is unsuitable for guiding individualized treatment. AKI is a complex syndrome with multiple presentations and causes. Targeted AKI management will only be possible if different phenotypes and subphenotypes of AKI are recognised, based on causation and related pathophysiology. Molecular signatures to identify subphenotypes are being recognised, as specific biomarkers reveal activated pathways. Assessment of individual clinical risk needs wider dissemination to allow identification of patients at high risk of AKI. New and more timely markers for glomerular filtration rate (GFR) are available. However, AKI diagnosis and classification should not be limited to GFR, but include tubular function and damage. Combining damage and stress biomarkers with functional markers enhances risk prediction, and identifies a population enriched for clinical trials targeting AKI. We review novel developments and aim to encourage implementation of these new techniques into clinical practice as a strategy for individualizing AKI treatment akin to a precision medicine-based approach.
Collapse
Affiliation(s)
- Camila Eleuterio Rodrigues
- Nephrology DepartmentPrince of Wales Clinical School – UNSW MedicineSydneyNew South WalesAustralia
- Nephrology DepartmentHospital das Clínicas – University of São Paulo School of MedicineSão PauloBrazil
| | - Zoltán H. Endre
- Nephrology DepartmentPrince of Wales Clinical School – UNSW MedicineSydneyNew South WalesAustralia
| |
Collapse
|
42
|
Sise ME, Wang Q, Seethapathy H, Moreno D, Harden D, Smith RN, Rosales IA, Colvin RB, Chute S, Cornell LD, Herrmann SM, Fadden R, Sullivan RJ, Yang NJ, Barmettler S, Wells S, Gupta S, Villani AC, Reynolds KL, Farmer J. Soluble and cell-based markers of immune checkpoint inhibitor-associated nephritis. J Immunother Cancer 2023; 11:e006222. [PMID: 36657813 PMCID: PMC9853261 DOI: 10.1136/jitc-2022-006222] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2023] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Non-invasive biomarkers of immune checkpoint inhibitor-associated acute tubulointerstitial nephritis (ICI-nephritis) are urgently needed. Because ICIs block immune checkpoint pathways that include cytotoxic T lymphocyte antigen 4 (CTLA4), we hypothesized that biomarkers of immune dysregulationpreviously defined in patients with congenital CTLA4 deficiency, including elevated soluble interleukin-2 receptor alpha (sIL-2R) and flow cytometric cell-based markers of B and T cell dysregulation in peripheral blood may aid the diagnosis of ICI-nephritis. METHODS A retrospective cohort of patients diagnosed with ICI-nephritis was compared with three prospectively enrolled control cohorts: ICI-treated controls without immune-related adverse events, patients not on ICIs with hemodynamic acute kidney injury (hemodynamic AKI), and patients not on ICIs with biopsy proven acute interstitial nephritis from other causes (non-ICI-nephritis). sIL-2R level and flow cytometric parameters were compared between groups using Wilcoxon rank sum test or Kruskal-Wallis test. Receiver operating characteristic curves were generated to define the accuracy of sIL-2R and flow cytometric biomarkers in diagnosing ICI-nephritis. The downstream impact of T cell activation in the affected kidney was investigated using archived biopsy samples to evaluate the gene expression of IL2RA, IL-2 signaling, and T cell receptor signaling in patients with ICI-nephritis compared with other causes of drug-induced nephritis, acute tubular injury, and histologically normal controls. RESULTS sIL-2R level in peripheral blood was significantly higher in patients with ICI-nephritis (N=24) (median 2.5-fold upper limit of normal (ULN), IQR 1.9-3.3), compared with ICI-treated controls (N=10) (median 0.8-fold ULN, IQR 0.5-0.9, p<0.001) and hemodynamic AKI controls (N=6) (median 0.9-fold-ULN, IQR 0.7-1.1, p=0.008). A sIL-2R cut-off point of 1.75-fold ULN was highly diagnostic of ICI-nephritis (area under the curve >96%) when compared with either ICI-treated or hemodynamic AKI controls. By peripheral blood flow cytometry analysis, lower absolute CD8+T cells, CD45RA+CD8+ T cells, memory CD27+B cells, and expansion of plasmablasts were prominent features of ICI-nephritis compared with ICI-treated controls. Gene expressions for IL2RA, IL-2 signaling, and T cell receptor signaling in the kidney tissue with ICI-nephritis were significantly higher compared with controls. CONCLUSION Elevated sIL-2R level and flow cytometric markers of both B and T cell dysregulation may aid the diagnosis of ICI-nephritis.
Collapse
Affiliation(s)
- Meghan E Sise
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Qiyu Wang
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Harish Seethapathy
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daiana Moreno
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Destiny Harden
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - R Neal Smith
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robert B Colvin
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sarah Chute
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lynn D Cornell
- Department of Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sandra M Herrmann
- Department of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Riley Fadden
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ryan J Sullivan
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nancy J Yang
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sara Barmettler
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sophia Wells
- Department of Medicine, Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Shruti Gupta
- Department of Medicine, Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Oncology, Adult Survivorship Program, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Alexandra-Chloe Villani
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kerry L Reynolds
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jocelyn Farmer
- Division of Allergy and Inflammation, Beth Israel Lahey Health, Boston, Massachusetts, USA
| |
Collapse
|
43
|
Lei R, Vu B, Kourentzi K, Soomro S, Danthanarayana AN, Brgoch J, Nadimpalli S, Petri M, Mohan C, Willson RC. A novel technology for home monitoring of lupus nephritis that tracks the pathogenic urine biomarker ALCAM. Front Immunol 2022; 13:1044743. [PMID: 36569940 PMCID: PMC9780296 DOI: 10.3389/fimmu.2022.1044743] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Introduction The gold standard for diagnosis of active lupus nephritis (ALN), a kidney biopsy, is invasive with attendant morbidity and cannot be serially repeated. Urinary ALCAM (uALCAM) has shown high diagnostic accuracy for renal pathology activity in ALN patients. Methods Lateral flow assays (LFA) for assaying uALCAM were engineered using persistent luminescent nanoparticles, read by a smartphone. The stability and reproducibility of the assembled LFA strips and freeze-dried conjugated nanoparticles were verified, as was analyte specificity. Results The LFA tests for both un-normalized uALCAM (AUC=0.93) and urine normalizer (HVEM)-normalized uALCAM (AUC=0.91) exhibited excellent accuracies in distinguishing ALN from healthy controls. The accuracies for distinguishing ALN from all other lupus patients were 0.86 and 0.74, respectively. Conclusion Periodic monitoring of uALCAM using this easy-to-use LFA test by the patient at home could potentially accelerate early detection of renal involvement or disease flares in lupus patients, and hence reduce morbidity and mortality.
Collapse
Affiliation(s)
- Rongwei Lei
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Binh Vu
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, United States
| | - Katerina Kourentzi
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, United States
| | - Sanam Soomro
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | | | - Jakoah Brgoch
- Department of Chemistry, University of Houston, Houston, TX, United States
| | - Suma Nadimpalli
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Richard C. Willson
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, United States
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
- Escuela de Medicina y Ciencias de Salud, Tecnológico de Monterrey, Monterrey, NL, Mexico
| |
Collapse
|
44
|
Medeiros T, Alves LS, Cabral-Castro MJ, Silva ARO, Xavier AR, Burger D, Almeida JR, Silva AA. Exploring Urinary Extracellular Vesicles and Immune Mediators as Biomarkers of Kidney Injury in COVID-19 Hospitalized Patients. Diagnostics (Basel) 2022; 12:diagnostics12112600. [PMID: 36359444 PMCID: PMC9689919 DOI: 10.3390/diagnostics12112600] [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: 09/05/2022] [Revised: 10/11/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Kidney injury is an important outcome associated with COVID-19 severity. In this regard, alterations in urinary extracellular vesicles (uEVs) could be detected in the early phases of renal injury and may be reflective of the inflammatory process. This is an observational study performed with a case series of COVID-19 hospitalized patients presenting mild-to-critical disease. Total and podocyte-derived uEVs were identified by nanoscale flow cytometry, and urinary immune mediators were assessed by a multiplex assay. We studied 36 patients, where 24 (66.7%) were considered as mild/moderate and 12 (33.3%) as severe/critical. Increased levels of total uEVs were observed (p = 0.0001). Importantly, total uEVs were significantly higher in severe/critical patients who underwent hemodialysis (p = 0.03) and were able to predict this clinical outcome (AUC 0.93, p = 0.02). Severe/critical patients also presented elevated urinary levels (p < 0.05) of IL-1β, IL-4, IL-6, IL-7, IL-16, IL-17A, LIF, CCL-2, CCL-3, CCL-11, CXCL-10, FGFb, M-CSF, and CTAcK. Lastly, we observed that total uEVs were associated with urinary immune mediators. In conclusion, our results show that early alterations in urinary EVs could identify patients at higher risk of developing renal dysfunction in COVID-19. This could also be relevant in different scenarios of systemic and/or infectious disease.
Collapse
Affiliation(s)
- Thalia Medeiros
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences (LAMAP), Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
- Department of Pathology, Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
- Correspondence: (T.M.); (A.A.S.); Tel.: +55-21-3674-7282 (A.A.S.)
| | - Lilian Santos Alves
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences (LAMAP), Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
| | - Mauro Jorge Cabral-Castro
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences (LAMAP), Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Rio de Janeiro, Brazil
| | - Alice Ramos Oliveira Silva
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences (LAMAP), Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
| | - Analúcia Rampazzo Xavier
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences (LAMAP), Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
- Department of Pathology, Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
| | - Dylan Burger
- Kidney Research Centre, Department of Cellular and Molecular Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jorge Reis Almeida
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences (LAMAP), Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
- Department of Clinical Medicine; Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
| | - Andrea Alice Silva
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences (LAMAP), Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
- Department of Pathology, Faculty of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Rio de Janeiro, Brazil
- Correspondence: (T.M.); (A.A.S.); Tel.: +55-21-3674-7282 (A.A.S.)
| |
Collapse
|
45
|
Miao J, Sise ME, Herrmann SM. Immune checkpoint inhibitor related nephrotoxicity: Advances in clinicopathologic features, noninvasive approaches, and therapeutic strategy and rechallenge. FRONTIERS IN NEPHROLOGY 2022; 2:1017921. [PMID: 37674988 PMCID: PMC10479679 DOI: 10.3389/fneph.2022.1017921] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/28/2022] [Indexed: 09/08/2023]
Abstract
Immune checkpoint inhibitors (ICIs) are used increasingly to treat more than 17 cancers and have shown promising therapeutic results. However, ICI use can result in a variety of immune-related adverse events (IRAEs) which can occur in any organ, including the kidneys. Acute kidney injury (AKI) is the most common nephrotoxicity, classically related to acute interstitial nephritis. Much more diverse patterns and presentations of ICI-related kidney injury can occur, and have implications for diagnostic and therapeutic management approaches. In this review, we summarize the recently approved ICIs for cancer, the incidence and risk factors for nephrotoxicity, our current understanding of the pathophysiological mechanisms and the key clinicopathological features of ICI-related AKI, and therapeutic strategies. We also explore important knowledge that require further investigation, such as the risks/benefits of ICI rechallenge in patients who recover from an episode of ICI-related AKI, and the application of liquid biopsy and microbiome to identify noninvasive biomarkers to diagnose and predict kidney injury and guide ICI therapy.
Collapse
Affiliation(s)
- Jing Miao
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Meghan E. Sise
- Department of Internal Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, MA, United States
| | - Sandra M. Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
46
|
Moledina DG, Eadon MT, Calderon F, Yamamoto Y, Shaw M, Perazella MA, Simonov M, Luciano R, Schwantes-An TH, Moeckel G, Kashgarian M, Kuperman M, Obeid W, Cantley LG, Parikh CR, Wilson FP. Development and external validation of a diagnostic model for biopsy-proven acute interstitial nephritis using electronic health record data. Nephrol Dial Transplant 2022; 37:2214-2222. [PMID: 34865148 PMCID: PMC9755995 DOI: 10.1093/ndt/gfab346] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Patients with acute interstitial nephritis (AIN) can present without typical clinical features, leading to a delay in diagnosis and treatment. We therefore developed and validated a diagnostic model to identify patients at risk of AIN using variables from the electronic health record. METHODS In patients who underwent a kidney biopsy at Yale University between 2013 and 2018, we tested the association of >150 variables with AIN, including demographics, comorbidities, vital signs and laboratory tests (training set 70%). We used least absolute shrinkage and selection operator methodology to select prebiopsy features associated with AIN. We performed area under the receiver operating characteristics curve (AUC) analysis with internal (held-out test set 30%) and external validation (Biopsy Biobank Cohort of Indiana). We tested the change in model performance after the addition of urine biomarkers in the Yale AIN study. RESULTS We included 393 patients (AIN 22%) in the training set, 158 patients (AIN 27%) in the test set, 1118 patients (AIN 11%) in the validation set and 265 patients (AIN 11%) in the Yale AIN study. Variables in the selected model included serum creatinine {adjusted odds ratio [aOR] 2.31 [95% confidence interval (CI) 1.42-3.76]}, blood urea nitrogen:creatinine ratio [aOR 0.40 (95% CI 0.20-0.78)] and urine dipstick specific gravity [aOR 0.95 (95% CI 0.91-0.99)] and protein [aOR 0.39 (95% CI 0.23-0.68)]. This model showed an AUC of 0.73 (95% CI 0.64-0.81) in the test set, which was similar to the AUC in the external validation cohort [0.74 (95% CI 0.69-0.79)]. The AUC improved to 0.84 (95% CI 0.76-0.91) upon the addition of urine interleukin-9 and tumor necrosis factor-α. CONCLUSIONS We developed and validated a statistical model that showed a modest AUC for AIN diagnosis, which improved upon the addition of urine biomarkers. Future studies could evaluate this model and biomarkers to identify unrecognized cases of AIN.
Collapse
Affiliation(s)
| | - Michael T Eadon
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Frida Calderon
- Section of Nephrology and Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Yu Yamamoto
- Section of Nephrology and Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Melissa Shaw
- Section of Nephrology and Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Mark A Perazella
- Section of Nephrology and Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Michael Simonov
- Section of Nephrology and Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Randy Luciano
- Section of Nephrology and Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Gilbert Moeckel
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | | | | | | | - Lloyd G Cantley
- Section of Nephrology and Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | | |
Collapse
|
47
|
Sprangers B, Leaf DE, Porta C, Soler MJ, Perazella MA. Diagnosis and management of immune checkpoint inhibitor-associated acute kidney injury. Nat Rev Nephrol 2022; 18:794-805. [PMID: 36168055 DOI: 10.1038/s41581-022-00630-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 11/10/2022]
Abstract
Since their introduction into clinical practice a decade ago, immune checkpoint inhibitors (ICIs) have had an overwhelming impact on cancer treatment. Use of these agents in oncology continues to grow; however, the increased use of these agents has been associated with a parallel increase in ICI-associated immune-related adverse events, which can affect virtually any organ, including the kidneys. ICI-associated acute kidney injury (ICI-AKI) occurs in 2-5% of patients treated with ICIs. Its occurrence can have important consequences, including the temporary or permanent discontinuation of ICIs or other concomitant anticancer therapies and the need for prolonged treatment with corticosteroids. Various mechanisms have been proposed to underlie the development of ICI-AKI, including loss of tolerance to self-antigens, reactivation of drug-specific effector T cells, and the production of kidney-specific autoantibodies. ICI-AKI most commonly manifests as acute tubulo-interstitial nephritis on kidney biopsy and generally shows a favourable response to early initiation of corticosteroids, with complete or partial remission achieved in most patients. The evaluation of patients with suspected ICI-AKI requires careful diagnostic work-up and kidney biopsy for patients with moderate-to-severe ICI-AKI to ensure accurate diagnosis and inform appropriate treatment.
Collapse
Affiliation(s)
- Ben Sprangers
- Division of Nephrology, Ziekenhuis Oost-Limburg, Genk, Belgium. .,Biomedical Research Institute, Department of Immunology and Infection, UHasselt, Diepenbeek, Belgium.
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Camillo Porta
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Corsorziale Policlinico di Bari, Bari, Italy.,Oncology, Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Maria José Soler
- Nephrology Research Group, Vall d'hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Mark A Perazella
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut, USA.,Veterans Affairs Medical Center, West Haven, Connecticut, USA
| |
Collapse
|
48
|
Claure-Del Granado R, Macedo E, Chávez-Íñiguez JS. Biomarkers for Early Diagnosis of AKI: Could It Backfire? KIDNEY360 2022; 3:1780-1784. [PMID: 36514722 PMCID: PMC9717653 DOI: 10.34067/kid.0001012022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/29/2022] [Indexed: 01/12/2023]
Affiliation(s)
- Rolando Claure-Del Granado
- Division of Nephrology Hospital Obrero No. 2—CNS, Cochabamba, Bolivia,IIBISMED, Universidad Mayor de San Simon, School of Medicine, Cochabamba, Bolivia
| | - Etienne Macedo
- Division of Nephrology-Hypertension University of California, San Diego, San Diego, California
| | - Jonathan S. Chávez-Íñiguez
- Division of Nephrology Hospital Civil de Guadalajara “Fray Antonio Alcalde,” Guadalajara, Jalisco, Mexico,University of Guadalajara Health Sciences Center, Guadalajara, Mexico
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICIs) have changed the landscape of cancer treatment. However, use of ICIs can be limited by inflammatory toxicities referred to as immune-related adverse events (irAEs). ICI-associated acute kidney injury (ICI-associated AKI) affects 3-5% of ICI users. RECENT FINDINGS With the rapidly growing indication of ICI, knowledge of ICI-associated kidney toxicity has also expanded from case series to large multicentre cohort studies. In this review, we discuss the clinical features, risk factors, clinicopathological correlations and prognosis of ICI-associated AKI from the most recent rigorously conducted retrospective cohort studies. We also discuss recent advances in diagnostic biomarker investigation, treatment and the unique challenge faced in the kidney transplant population. SUMMARY With more comprehensive understanding of the clinical features and risk factors, ICI-associated AKI is commonly diagnosed clinically, especially given the inherent challenges performing a kidney biopsy in the cancer population; however, this highlights the urgent need for improved noninvasive diagnostic biomarkers to aid diagnosis and prognosis. Prospective studies are needed to better define the optimal treatment of ICI-associated AKI and to minimize the risk of graft loss in patients with kidney transplant who require ICIs.
Collapse
|
50
|
Melchinger H, Calderon-Gutierrez F, Obeid W, Xu L, Shaw MM, Luciano RL, Kuperman M, Moeckel GW, Kashgarian M, Wilson FP, Parikh CR, Moledina DG. Urine Uromodulin as a Biomarker of Kidney Tubulointerstitial Fibrosis. Clin J Am Soc Nephrol 2022; 17:1284-1292. [PMID: 35948365 PMCID: PMC9625093 DOI: 10.2215/cjn.04360422] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/15/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND OBJECTIVES Uromodulin, produced exclusively in the kidney's thick ascending limb, is a biomarker of kidney tubular health. However, the relationship between urine uromodulin and histologic changes in the kidney tubulointerstitium has not been characterized. In this study, we test the association of urine uromodulin with kidney histologic findings in humans and mice. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We investigated the independent association of urine uromodulin measured at the time of kidney biopsy with histologic features in 364 participants at two academic medical centers from 2015 to 2018 using multivariable linear regression models. This relationship was further examined by comparison of uromodulin staining in murine models of kidney fibrosis and repair. RESULTS We found urine uromodulin to be correlated with serum creatinine (rho=-0.43; P<0.001), bicarbonate (0.20; P<0.001), and hemoglobin (0.11; P=0.03) at the time of biopsy but not with urine albumin (-0.07; P=0.34). Multivariable models controlling for prebiopsy GFR, serum creatinine at biopsy, and urine albumin showed higher uromodulin to be associated with lower severity of interstitial fibrosis/tubular atrophy and glomerulosclerosis (interstitial fibrosis/tubular atrophy: -3.5% [95% confidence intervals, -5.7% to -1.2%] and glomerulosclerosis: -3.3% [95% confidence intervals, -5.9% to -0.6%] per two-fold difference in uromodulin). However, when both interstitial fibrosis/tubular atrophy and glomerulosclerosis were included in multivariable analysis, only interstitial fibrosis/tubular atrophy was independently associated with uromodulin (interstitial fibrosis/tubular atrophy: -2.5% [95% confidence intervals, -4.6% to -0.4%] and glomerulosclerosis: -0.9% [95% confidence intervals, -3.4% to 1.5%] per two-fold difference in uromodulin). In mouse kidneys, uromodulin staining was found to be lower in the fibrotic model than in normal or repaired models. CONCLUSIONS Higher urine uromodulin is independently associated with lower tubulointerstitial fibrosis in both human kidney biopsies and a mouse model of fibrosis. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2022_08_10_CJN04360422.mp3.
Collapse
Affiliation(s)
- Hannah Melchinger
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Frida Calderon-Gutierrez
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Wassim Obeid
- Division of Nephrology, Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Leyuan Xu
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Melissa M. Shaw
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Randy L. Luciano
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Michael Kuperman
- Division of Nephropathology, Arkana Laboratories, Little Rock, Arkansas
| | - Gilbert W. Moeckel
- Section of Renal Pathology, Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Michael Kashgarian
- Section of Renal Pathology, Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - F. Perry Wilson
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Chirag R. Parikh
- Division of Nephrology, Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Dennis G. Moledina
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|