European Journal of Internal Medicine xxx (xxxx) xxx Contents lists available at ScienceDirect European Journal of Internal Medicine journal homepage: www.elsevier.com/locate/ejim Review Article Toxic nephropathy: Adverse renal effects caused by drugs Robert J. Unwin Department of Renal Medicine, Royal Free Hospital Trust, University College London, Rowland Hill Street, London NW3 2PF, UK A R T I C L E I N F O A B S T R A C T Keywords: Drug toxicity Nephrotoxicity Kidney Proximal tubule Mitochondria This is a brief overview of toxic nephropathy, which is an increasingly recognised problem with the continual introduction of new drugs and novel drug modalities, especially in oncology, and the risks associated with polypharmacy in many patients; although it is important to remember that it may not always be caused by a drug. It is also important to note that several possibly harmful drugs are now available without prescription (‘over-the-counter’) and can be purchased easily over the internet, including some poorly characterised herbal remedies. Knowing exactly what our patients are taking as medication is not always easy and patients often fail to mention drugs that may not have been prescribed by a doctor or recommended by a pharmacist. Moreover, patients with several comorbidities often require care from more than one doctor in other specialties, which can also lead to drug prescribing in isolation. This article will summarise some key aspects of drug nephrotoxicity and provide a few clinical pointers to consider, bearing in mind that there is rarely any antidote available, and effective treatment relies on early detection, prompt drug withdrawal, and supportive care. This short review is intended only as a primer to highlight some of the more practical aspects of toxic nephropathy; its content is based on a lecture delivered during the 2021 European Congress of Internal Medicine. 1. Introduction According to Paracelsus, the Swiss Physician in the 1500s who has been likened to Hippocrates, ‘only the dose makes the poison’; while this is still largely true for most drugs or known poisons, it is not so for many idiosyncratic or ‘allergic’ drug reactions, or for those compounds with a narrow margin of safety (MOS).1 The kidneys are often the main target organ affected by many poisons and drug-related adverse reactions; however, the topic of toxic nephropathy is potentially very broad, since there is a lot that we cannot know or predict about potential environ­ mental toxins or until some time after a new therapeutic agent has been introduced clinically and used on a large-scale, which can take several years of post-marketing surveillance. Therefore, vigilance is paramount and it is important to be aware of some general principles when it comes to deciding whether an ingested product or new drug is a cause of renal injury. I will try to summarise some topical aspects, including acute versus chronic kidney injury, and cover nephrotoxicity more generally, as well as some specific developments with already known potential drug nephrotoxins still used widely in clinical practice. I will also consider the ongoing and slightly contentious issue of ’analgesic nephropathy’, 1 especially in relation to commonly prescribed non-steroidal anti-in­ flammatory drugs (NSAID). 2. Early history and some examples Since mediaeval times, poisoning has been a popular means of murder or assassination, and if not sudden or immediate, often kidney failure has been the likely and eventual cause of death, although difficult to confirm. Commonly used poisons for this purpose have been arsenic and strychnine, which can both affect the kidneys. Arsenic, as for other heavy metals, which are all potential environmental toxins [1], can cause acute and chronic renal toxicity. The free ionised form is directly toxic to renal tubular cells, although the means by which it enters these cells is still unclear, but may involve the glucose transporter GLUT1 [2] present in the proximal tubule, where As3+ then depletes cellular glutathione, triggers inflammatory cytokine release, and disrupts mito­ chondrial function [3]; chronic toxicity results from the protein bound and inert form that is conjugated to metallothioneins and glutathione in the liver (which also expresses GLUT1, as well AQP9, a more important uptake mechanism for arsenic and confined to hepatocytes [4]), which when released into the circulation is again reabsorbed by the proximal E-mail address: robert.unwin@ucl.ac.uk. MOS = ratio of the lethal dose in 1% of the population to the effective dose in 99% of the population = LD1/ED99 https://doi.org/10.1016/j.ejim.2021.09.008 Received 22 July 2021; Received in revised form 29 August 2021; Accepted 15 September 2021 0953-6205/© 2021 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved. Please cite this article as: Robert J. Unwin, European Journal of Internal Medicine, https://doi.org/10.1016/j.ejim.2021.09.008 Descargado para Ronald Eduardo Lozano Acosta (loacro@yahoo.com) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en noviembre 10, 2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados. R.J. Unwin European Journal of Internal Medicine xxx (xxxx) xxx tubule, this time by a megalin-dependent receptor-mediated endocytic process (see Fig. 1), and once inside the cell is slowly released to cause chronic damage [5]. A similar pattern of toxicity is seen with cadmium and lead [6,7]. The renal proximal tubule is particularly sensitive to all forms of toxic damage (see later), including ischaemic injury as a cause of acute kidney injury (AKI or what used to be called ATN, acute tubular ne­ crosis). This sensitivity to injury is probably for several reasons [8]. The proximal tubule is an important site of solute reabsorption, requiring a lot of energy (in the form of ATP), which is reflected in the large number of mitochondria these tubular cells contain, and their dependence for ATP generation on fatty acid oxidation, rather than glycolysis. The proximal tubule is also a major site for xenobiotic metabolism (cyto­ chrome P450 and conjugating enzymes, cf. liver) and excretion [9,10]. It also has the distinctive and promiscuous megalin-dependent transport mechanism already mentioned for the uptake of various filtered mole­ cules, including some drugs, especially antibiotics, and small (low mo­ lecular weight) proteins (LMWP) and peptides (Fig. 1). Thus, given the rich blood supply to the kidney for filtration, the proximal tubule, which is the first part of the nephron (Fig. 1), is exposed to high levels of most circulating factors, including drugs. In contrast to arsenic, another favoured poison is strychnine, a neurotoxin that causes kidney injury indirectly as a result of intense muscle spasm (opisthotonos), leading to muscle injury and rhabdo­ myolysis with release of myoglobin (myoglobinuria). Myoglobin is also reabsorbed by the megalin-dependent mechanism in the early proximal tubule (Fig. 1) and transferred to lysosomes from which the toxic ironheme portion of myoglobin is released into the cell causing acute tubular cell injury (AKI). In the tropics, snake venom is a common cause of poisoning and can have a similar effect, resulting in acute haemolysis (with haemoglobinuria), muscle necrosis (myoglobinuria) and severe coagulation defects (disseminated intravascular coagulation, DIC, and thrombotic microangiopathy, TMA, which can both also be infrequently drug-related), leading to AKI with a high mortality [11] (see Fig. 2). Furthermore, complications similar to those seen after snake venom poisoning can occur in patients with the not uncommon genetically X-linked deficiency of the enzyme glucose-6-phosphate dehydrogenase (G6PD) following exposure to various drugs, particularly sulphonamide derivatives; G6PD deficiency is found more commonly in Africa, the Middle East and Southeast Asia, and in up to 10% of African American males [12]. This is why alerting and reporting schemes are an important part of long-term monitoring and surveillance, and can help to avoid prescrip­ tion errors and ensure a regular review of medication [20,21]. Patients with CKD often take more than five different drugs singly or in combi­ nation, and may also take ‘over-the-counter’ (OTC) remedies that do not require a prescription. A recent CKD cohort study from Germany using an untargeted mass spectrometry-based approach to analyse for drug metabolites in urine, found that while this confirmed a generally good match with self-reported medications, and therefore treatment compli­ ance; it also revealed use of OTC analgesics, mainly NSAID, than was disclosed by patients, as well as combination drugs that patients may be unaware of, and drugs prescribed by other specialists such as neurolo­ gists or psychiatrists [22]. Efforts are ongoing to identify and develop biomarkers, particularly in urine, which can be used to detect kidney glomerular or tubular injury: the Predictive Safety Testing Consortium (PSTC) has qualified at least seven biomarkers for approved preclinical and clinical use in drug safety - see Table 1 [23]. While the markers listed may be of use in screening for AKI preclinically and in early phase clinical trials, less is known about any variation in levels in normal urine or in established CKD, other than for albumin and protein, and how these newer injury biomarkers may change with treatment. Six reminders or R’s for DIKD have been proposed by Awdishu and Mehta [24], which are quite useful in highlighting what should be considered when faced with a case of suspected nephrotoxicity. The first step is identify who is at Risk (1) in relation to the drug and patient characteristics; Recognise (2) a sudden deterioration in renal function (AKI/DIKD2); Respond (3) promptly by withdrawing the causative drug and provide whatever support is available and appropriate, including Renal (4) support; Rehabilitate (5) by careful follow up, including drug monitoring, if the drug implicated is still required, or avoid drug re-exposure; and support Research (6) through good documentation and participation in local and national adverse drug reporting schemes. Drug targets and mechanisms that commonly underlie nephrotoxi­ city are summarised in Table 2 with some examples. However, most are based on clinical observation and experience, rather than any detailed understanding of pathological mechanisms or ability to predict [25]. We have only a rudimentary knowledge of the pathobiology of renal toxicity for most drugs and this is still an important area for future research, but it depends critically on an alert physician recognising and reporting any clinically observed association between a drug and DIKD. 3. A little more background 4. Predisposing factors and sites of injury Adverse drug reactions can be broadly classified as either Type A and dose-dependent or Type B and idiosyncratic (‘allergic’). Drug-induced kidney damage (DIKD), a term beginning to be used to help stage or grade nephrotoxicity2, is estimated to cause around 20% of community and in-hospital AKI in adults and children; it is also more common in an intensive care setting with multiple drug (mainly high dose antibiotic) use, in the elderly, and in those with significant co-morbidities such as diabetes, cardiovascular disease (CVD) and chronic kidney disease (CKD) [13]. Of those drugs known to have adverse renal effects and may be taken deliberately to self-harm, or accidentally, over 50% of patients will experience serious kidney-related complications and are more likely to need renal replacement (dialysis support). The most commonly implicated drugs causing more acute toxicity are antibiotics, antivirals, chemotherapeutics, angiotensin-converting enzyme inhibitors (ACEi), NSAID, and unspecified herbal remedies, especially those containing aristolochic acid [14,15]. Kidney-related safety failures in drug development make up about 10% [16], and while in vitro methods for acute nephrotoxicity screening are improving with, for example, the use of organoids and ‘kid­ ney-on-chip’, and there is a better understanding of some of the key enzymes involved in drug metabolism (e.g., P450 family mentioned earlier) [17–19], it is still difficult to reliably predict chronic toxicity. The kidney receives approximately 20% of the cardiac output and so drug delivery and exposure are high, and it is important to consider those factors that can determine the potential for drug toxicity: the drug itself, for example, if renally excreted, its urine solubility (precipitation – ampicillin, indinavir), possible direct actions on the kidney, interactions with other drugs, including effects they may have on excretion, and patient characteristics, including age, sex and race, which can all affect drug handling and metabolism, as well as co-morbidities such as CKD or liver disease [19]. Pharmacogenetics: genetic polymorphisms that sub­ tly alter drug transport or metabolism are becoming increasingly important and are beginning to be screened for; an early example was the discovery of ‘fast’ or ‘slow’ acetylator status for the antihypertensive drug hydralazine, which can affect blood pressure control and the risk of a lupus-like adverse drug reaction [26,27]. Both drug-related glomerular and tubular cell toxicity can occur. Direct glomerular podocyte injury leads to proteinuria (minimal change 2 KDIGO definition of Acute Kidney Injury, AKI, is a 50% rise in serum creatinine in <7 days, often within 24-48 h; AKD, Acute Kidney Disease is duration of 7-90 days and established CKD > 90 days. DIKD can be defined similarly as acute (1-7 days), sub-acute (8-90 days) and chronic (>90 days). 2 Descargado para Ronald Eduardo Lozano Acosta (loacro@yahoo.com) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en noviembre 10, 2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados. European Journal of Internal Medicine xxx (xxxx) xxx R.J. Unwin Fig. 1.. Diagram of the proximal tubule showing individual cells with the transport mechanisms discussed (see text for details). The main solute transporters for bicarbonate (sodium-hydrogen ion exchanger NHE-3), glucose (SGLT2), phosphate (NPT2a and 2c) and amino acids (aa) are shown, together with the megalin- (with cubilin) dependent uptake mechanism for low molecular weight proteins (LMWP) and peptides, which cycles its ligands through endosomes to lysosomes. The sodium pump that energises sodium-coupled transport is shown at the basolateral membrane of the cell. The small figure inset (top right) for orientation is a schematic of a nephron: G, glomerulus; PT, proximal tubule; LOH, loop of Henle; DT, distal tubule; CD, collecting duct. Fig. 2.. Schematic of the mechanisms involved in snake venom-mediated kidney injury. GFR, glomerular filtration rate; RBF, renal blood flow; AKI, acute kidney injury. Many of these pathological mechanisms are also seen with some drug-related causes of nephrotoxicity. Table 1. See text for details. Examples of approved urinary biomarkers for detecting kidney glomerular and tubular damage. KIM-1, kidney injury marker 1; CLU, clusterin; TFF3, trefoil factor 3; SCr, serum creatinine; BUN, blood urea nitrogen. Apart from albumin and total protein, the most widely used markers currently are KIM-1 and cystatin-C. While β2-microglobulin as a low molecular weight protein (LMWP) can be used as an early marker of proximal tubular injury, because its reabsorption is megalin-dependent; circulating levels can be elevated in a variety of situations, including CKD. The LMWPs retinol binding protein (RBP) and/or α1-microglubulin may be more useful for this purpose in clinical practice. Other biomarkers of tubular injury include lipocalin (NGAL) and osteopontin, but they have limited specificity. Biomarker (urine) Preclinical Clinical Complements SCr/BUN Superior to SCr/BUN KIM-1 Albumin CLU TFF3 Total protein Cystatin C β2-microglobulin Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y Y N Y Y Y 3 Descargado para Ronald Eduardo Lozano Acosta (loacro@yahoo.com) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en noviembre 10, 2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados. R.J. Unwin European Journal of Internal Medicine xxx (xxxx) xxx and has significant energy demands with a heavy reliance on mito­ chondrial ATP generation. Tenofovir is taken up by proximal tubular cells via the organic anion transporter, OAT1, expressed on the basolateral cell membrane, and is eliminated by secretion via the multidrug resistant protein, MRP4, on the apical membrane. Disturbed secretion by, for example, a drug-drug interaction (DDI) inhibiting or competing with MRP (e.g., NSAIDs), can lead to the intracellular accumulation of tenofovir causing mitochon­ drial damage. Probenecid is a well-described inhibitor of OAT1 and has been shown to reduce the toxicity of the related drug cidofovir [33], but it can be difficult to predict the outcome of such drug interactions, since probenecid is not selective and can also inhibit MRP (Fig. 1). A related example is cisplatin nephrotoxicity. Cisplatin is also taken up by prox­ imal tubular cells, but in this instance via the organic cation transporter, OCT2 (Fig. 1) and it is secreted via the multidrug and toxin extrusion protein, MATE1. As with probenecid, in this case cimetidine can be used to block OCT2 and cisplatin uptake, but it can also inhibit MATE1 responsible for cisplatin extrusion. However, in practice cimetidine’s overall effect is to limit cisplatin-related nephrotoxicity [34]. Finally, an important feature of this type of nephrotoxicity is that the tubular injury markers in urine described earlier (see Table 1), including LMWP like RBP and other indices of proximal tubular function (e.g., glycosuria with normoglycaemia), can be used to detect proximal tubular damage. These urinary biomarkers should be used more widely in conjunction with changes in serum creatinine or urea when a suspicion of drug nephro­ toxicity is raised. However, efforts to identify nephron site-specific (Fig. 1) injury biomarkers in urine are still in their infancy, although certain urinary miRNAs show some promise [35]. Returning briefly to tenofovir, there are currently two forms avail­ able, TDF (tenofovir disoproxil fumarate) and TAV (tenofovir alafena­ mide), the former being the first shown to be nephrotoxic, as described above. While TAF still has the potential to cause renal toxicity, since it is also excreted by the kidney, its different pharmacokinetic (PK) proper­ ties are such that it accumulates more rapidly in peripheral blood monocytes (the target cells) and its peak plasma levels tend to be lower than with TDF, and so it is less of a renal burden for excretion; these PK differences make it less nephrotoxic compared with TDF [36]. It is worth mentioning at this point that creatinine excretion is also dependent on MATE1, OCT2 and (preferentially) OAT2; thus, a rise in serum creatinine may indicate a DDI, rather than renal toxicity, for example, with trimethoprim, cimetidine (see above) or pyrimethamine. These drugs can also reduce metformin excretion, which is MATE1- Table 2. Sites of nephrotoxicity, mechanism, and some corresponding examples. Pathophysiology and Target Corresponding Examples Glomerular (MCD, FSGS, MN, TMA, Inflammatory) Tubular Crystal (Nephrolithiasis) NSAID, IFN, VEGF, CNI, Hydralazine Acute Interstitial Nephritis (AIN/AKI) Chronic Interstitial Nephritis (CKD) Rhabdomyolysis (AKI) Hyperosmolarity Antibiotics, Antiretrovirals Antibiotics, Antiretrovirals, CA inhibitors Antibiotics, NSAIDS, PPIs, Any drug Analgesics(?), Herbal (aristolochic acid) Statins, Drugs of abuse (cocaine, MDA, alcohol) IV dextrans, IgG disease and focal segmental glomerulosclerosis, both causes of the nephrotic syndrome), endothelial injury, microthrombosis (TMA mentioned earlier), and mesangial cell injury to nodular sclerosis. Glomerular immune-mediated injury can also occur with some drugs that induce immune complex formation (causing a lupus-like nephritis see hydralazine above - or membranous nephropathy with proteinuria) and ANCA-related small vessel vasculitis, again with hydralazine [28]. Tubular injury usually results from mitochondrial or lysosomal toxicity (see Fig. 2). Because these sites of injury are relatively localised, prog­ ress has been made in creating ‘organs on a chip’, perfused tubules and isolated glomeruli or ‘organoids’ containing all the cell types referred to above; these are becoming a valuable means of screening new drugs for potential nephrotoxicity. As an example: an interesting case illustrated in Fig. 2 is the toxicity of the antiretroviral drug tenofovir [29]. This only became apparent when patients treated with this drug became hypophosphataemic and complained of musculoskeletal aches and pains, prompting in some cases bone scans that showed signs of osteomalacia [30] and features seen typically in patients with oncogenic osteomalacia [31] (Fig. 3). Many patients were also found to have proteinuria on urine dipstick testing, which turned out to be mainly albumin and LMWP such as retinol binding protein (RBP). These findings pointed to an abnormality of kidney proximal tubular reabsorptive capacity, and disturbed mito­ chondrial function was soon identified as the underlying cause with typical features of a renal Fanconi syndrome [32]. On stopping the drug these abnormalities gradually reversed. As mentioned earlier, the proximal tubule is something of a reabsorbing and secreting ‘workhorse’ Fig. 3.. (a) 99m-Technectium bone appearances in a patient on tenofovir showing an increased bone to soft tissue ratio and ‘hot spot’ pattern typical of osteomalacia and also seen in patients with oncogenic osteomalacia. (b) Renal biopsy electron micrographs (EM) from a patient on tenofovir showing abnormally enlarged mitochondria that have lost their normal crista pattern. 4 Descargado para Ronald Eduardo Lozano Acosta (loacro@yahoo.com) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en noviembre 10, 2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados. R.J. Unwin European Journal of Internal Medicine xxx (xxxx) xxx dependent, and is an important DDI to look out for in diabetic patients [33]. Remember also that initial increases in serum creatinine are often seen with drugs that have short-term haemodynamic effects that can reduce GFR, including RAAS and SGLT2 inhibitors, which do not herald nephrotoxicity, although still need monitoring, and that changes in muscle mass can be an additional confounder. and macroscopic haematuria occasional. Proteinuria is unusual and rarely nephrotic range, but when this occurs it is usually due to NSAIDs. AIN is increasingly associated with other drugs such as proton pump inhibitors, 5-aminosalicylates and NSAIDs, and may not be evident early on; however, almost any drug can be implicated and AIN should always be considered when confronted with a patient with unexplained loss of renal function, even if not acute. AIN is not dependent on the drug dose and is an idiosyncratic (Type B) reaction that still has an unclear im­ mune basis and it may take time to develop. There is no known bio­ logical predilection, other than polypharmacy. The topic of AIN has been reviewed comprehensively by Perazella and co-workers, who have written extensively on the subject in recent years [47]. As to treatment, this remains uncertain, although the current consensus is a trial of high-dose steroids started early and for up to one month [48]. More recently, immunotherapy with immune checkpoint inhibitors, a class of drug that has revolutionised cancer therapy, is increasingly being re­ ported as a cause of drug-related AKI, which seems to be due to immune-related AIN and can be responsive to a course of steroids [49]. However, the need to stop the inhibitor may be problematic for the patient, as may the use of high-dose steroids, and confirmation of AIN with a renal biopsy is generally recommended [50]. 5. Analgesic nephropathy: does it exist? This has been a hotly debated topic for many years, the first reports coming from Switzerland in the 1950s [37] and a seeming epidemic of cases in reports from Australia in the 1960s of phenacetin-containing analgesics associated with renal papillary necrosis, particularly in women, which initially was taken to be ‘cause-and-effect’ [38]. This led to the withdrawal of phenacetin, which did seem to be associated with a decline in papillary necrosis cases [39], but there was always some concern that this drug was often taken in combination with other compounds, including opiates and caffeine [40], and it wasn’t always clear what else might have been taken by affected patients, usually for recurrent or chronic headache. Papillary necrosis with calcification can be diagnosed on a CT scan, which has been advocated as a screening examination with a claimed high sensitivity and specificity for the diagnosis of analgesic nephropathy [41]; although there are other cau­ ses of papillary necrosis that may be coincidentally associated with increased analgesic use, such as diabetic kidney disease, recurrent uri­ nary tract infections (pyelonephritis), urinary tract obstruction, and sickle cell disease. Paracetamol was also invoked as a cause of analgesic nephropathy early on [42] (prompting widespread disagreement at the time and since), probably because it was sometimes co-prescribed with phenac­ etin and that a major product of phenacetin metabolism is paracetamol. But it is now thought that the other metabolite of phenacetin, p-phe­ netidine, is probably the toxic culprit, being an extremely potent in­ hibitor of prostaglandin (PGE2) synthesis, unlike paracetamol or phenacetin itself [43]. Moreover, subsequent case series have not shown that paracetamol is a cause of analgesic nephropathy [44,45]. It was later claimed that chronic NSAID use is another potential cause of analgesic nephropathy and chronic kidney disease (CKD). Although there are certainly situations in which this might occur more acutely (AKI), particularly in combination with an ACEi or angiotensin receptor blocker (ARB), or when given to an elderly and dehydrated patient, it has not been clearly established that NSAIDs cause analgesic nephropathy. However, they are a recognised cause of acute interstitial nephritis (AIN) (see later), which can lead to residual renal damage and CKD. However, a recent US cohort study in 3000 older (mean age 74) patients with a record of GFR values for ten years could find no evidence of an association between self-reported NSAID use and loss of renal function, including measurement of the renal injury biomarkers IL-18 and KIM-1: mean baseline eGFR was 70 ml/min and did not change significantly [46]. That said, some caution is still appropriate in those with pre-existing renal impairment and occasional monitoring of renal function, especially soon after initiation of treatment, is recommended. However, bear in mind that NSAIDs can be an important means of controlling pain and discomfort in those with chronic arthritis and should not be withheld without good reason. 7. What next? As mentioned already, awareness is the key, particularly with the ever-increasing number of new drugs being introduced, including the newer modalities (monoclonal antibodies, antisense oligonucleotides, RNA- and DNA- based therapies, etc) and the increasing tendency for patients to be taking more than one drug or drug combination. Efforts to improve early drug screening for potential nephrotoxicity are proceed­ ing apace, both with novel biomarkers of kidney injury that can also indicate the likely nephron site of toxicity, more sophisticated and in­ tegrated cell-based kidney models in vitro [8,18], as well as electronic alerting systems being introduced and trialled to detect early cases of AKI that are often drug-related [20]. Finally, the clinical challenge is knowing when a drug is a potential cause of nephrotoxicity and in always being cognizant of that possibility. Acknowledgments The author is currently employed by AstraZeneca Bio­ pharmaceuticals R&D, Cardiovascular, Renal and Metabolism (CVRM), Cambridge UK. References [1] Ali H, Khan E, Ilahi I. Environmental chemistry and ecotoxicology of hazardous heavy metals: environmental persistence, toxicity, and bioaccumulation. J ChemNy 2019;2019:1–14. https://doi.org/10.1155/2019/6730305. [2] Wei H, Hu Q, Wu J, Yao C, Xu L, Xing F, et al. Molecular mechanism of the increased tissue uptake of trivalent inorganic arsenic in mice with type 1 diabetes mellitus. Biochem Bioph Res Co 2018;504:393–9. https://doi.org/10.1016/j. bbrc.2018.06.029. [3] Robles-Osorio ML, Sabath-Silva E, Sabath E. Arsenic-mediated nephrotoxicity. Renal Failure 2015;37:542–7. https://doi.org/10.3109/0886022x.2015.1013419. [4] Lindskog C, Asplund A, Catrina A, Nielsen S, Rützler M. A systematic characterization of Aquaporin-9 expression in human normal and pathological tissues. J Histochem Cytochem 2016;64:287–300. https://doi.org/10.1369/ 0022155416641028. [5] Lentini P, Zanoli L, Granata A, Signorelli SS, Castellino P, Dellaquila R. Kidney and heavy metals - the role of environmental exposure. Mol Med Rep 2017;15:3413–9. https://doi.org/10.3892/mmr.2017.6389. [6] Johri N, Jacquillet G, Unwin RJ. Heavy metal poisoning: the effects of cadmium on the kidney. Biometals 2010;23:783–92. https://doi.org/10.1007/s10534-0109328-y. [7] Menon S, Kirkendall ES, Nguyen H, Goldstein SL. Acute kidney injury associated with high nephrotoxic medication exposure leads to chronic kidney disease after 6 months. J Pediatr 2014. https://doi.org/10.1016/j.jpeds.2014.04.058. [8] Hall AM, Trepiccione F, Unwin RJ. Drug toxicity in the proximal tubule: new models, methods and mechanisms. Pediatr Nephrol 2021:1–10. https://doi.org/ 10.1007/s00467-021-05121-9. 6. Acute interstitial nephritis (AIN) and AKI This is characterised by a sudden decline in renal function (AKI) associated with an interstitial inflammatory infiltrate seen in biopsy tissue from the kidney. While an inflammatory infiltrate is detected in around 1-3% of all renal biopsies, it is found in up to 30% of patients with AKI, but is not consistently diagnostic of AIN. Approximately 75% of cases of AIN are drug-related with antibiotics causing a third or more of cases and often associated with a skin rash, and blood and urine eosinophilia. Fever and arthralgia may also occur, oliguria is common, 5 Descargado para Ronald Eduardo Lozano Acosta (loacro@yahoo.com) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en noviembre 10, 2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados. R.J. Unwin European Journal of Internal Medicine xxx (xxxx) xxx [30] Woodward C, Hall A, Williams I, Madge S, Copas A, Nair D, et al. Tenofovirassociated renal and bone toxicity. Hiv Med 2009;10:482–7. https://doi.org/ 10.1111/j.1468-1293.2009.00716.x. [31] Sood A, Agarwal K, Shukla J, Goel R, Dhir V, Bhattacharya A, et al. Bone scintigraphic patterns in patients of tumor induced osteomalacia. Indian J Nucl Medicine 2013;28:173–5. https://doi.org/10.4103/0972-3919.119541. [32] Hall AM, Bass P, Unwin RJ. Drug-induced renal Fanconi syndrome. Qjm Int J Medicine 2014;107:261–9. https://doi.org/10.1093/qjmed/hct258. [33] Lepist E, Ray AS. Renal transporter-mediated drug-drug interactions: are they clinically relevant? J Clin Pharmacol 2016;56:S73–81. https://doi.org/10.1002/ jcph.735. [34] Katsuda H, Yamashita M, Katsura H, Yu J, Waki Y, Nagata N, et al. Protecting cisplatin-induced nephrotoxicity with cimetidine does not affect antitumor activity. Biol Pharm Bull 2010;33:1867–71. https://doi.org/10.1248/bpb.33.1867. [35] Chorley BN, Ellinger-Ziegelbauer H, Tackett M, Simutis FJ, Harrill AH, McDuffie J, et al. Urinary miRNA biomarkers of drug-induced kidney injury and their site specificity within the nephron. Toxicol Sci 2020;180:kfaa181. https://doi.org/ 10.1093/toxsci/kfaa181. [36] Ray AS, Fordyce MW, Hitchcock MJM. Tenofovir alafenamide: a novel prodrug of tenofovir for the treatment of human immunodeficiency virus. Antivir Res 2016; 125:63–70. https://doi.org/10.1016/j.antiviral.2015.11.009. [37] Dubach UC, Rosner B, Levy PS, Baumeler H-R, Müller A, Peyer A, et al. Epidemiological study in Switzerland. Kidney Int 1978;13:41–9. https://doi.org/ 10.1038/ki.1978.6. [38] Nanra RS, Stuart-Taylor J, Leon de AH, White KH. Analgesic nephropathy: etiology, clinical syndrome, and clinicopathologic correlations in Australia. Kidney Int 1978;13:79–92. https://doi.org/10.1038/ki.1978.11. [39] Waddington F, Naunton M, Thomas J. Paracetamol and analgesic nephropathy: are you kidneying me? Int Medical Case Reports J 2014;Volume 8:1–5. https://doi. org/10.2147/imcrj.s71471. [40] Elseviers MM, Broe MED. Analgesic nephropathy. Drug Safety 1999;20:15–24. https://doi.org/10.2165/00002018-199920010-00003. [41] Elseviers MM, Schepper AD, Corthouts R, Bosmans J-L, Cosyn L, Lins RL, et al. High diagnostic performance of CT scan for analgesic nephropathy in patients with incipient to severe renal failure. Kidney Int 1995;48:1316–23. https://doi.org/ 10.1038/ki.1995.416. [42] Koutsaimanis KG, Wardener de HE. Phenacetin nephropathy, with particular reference to the effect of surgery. Brit Med J 1970;4:131–4. https://doi.org/ 10.1136/bmj.4.5728.131. [43] Kankuri E, Solatunturi E, Vapaatalo H. Effects of phenacetin and its metabolite pphenetidine on COX-1 and COX-2 activities and expression in vitro. Thromb Res 2003;110:299–303. https://doi.org/10.1016/s0049-3848(03)00416-x. [44] Mihatsch MJ, Khanlari B, Brunner FP. Obituary to analgesic nephropathy—an autopsy study. Nephrol Dial Transpl 2006;21:3139–45. https://doi.org/10.1093/ ndt/gfl390. [45] Michielsen P, Heinemann L, Mihatsch M, Schnülle P, Graf H, Koch K-M. Nonphenacetin analgesics and analgesic nephropathy clinical assessment of high users from a case-control study. Nephrol Dial Transpl 2008;24:1253–9. https://doi.org/ 10.1093/ndt/gfn643. [46] Amatruda JG, Katz R, Peralta CA, Estrella MM, Sarathy H, Fried LF, et al. Association of non-steroidal anti-inflammatory drugs with kidney health in ambulatory older adults. J Am Geriatr Soc 2021;69:726–34. https://doi.org/ 10.1111/jgs.16961. [47] Moledina DG, Perazella MA. The challenges of acute interstitial nephritis: time to standardize. Kidney360 2021;2:1049–53. https://doi.org/10.34067/ kid.0001742021. [48] Fernandez-Juarez G, Perez JV, Caravaca-Fontán F, Quintana L, Shabaka A, Rodriguez E, et al. Duration of treatment with corticosteroids and recovery of kidney function in acute interstitial nephritis. Clin J Am Soc Nephro 2018;13: 1851–8. https://doi.org/10.2215/cjn.01390118. [49] Cortazar FB, Kibbelaar ZA, Glezerman IG, Abudayyeh A, Mamlouk O, Motwani SS, et al. Clinical features and outcomes of immune checkpoint inhibitor-associated AKI: a multicenter study. J Am Soc Nephrol 2020;31:435–46. https://doi.org/ 10.1681/asn.2019070676. [50] Perazella MA, Shirali AC. Immune checkpoint inhibitor nephrotoxicity: what do we know and what should we do? Kidney Int 2020;97:62–74. https://doi.org/ 10.1016/j.kint.2019.07.022. [9] Knights KM, Rowland A, Miners JO. Renal drug metabolism in humans: the potential for drug–endobiotic interactions involving cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT). Brit J Clin Pharmaco 2013;76:587–602. https://doi.org/10.1111/bcp.12086. [10] Bajaj P, Chowdhury SK, Yucha R, Kelly EJ, Xiao G. Emerging kidney models to investigate metabolism, transport and toxicity of drugs and xenobiotics. Drug Metab Dispos 2018;46:dmd.118.082958. https://doi.org/10.1124/ dmd.118.082958. [11] Chugh KS. Snake-bite-induced acute renal failure in India. Kidney Int 2010;35: 891–907. https://doi.org/10.1038/ki.1989.70. [12] Bubp J, Jen M, Matsuszewski K. Caring for glucose-6-phosphate dehydrogenase (G6PD)–deficient patients: implications for pharmacy. P&T 2015;40:572–4. [13] Petejova N, Martinek A, Zadrazil J, Teplan V. Acute toxic kidney injury. Renal Failure 2019;41:576–94. https://doi.org/10.1080/0886022x.2019.1628780. [14] Mody H, Ramakrishnan V, Chaar M, Lezeau J, Rump A, Taha K, et al. A review on drug-induced nephrotoxicity: pathophysiological mechanisms, drug classes, clinical management, and recent advances in mathematical modeling and simulation approaches. Clin Pharm Drug Dev 2020;9:896–909. https://doi.org/ 10.1002/cpdd.879. [15] Yang B, Xie Y, Guo M, Rosner MH, Yang H, Ronco C. Nephrotoxicity and Chinese herbal medicine. Clin J Am Soc Nephro 2018;13:1605–11. https://doi.org/ 10.2215/cjn.11571017. [16] Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G, et al. Lessons learned from the fate of AstraZeneca’s drug pipeline: a five-dimensional framework, 13. Nature Publishing Group; 2014. p. 419–31. https://doi.org/ 10.1038/nrd4309. [17] Kim S, LesherPerez SC, Kim B choul C, Yamanishi C, Labuz JM, Leung B, et al. Pharmacokinetic profile that reduces nephrotoxicity of gentamicin in a perfused kidney-on-a-chip. Biofabrication 2016;8:1–10. https://doi.org/10.1088/17585090/8/1/015021. [18] Wiraja C, Mori Y, Ichimura T, Hwang J, Xu C, Bonventre JV. Nephrotoxicity assessment with human kidney tubuloids using spherical nucleic acid-based mRNA nanoflares. Nano Lett 2021. https://doi.org/10.1021/acs.nanolett.1c01840. [19] Perazella MA. Pharmacology behind common drug nephrotoxicities. Clin J Am Soc Nephrol 2018:CJN.00150118. https://doi.org/10.2215/cjn.00150118. [20] Martin M, Wilson FP. Utility of electronic medical record alerts to prevent drug nephrotoxicity. Clin J Am Soc Nephro 2019;14:115–23. https://doi.org/10.2215/ cjn.13841217. [21] Ong SW, Jassal SV, Porter EC, Min KK, Uddin A, Cafazzo JA, et al. Digital applications targeting medication safety in ambulatory high-risk CKD patients: randomized controlled clinical trial. Clin J Am Soc Nephro 2021;16:532–42. https://doi.org/10.2215/cjn.15020920. [22] Kotsis F, Schultheiss U, Wuttke M, Schlosser P, Mielke J, Becker M, et al. Selfreported medication use and urinary drug metabolites in the German chronic kidney disease (GCKD) study. J Am Soc Nephrol 2021:ASN.2021010063. https:// doi.org/10.1681/asn.2021010063. [23] Dieterle F, Sistare F, Goodsaid F, Papaluca M, Ozer JS, Webb CP, et al. Renal biomarker qualification submission: a dialog between the FDA-EMEA and predictive safety testing consortium. Nat Biotechnol 2010;28:455–62. https://doi. org/10.1038/nbt.1625. [24] Awdishu L, Mehta RL. The 6R’s of drug induced nephrotoxicity. Bmc Nephrol 2017;18:124. https://doi.org/10.1186/s12882-017-0536-3. [25] Sales GTM, Foresto RD. Drug-induced nephrotoxicity. Revista Da Assoc Médica Brasileira 2020;66:s82–90. https://doi.org/10.1590/1806-9282.66.s1.82. [26] Ramsay LE, Silas JH, Ollerenshaw JD, Tucker GT, Phillips FC, Freestone S. Should the acetylator phenotype be determined when prescribing hydralazine for hypertension? Eur J Clin Pharmacol 1984;26:39–42. https://doi.org/10.1007/ bf00546706. [27] Collins KS, Raviele ALJ, Elchynski AL, Woodcock AM, Zhao Y, Cooper-DeHoff RM, et al. Genotype-guided hydralazine therapy. Am J Nephrol 2020;51:764–76. https://doi.org/10.1159/000510433. [28] Santoriello D, Bomback AS, Kudose S, Batal I, Stokes MB, Canetta P, et al. Antineutrophil cytoplasmic antibody associated glomerulonephritis complicating treatment with hydralazine. Kidney Int 2021;100:440–6. https://doi.org/10.1016/ j.kint.2021.03.029. [29] Hall AM, Edwards SG, Lapsley M, Connolly JO, Chetty K, O’Farrell S, et al. Subclinical tubular injury in HIV-infected individuals on antiretroviral therapy: a cross-sectional analysis. Am J Kidney Dis 2009;54:1034–42. https://doi.org/ 10.1053/j.ajkd.2009.07.012. 6 Descargado para Ronald Eduardo Lozano Acosta (loacro@yahoo.com) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en noviembre 10, 2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.