You are on page 1of 13

www.kidney-international.

org meeting report

Summary of the International Conference


on Onco-Nephrology: an emerging field
in medicine
Anna Capasso1, Ariella Benigni2, Umberto Capitanio3, Farhad R. Danesh4, Vincenzo Di Marzo5,6,
Loreto Gesualdo7, Giuseppe Grandaliano8, Edgar A. Jaimes9, Jolanta Malyszko10, Mark A. Perazella11,
Qi Qian12, Pierre Ronco13,14, Mitchell H. Rosner15, Francesco Trepiccione16,17, Davide Viggiano17,
Carmine Zoccali18 and Giovambattista Capasso16,17; for the International Conference on
Onco-Nephrology Participants19
1
Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas, Austin, Texas, USA; 2Istituto di
Ricerche Farmacologiche Mario Negri IRCSS, Bergamo, Italy; 3Unit of Urology, Division of Experimental Oncology, Urological Research
Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy; 4Section of Nephrology, The University of Texas MD Anderson Cancer Center,
Houston, Texas, USA; 5Consiglio Nazionale delle Ricerche - Istituto di Chimica Biomolecolare, Pozzuoli, Italy; 6Canada Research Excellence
Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Quebec City, Quebec, Canada; 7Department of
Nephrology, Aldo Moro University of Bari, Bari, Italy; 8Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy;
9
Renal Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; 10Department of Nephrology, Dialysis and Internal
Medicine, Warsaw Medical University, Warsaw, Poland; 11Department of Medicine, Yale University School of Medicine, New Haven,
Connecticut, USA; 12Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA; 13Inserm UMR-S1155, Sorbonne Université,
Paris, France; 14Assistance Publique-Hôpitaux de Paris, Department of Nephrology and Dialysis, Hôpital Tenon, Paris, France; 15University
of Virginia Health System, Charlottesville, Virginia, USA; 16Department of Translational Medical Sciences, University Luigi Vanvitelli,
Naples, Italy; 17BioGeM Institute, Ariano Irpino, Italy; and 18CNR-IFC, c/o Ospedali Riuniti, Reggio Calabria, Italy

C
Onco-nephrology is an emerging field in medicine. Patients ancer and kidney disease are both associated with
with cancer may suffer from kidney diseases because of the significant morbidity and mortality.1 Over the past
cancer itself and cancer-related therapy. It is critical for decade, nephrologists, oncologists, and other clinicians
nephrologists to be knowledgeable of cancer biology and recognized kidney disease and electrolyte/acid-base disturbances
therapy in order to be fully integrated in the as complications of a malignant tumor. Moreover, chronic
multidisciplinary team and optimally manage patients with kidney disease (CKD) is associated with an increased cancer
cancer and kidney diseases. In a recent international risk, making the relationship bidirectional. The importance of
meeting, the key issues in this challenging clinical interface this cancer–kidney disease connection has prompted the crea-
were addressed, including many unresolved basic science tion of the subfield “onco-nephrology” (Figure 1).2
questions, such as the high tumor incidence in kidney Both acute kidney injury (AKI) and CKD are prevalent in
transplant recipients. To this end, 70 highly qualified patients with cancer.3–5 Incidence and severity of AKI varies
faculty members were gathered from all over the world to depending on type/stage of cancer, treatment regimen, and
discuss these issues in 8 plenary sessions, including 5 underlying comorbidities.4–6 Patients with incident cancer
keynote lectures. In addition, 48 young nephrologists and have 1- and 5-year AKI risk of 17.5% and 27%, respectively.7
oncologists were invited to present their original Critically ill patients with cancer are at an even higher risk,
observations that were highlighted in 2 large poster with w13% to 54% of patients developing AKI and 8% to
sessions. 60% requiring dialysis.8,9 As observed in the Renal
Kidney International (2019) 96, 555–567; https://doi.org/10.1016/ Insufficiency and Cancer Medications (IRMA) Study 1 and
j.kint.2019.04.043 2, patients with cancer had a stage 3 CKD prevalence of
KEYWORDS: acute kidney injury; cancer; chemotherapy; chronic kidney w12%.10,11 In other cancer cohorts, stage 3 CKD was
disease; nephrotoxicity; renal cell carcinoma observed in w13% to 30% of patients.12,13
Copyright ª 2019, International Society of Nephrology. Published by Major risks of AKI include cancer-related metabolic
Elsevier Inc. All rights reserved.
disturbances, tissue deposition of paraproteins, treatment
with nephrotoxic anticancer drugs, and hematopoietic stem
Correspondence: Giovambattista Capasso, Department of Translational cell transplantation.14 Total and partial nephrectomy for
Medical Sciences, University Luigi Vanvitelli, Padiglione 17 Policlinico Nuovo, kidney cancer also increases the risk of acute kidney disease/
Via Pansini 5, 80131 Naples, Italy. E-mail: gb.capasso@unicampania.it CKD.15
19
See Appendix for the International Conference on Onco-Nephrology Morbidity/mortality is higher in patients with cancer and
Participants. kidney diseases,6,10 with the highest mortality in patients with
Received 30 January 2019; revised 2 April 2019; accepted 5 April 2019; dialysis-requiring AKI.4 Some patients with cancer and CKD
published online 31 May 2019 may also have an increased risk of death.12,13,16 Mortality risk

Kidney International (2019) 96, 555–567 555


meeting report A Capasso et al.: Onco-nephrology: an emerging field in medicine

Onco-Nephrology mice17 with the advantage of recapitulating intra- and intertumor


heterogeneity.18 However, the absence of an immune system in
these mice limits their use to study immune-based treat-
ments.19 With immunotherapy likely being a permanent
Electrolyte -Acid/Base component of the anticancer arsenal, better in vitro and
Disorders in vivo preclinical models are needed.20,21 Ex vivo spheroid
models have been generated to screen immunotherapy
combinations, but such approach is also limited by lacking
the ability to study the tumor–immune system interactions.22
Likewise, the use of syngenic murine-derived cancer models
An cancer Drugs
in immune-competent mice is regrettable on the basis of
Malignancy-related Kidney Injury current knowledge of the molecular heterogeneity of human
AKI CKD
Radia on/Nephrectomy malignancies, limited available models, and differences be-
Uremic Milleu (CKD, ESRD) tween mouse and human immunology.23–25
To gain a better biological understanding of the human
immune system, “hematopoietic humanized mice” have been
developed. These models provide an opportunity to study the
human biological process that would not otherwise be
CANCER possible.
Currently several humanized mouse models are being used
to study human diseases.26 They can be generated by trans-
Figure 1 | The kidney disease–cancer connection. AKI, acute plantation of (i) human peripheral blood mononuclear cells,
kidney injury; CKD, chronic kidney disease; ESRD, end-stage renal
disorder. (ii) tumor infiltrating lymphocytes, (iii) CD34þ human he-
matopoietic stem, or (iv) transplantation of human fetal liver
and thymus under the kidney capsule and injection of
is highest for hematologic malignancies, gynecologic cancers, autologous fetal liver human stem cells.27–29
and renal carcinomas in the setting of CKD.12,13 Because of their immunosuppressed environment, these
Kidney disease increases the risk of systemic chemotherapy humanized models have shown to accept allogeneic tumors,
toxicity, jeopardizes continued cancer therapy, and limits pa- providing a novel preclinical platform to study innovative
tient participation in clinical trials. Temporary or permanent therapeutic combination in cancer.30
cessation of effective chemotherapeutic regimens allows un- These models allow a comprehensive analysis of the hu-
hindered tumor growth, whereas underdosing of potentially man immune system, granting access to lymph nodes, spleen,
curative regimens or use of suboptimal alternatives reduces and tumor tissues and providing more information about
treatment efficacy.2,5 Altered drug pharmacokinetics risk sys- immune response. Immunotherapies have started a new era
temic toxicity, whereas uremia impairs immune surveillance, in cancer therapy, but the impressive immune-mediated re-
allowing cancers to grow/metastasize. It is also possible that sponses are still limited to a minority of patients. Despite the
anticancer drug nephrotoxicity leads to progression of AKI and/ encouraging results obtained in the clinical setting and the
or CKD, contributing to all-cause mortality unrelated to cancer. initial evidence of the importance of tumor mutation load
It is critical that clinicians and researchers from multiple and genetic alterations, we still lack the full understanding of
specialties develop familiarity with onco-nephrology as the mechanisms responsible for tumor immune rejection. Hu-
number of patients surviving cancer and kidney disease in- manized mouse models can improve our knowledge and
creases. To this end, the meeting was designed. The summary facilitated testing of novel therapeutic combinations.31
of the plenary sessions is reported below.
The full conference program and the video recordings of How to measure kidney function in patients with cancer
all presentations and discussions can be found at the following Measuring glomerular filtration rate (GFR) in patients with
website: https://www.en.fondazione-menarini.it/Home/News/ neoplasia is fundamental for profiling both the risk and the
Onco-Nephrology-an-Emerging-Field-in-Medicine/Presentation. appropriate dose of chemotherapeutic agents. Ideally GFR
should be measured using criterion standard, that is, by
Advanced technologies chromium-51-ethylenediaminetetraacetic acid (51Cr-EDTA)
Researchers worldwide base their experiments using cancer cells or technetium-99m–diethylenetriaminepentaacetic acid
in vitro, but to acquire a comprehensive understanding of the ( mTc-DTPA) or by inulin or iothalamate clearance32 (see
99

complexity of cancer biological processes, in vivo experiments are Canadian Cancer Association: Glomerular Filtration Rate
required. In cancer translational research and in the development study, http://www.cancer.ca). However, these methods are
of new therapeutics, many important advances have been time-consuming and costly. For this reason, in the vast ma-
accomplished by the use of patient-derived xenografts, that is, jority of patients with cancer, GFR is estimated by creatinine-
patient-derived tumors transplanted in immunocompromised based formulas. Several studies in patients with cancer

556 Kidney International (2019) 96, 555–567


A Capasso et al.: Onco-nephrology: an emerging field in medicine meeting report

examined the reliability of current creatinine-based methods, posttransplant neoplasia is well known to be linked to the
namely, the Cockcroft-Gault equation, the 4-variable Modi- action of specific viruses.47 In addition, immunosuppressive
fication of Diet in Renal Disease equation based on either drugs may have direct effects on the development of partic-
uncalibrated creatinine or calibrated creatinine, and the ular types of cancer, as demonstrated for azathioprine.48
Chronic Kidney Disease Epidemiology Collaboration (CKD- Finally, immunosuppression can facilitate the process of tu-
EPI) equation. Nevertheless, these studies focused on a mor immune escape. The reduced ability of the immune
specific type of cancer,33–36 did not test the CKD-EPI system to control neoplastic cell growth might also explain
equation,37 and included a small number of patients.34,36 the particular aggressiveness of neoplasia in this setting.
The largest and most thorough study so far, by Janowitz Indeed, the mortality rate for any specific neoplasia is
et al.,38 included >2000 patients with various types of cancers. significantly higher in patients with a transplant than in the
The study tested all main creatinine-based GFR formulas as general population.49
well as a new Janowitz equation validated against an estab- Given the high risk of neoplasia, prevention should be the
lished criterion standard such as 51Cr-EDTA. The new main objective of clinical care in this patient population. In
equation was slightly more precise than the CKD-EPI equation this perspective, the first aim is to exclude that either the
and showed no bias, denoting adequate accuracy.38 A GFR organ or the patient has an occult neoplasm at trans-
calculator based on the Janowitz equation is available online at plantation.50 Both donors’ and recipients’ screening has
http://tavarelab.cruk.cam.ac.uk/JanowitzWilliamsGFR/. For significantly changed in the last few years, because a general
extensive validation in diverse populations and the satisfactory history of neoplasia does not represent per se a contraindi-
accuracy and precision in patients with cancer, the CKD-EPI cation to transplantation, but is considered on a case-by-case
formula appears suitable for application in this population. basis.50,51 Screening needs to be expanded to include the
If further validated in other large databases, the new equa- whole posttransplant period, aiming for a timely diagnosis,
tion38 can emerge as the new standard of care for patients with which is particularly relevant.52 Finally, available evidence
cancer. suggests that in high-risk recipients the use of the mammalian
A formula based on the combination of creatinine and target of rapamycin (mTOR) inhibitor–based immunosup-
cystatin C measurements is the most reliable estimate of GFR pressive regimen might be beneficial.46,52
in the general population and in patients with CKD,39,40 but
there is no adequate validation study of this formula in the Cancer in patients with CKD
population with cancer. Cystatin C is influenced by inflam- Albuminuria is well recognized as an independent risk factor
mation and cell turnover, and in B-cell non-Hodgkin for cardiovascular disease, especially in patients with diabetes
lymphoma its synthesis is increased in immature dendritic and hypertension.53,54
cells,41,42 making this marker a less than ideal GFR surrogate. Clinical and experimental evidence suggests that endo-
Future directions for the direct measurement of GFR in pa- thelial dysfunction, chronic inflammation, and transvascular
tients with cancer include the development of fluorescence leakage of macromolecules may be responsible for the asso-
methods for real-time GFR determination.43 Specifically, dex- ciation between albuminuria and cardiovascular disease.55,56
trans of different molecular weight can be labeled by different In addition to this well-known connection, emerging evi-
fluorochromes and thus be used with 2-compartmental models dence indicates that albuminuria is linked to an increased risk
to measure the plasma volume (unfiltered dextrans) and GFR of cancer mortality. Studies using data from the Third Na-
(filtered dextrans). Furthermore, fluorescence could be detected tional Health and Nutrition Examination Survey (NHANES
at the level of the skin, thus avoiding taking blood samples. III) demonstrated that albuminuria is associated with an
Theoretically, this method could easily and rapidly measure GFR increased risk of cancer death from all causes, lung cancer,
and plasma volume and potentially estimate renal reserve (i.e., and prostate cancer in men 50 years and older. Interestingly,
the increase in GFR after a protein meal or infusion of amino this association was present only in men, as it was not sig-
acids44). This may be particularly useful in allowing for rapid nificant for any type of cancer in women of the same age.57 In
dose adjustments of potentially toxic drugs. However, several addition to this link, several studies have shown that baseline
questions remain regarding this new technology and it has yet to albuminuria is linked to increased cancer incidence. In one of
be implemented in clinical practice. these studies, subjects in the highest quartile of albuminuria
were 8.3- and 2.4-fold more likely to receive a diagnosis of
Cancer in patients with a solid organ transplant bladder and lung cancer, respectively.58
Neoplasia is the third cause of mortality in transplant re- Patients with CKD have a 5- to 15-fold increased risk of
cipients, and its incidence has been increasing over the past cardiovascular mortality.59,60 In addition, population-based
decade.45 These patients have a 3- to 5-fold increased risk of studies have demonstrated that patients with CKD are at a
developing solid organ cancers and a 60- to 200-fold higher risk of cancer, even those with mild to moderate stages
increased risk of skin cancers in comparison to the general of CKD.61 Patients with moderate CKD have an increased risk
population.46 Immunosuppressive therapy is the main cause of cancer mortality, especially those with liver and urinary
of this dramatic increase. Immunosuppression induces a tract cancer,62 while others have shown that modest re-
significant increase in viral infections, and most of the ductions in GFR <60 ml/min per 1.73 m2 are linked to an

Kidney International (2019) 96, 555–567 557


meeting report A Capasso et al.: Onco-nephrology: an emerging field in medicine

increased risk of cancer death.12 In the latter study, the asso- Finally, vaccines have failed to show an improved survival to
ciation between reduced kidney function and cancer death date.78 More recently, combination strategies involving
appeared to be specific for breast cancer and urinary tract contemporary immunotherapy have emerged as key oppor-
cancer. Other large longitudinal studies have shown a correla- tunities to further shift the treatment landscape.78 With the
tion between the severity of CKD and cancer mortality. Spe- advent of new therapies, median survival for patients with
cifically, mortality from liver cancer, kidney cancer, and urinary good-to-intermediate- and poor-risk metastatic RCC has
tract cancer increased incrementally with the severity of CKD.62 increased from 26, 14, and 7 months to 43, 23, and 8 months.78
These epidemiological data introduce a number of addi- For all those reasons, functional outcomes, overall survival,
tional topics concerning the difficulties in cancer treatment, and quality of life of cancer survivors gained increasing sci-
screening, and biopsy in patients with CKD. The treatment of entific attention in the past years.79 Efforts have been made to
cancer is difficult because of the reduced renal excretion clarify the pathophysiology and mechanisms underlying the
of drugs and different pharmacodynamics/pharmacokinetics development of CKD after nephron-sparing surgery or radical
of anticancer drugs in patients with CKD. For screening nephrectomy. Beyond the surgical aspects, much more
cancer in patients with CKD, we rely on standard guidelines emphasis has been placed to patients’ comorbidities and
for screening in the general population: specific surveillance baseline characteristics. That said, a multidisciplinary
protocols await further validation, and at present, consider- approach to the decision making in patients with a renal mass
able differences among nephrologists exist.63 Finally, the may maximize those outcomes (Figure 2). Unfortunately,
therapeutic and diagnostic strategies with regard to renal nowadays such an approach is not pursued in many urological
complications in patients with cancer are probably subopti- cancer centers. To investigate this aspect, the Kidney Cancer
mal at present. Renal biopsy has been recommended in case Young Academic Urologists Working party recently promoted
of glomerular damage (hypertension, edema, proteinuria, and an Internet-based survey with the aim to better understand
hematuria), interstitial disease (eosinophiluria), vascular dis- how urologists take care of renal function.80 Although virtually
ease/vasculitis, or unresolved tubular disease.64 all urologists (98%) answered that renal function is a critical
variable for decision making, the consultation of a nephrologist
Renal cancer: multidisciplinary treatment approaches after surgery for a patient with established CKD risk factors is
When Robson et al. initially described the management of contemplated in merely 17% of the cases. This is even more
renal cancer, surgery included the removal of the entire kid- important if we considered that only a quarter of the urologists
ney.65 Therefore, surgical extirpation was usually affected by a acknowledged albuminuria and obesity as key determinants of
significant decline in renal function and an increased risk of postoperative CKD. Of note, the latest European guidelines do
cardiovascular morbidity. Nowadays, the maximum preser- not suggest referring patients with RCC for a perioperative
vation of normal renal parenchyma is standard of care, when nephrological assessment. Conversely, the American guidelines
technically feasible.66 Nonetheless, up to 40% to 50% of pa- recommend nephrology referral after urological treatment, at
tients treated with radical nephrectomy and up to 10% to least when CKD and/or proteinuria is detected.
15% of nephron-sparing surgery cases experience renal A multidisciplinary management according to patients’
functional impairment at 1 year after surgery.67 individual risk factors appears mandatory to shift from a
In cases of metastatic renal cell carcinoma (RCC), systemic purely surgeon-oriented point of view to a holistic consid-
therapy has evolved rapidly over the past decade (Table 168–77). eration to further improve functional outcomes and quality of
Cytokines have largely been replaced in current practice by life of patients with RCC.81,82
tyrosine kinase inhibitors.78 The mammalian target of rapa-
mycin (mTOR) inhibitor therapy is nowadays considered Glomerular diseases associated with cancer
largely inferior to vascular endothelial growth factor (VEGF)– There is a strong association between cancer and glomeru-
based therapy, although it has a role in combination strategies.78 lonephritis.83,84 One study58 found that proteinuria is more

Table 1 | First-line setting pivotal trials of targeted systemic therapy in metastatic clear cell renal cell carcinoma6
Study Year Experimental arm(s) Control arm(s) Median overall survival (mo) (HR; 95% CI)
Motzer et al.68 2007 Sunitinib (n ¼ 375) IFN (n ¼ 375) 26 vs. 21 (0.82; 0.57–1.00)
Escudier et al.69 2007 Bevacizumab þ IFN (n ¼ 327) Placebo þ IFN (n ¼ 322) 23 vs. 21 (0.86; 0.72–1.04)
Hudes et al.70 2007 IFN þ temsirolimus (n ¼ 210) IFN (n ¼ 207) 8 vs. 11 vs. 7 (0.96; 0.76–1.20 and
Temsirolimus (n ¼ 209) 0.73; 0.58–0.92)
Rini et al.71 2008 Bevacizumab þ IFN (n ¼ 369) IFN (n ¼ 363) 18 vs. 17 (0.86; 0.73–1.01)
Sternberg et al.72 2010 Pazopanib (n ¼ 290) Placebo (n ¼ 145) 23 vs. 20 (0.91; 0.71–1.16)
Motzer et al.73 2013 Pazopanib (n ¼ 557) Sunitinib (n ¼ 553) 28 vs. 29 (0.92; 0.79–1.06)
Motzer et al.74 2013 Tivozanib (n ¼ 260) Sorafenib (n ¼ 257) 29 vs. 29 (1.24; 0.95–1.62)
Choueiri et al.75 2018 Cabozantinib (n ¼ 79) Sunitinib (n ¼ 78) 27 vs. 21 (0.80; 0.53–1.21)
Escudier et al.76 2017 Nivolumab þ ipilimumab (n ¼ 550) Sunitinib (n ¼ 546) NR vs. 26 (0.63; 0.44–0.89)
Motzer et al.77 2018 Atezolizumab þ bevacizumab (n ¼ 454) Sunitinib (n ¼ 461) NR vs. 26 (0.63; 0.44–0.89)
CI, confidence interval; HR, hazard ratio; IFN, interferon; NR, not reported.

558 Kidney International (2019) 96, 555–567


A Capasso et al.: Onco-nephrology: an emerging field in medicine meeting report

lymphocytic leukemia, hairy cell leukemia, and B-cell non-


Hodgkin lymphoma.
Nephrologist IgA nephropathy is associated with respiratory tract, oral
mucosa, and nasopharyngeal cancer; RCC, and cutaneous T-
Interventional Urologist cell lymphoma. It has been hypothesized that the invasion of
radiologist
the site-specific lymphoid-associated tissue by cancer cells
leads to increased circulating levels of IgA because of over-
production of interleukin-6.94

Nephrotoxicity from chemotherapy


Nutritionist Patient Medical
oncologist Nephrotoxicity of conventional chemotherapeutic agents re-
mains a significant problem in patients with cancer not only
because of its impact on the survival of patients with cancer
but also because of its dose-limiting effect on the adequate
cancer treatment. Although nephrotoxicity is a major side
Hematologist Psychologist effect of many chemotherapeutic agents, not all patients
exposed to these agents develop kidney injury. This suggests
the presence of several factors that could increase the patient’s
Geriatrician
risk of nephrotoxicity, including the patient’s age, intravas-
cular volume depletion, and the presence of underlying AKI
or CKD.95
Figure 2 | A proposal for a multidisciplinary approach to Conventional cytotoxic agents can cause nephrotoxicity by
maximize functional outcomes and quality of life in patients
various mechanisms involving different parts of kidney anat-
with renal cancer.
omy including glomerulus, tubules, interstitium, and renal
microvasculature with kidney manifestations ranging from an
frequent in patients with malignancies than in controls. Data acute interstitial nephritis, focal segmental glomerulosclerosis,
from the Danish Kidney Biopsy Register show a higher inci- and hypertension to various electrolyte disorders, capillary leak
dence of cancer in patients with glomerulopathy than in the syndrome, and thrombotic microangiopathy (TMA).13
general population.85 Unfortunately, a subset of patients who develop nephro-
Membranous nephropathy is the most frequent cancer- toxicity can give rise to long-term complications such as
associated glomerulonephritis.86 The pathogenesis is due to chronic interstitial nephritis and CKD.
subepithelial deposition of tumor antigens87 associated with A major kidney complication of both mitomycin C and
an enhanced immune reaction triggered by a malignant tu- gemcitabine, 2 other important conventional chemothera-
mor.88 Seronegativity to anti-phospholipase A2 receptor an- peutic agents, is the development of TMA. Interestingly, there
tibodies, immunohistochemical identification of IgG1 and are often findings of both acute and chronic TMA with these
IgG2 subclasses,89 and >8 inflammatory cells per glomerulus drugs. Whereas acute TMA is characterized by the formation
are the main criteria to differentiate idiopathic membranous of fibrin microthrombi in arteries, arterioles, and glomerular
nephropathy from cancer-related membranous nephropathy. capillaries, chronic TMA is typically considered as sclerosis
Minimal change disease occurs in w1% of patients with and “onion skin” appearance of arteries and arterioles.
Hodgkin lymphoma, and the occurrence of focal segmental Mitomycin C–induced TMA is dose dependent,96,97 whereas
glomerulosclerosis is about one-tenth that of minimal change no clear relationship has been observed between the cumu-
disease. Interleukin-13 serum levels seem to play an impor- lative dose of gemcitabine and the risk of TMA. The under-
tant role.90 Minimal change disease and focal segmental lying molecular mechanism of gemcitabine-induced TMA is
glomerulosclerosis are also associated with solid tumors. not well established; it might involve both direct endothelial
VEGF has been potentially implicated in pathogenesis injury and diminished activity of a von Willebrand factor
because of its ability to increase glomerular permeability.91 named ADAMTS-13 (a disintegrin and metalloproteinase
Rapid progressive glomerulonephritis can develop in pa- with a thrombospondin type 1 motif, member 13).98
tients with RCC, gastric cancer, and lung cancer. Conversely, TMA has also been observed after treatment with VEGF
patients with antineutrophil cytoplasmic antibody–associated inhibitors (such as bevacizumab) and inhibitors of VEGF
(ANCA) vasculitis exhibit a higher risk of a malignant tumor tyrosine kinase domain (such as sunitinib).99 These drugs,
than did the general population.92 collectively called VEGF signaling pathway inhibitors, are
Membranoproliferative glomerulonephritis has been plagued by an increased risk of cardiovascular disease due to
associated with solid tumors (lung, renal, and gastric); this is vascular toxicity. Indeed, up to 42% of patients treated with
an immune complex disease caused by tumor antigen for- bevacizumab and 34% of those treated with sunitinib develop
mation and the inability of the host to effectively clear these hypertension (Micromedex and Lexicomp data). It has been
antigens.93 It may also develop in the context of chronic even suggested that hypertension could be used as a sign of

Kidney International (2019) 96, 555–567 559


meeting report A Capasso et al.: Onco-nephrology: an emerging field in medicine

efficacy of VEGF signaling pathway inhibitors.100 The mice experiencing more severe AKI and dying prematurely
mechanism of VEGF signaling pathway–induced hyperten- after receiving cisplatin.114 In wild-type mice with AKI,
sion is thought to involve nitric oxide regulation at the treatment with agents that increase SIRT3 expression and
vascular level and reduced natriuresis at the level of the activity improves renal function and decrease tubular injury
kidney.100 by preventing changes in mitochondrial dynamics.114
Overall, a better definition of these interactions and Enhancing SIRT3 preserves cellular cytoskeleton integrity,
further understanding of the molecular mechanisms whereby enabling the intercellular transfer of healthy mitochondria via
cytotoxic agents induce nephrotoxicity could provide better tubulin-rich projections in cisplatin-injured PTCs.115 This
strategies to manage the kidney side effects of cytotoxic reparative dialogue between adjacent PTCs favors bio-
agents. energetic cross talk and redox balance, which is abolished by
SIRT3 silencing in PTCs.115
Molecular mechanisms of cisplatin-induced AKI The natural outcome of these studies is the search for
Cisplatin is an effective chemotherapeutic drug for a broad SIRT3-activating compounds. In this context, honokiol, a
spectrum of solid organ malignancies. Its main dose-limiting biphenolic compound derived from the bark of magnolia
side effect is its nephrotoxicity, which can present in many trees,116 has been shown to selectively increase SIRT3 and has
forms, the most serious and common being AKI.101 In early anti-inflammatory and antioxidant effects.117,118
clinical use, a dose-related incidence of cisplatin-induced AKI Furthermore, NADþ precursors including nicotinamide,
was reported in 14% to 100% of patients, which decreased to nicotinamide riboside, nicotinamide mononucleotide, or
20% to 30% more recently with the use of saline hydration exogenous NADþ have been found to be beneficial in cardiac
and diuresis.102,103 Diuresis induced by the atrial natriuretic and renal diseases119,120 to the extent that nicotinamide
factor was also tested,104 and parathyroidectomy is reported supplementation has been associated with less frequent AKI
to exert some beneficial action.104,105 in hospitalized patients.121
Cisplatin is traditionally believed to exert its antitumor
effect through its interaction with DNA, leading to the for- Immunotherapy and interstitial nephritis
mation of inter- and intrastrand cross-links, causing DNA Cancer immunotherapy exploits the immune system to fight
synthesis and replication arrest.106 However, recently an tumor cells. The availability today of selective immune
important question was raised on why kidney proximal stimulants has finally solidified the original idea into a ther-
tubular cells (PTCs) with relatively low rates of cell prolifer- apeutic success. The dark side of immunotherapy is the un-
ation are especially prone to cisplatin-induced cytotoxicity. avoidable presence of autoimmune effects, such as interstitial
One recent persuasive explanation is that nephrotoxicity nephritis. Specifically, the class of immunotherapy drugs
could result from mitochondrial damage and the release of called immune checkpoint inhibitors results in clinically
high levels of mitochondrial reactive oxygen species.107,108 relevant kidney damage in 1% to 3% of patients; however, the
This hypothesis is reinforced by the finding that along the percentage of subclinical interstitial nephritis might be much
proximal tubular epithelium, there are 2 membrane trans- higher, up to one-third of patients with immune checkpoint
porters that mediate cisplatin uptake in PTCs.109 inhibitors, according to autoptic series.122 Clearly this de-
Cisplatin binds both nuclear and mitochondrial DNA, pends on our limited ability to identify this renal damage with
leading to cell cycle arrest and mitochondrial dysfunction.102 classical blood tests: better biomarkers are thus needed. Im-
Because of their high-energy functions in active trans- mune checkpoint inhibitors could determine interstitial
port,110,111 PTCs have high mitochondrial density and are nephritis when T cells are primed by drugs associated with
particularly susceptible to cisplatin. tubular damage, such as proton pump inhibitors and
Mitochondria are highly dynamic organelles that continu- nonsteroidal anti-inflammatory drugs. The treatment is the
ously divide and unite through fission and fusion events to meet same as it is for other T cell–mediated kidney injuries (such as
cell energy demands.107 There is a large body of evidence after HIV infection and renal allograft rejection) and mostly
demonstrating that NADþ-dependent deacetylase sirtuin 3 depends on the use of steroids.123
(SIRT3) maintains mitochondrial vitality by regulating processes
such as energy homeostasis and antioxidant defenses.112,113 Electrolyte disorders in patients with cancer
In experimental cisplatin-induced AKI, reduced SIRT3 Electrolyte disorders are commonly seen in patients with any
levels are associated with oxidative stress and mitochondrial form of cancer.124 Table 2 lists the common etiologies of these
damage,114 leading to metabolic and functional impairment electrolyte disorders.
of PTCs.114 Mechanistically, cisplatin-induced reduction of Hyponatremia is the most common electrolyte disorder
SIRT3 levels triggers the upregulation of dynamin-related encountered in patients with cancer.125–129 Hyponatremia is
protein 1 and mitochondrial fission factor and the down- associated with increased mortality and poor response to
regulation of optic atrophy 1, tipping mitochondrial dy- therapy.129–132 The syndrome of inappropriate antidiuretic
namics toward fragmentation, along with loss of hormone may result from numerous malignancies and
mitochondrial membrane potential and organelle disposal.114 numerous chemotherapeutic regimens.133–136 A diagnosis
The functional role of SIRT3 is highlighted by SIRT3-deficient of hyponatremia may also be a marker of occult

560 Kidney International (2019) 96, 555–567


Kidney International (2019) 96, 555–567

A Capasso et al.: Onco-nephrology: an emerging field in medicine


Table 2 | Electrolyte disorders in onco-nephrology
Etiology Hyponatremia Hypernatremia Hypokalemia Hyperkalemia Hypophosphatemia Hyperphosphatemia Hypomagnesemia

Tumor-related - Paraneoplastic - Central diabetes - Lysozymuria with acute - Tumor lysis syndrome - Hyperparathyroidism - Tumor lysis syndrome - None
SIADH (e.g., squamous insipidus leukemia - Pseudohyperkalemia (parathyroid - Hyperglobulinemia
cell carcinoma - Cushing - Mineralocorticoid excess - Adrenal insufficiency carcinoma) (pseudohyperparathyroidism)
of the lung) syndrome syndrome (metastatic disease) - PTHrP-secreting
- Paraneoplastic nephrotic - Primary hyperaldosteronism - Acute kidney injury tumors
syndrome (adrenal carcinoma) (tumor-induced) - Tumor-induced
- Adrenal insufficiency - Renin-producing tumors osteomalacia
(metastatic disease) (extremely rare) (mesenchymal
- Ectopic adrenocorticotropin tumors)
syndrome - Light chain–associated
- Intracellular shifts Fanconi syndrome
(pseudohypokalemia) in
the setting of high white
cell counts
Chemotherapy- - Medication-induced - Nephrogenic - Chemotherapeutic agents - Tumor lysis syndrome - Drug-induced - Acute kidney injury - Cisplatin
related SIADH (cyclophosphamide, diabetes (cisplatin and ifosfamide) - Acute kidney injury Fanconi - Bisphosphonates - Calcineurin
vinblastine, vincristine, insipidus - Antimicrobial and (medication-induced) syndrome inhibitors
etc.) - Diarrhea and antifungal agents - Drug-induced (heparin, - Tyrosine kinase - Antibodies to
- Salt-losing nephropathy increased free (aminoglycosides and trimethoprim, inhibitors (imatinib) EGF receptor
(cisplatin) water losses amphotericin B) calcineurin inhibitors, - mTOR inhibitors (cetuximab and
- Drug-induced Fanconi - Excessive gastrointestinal and ketoconazole) - VEGF inhibitors panitumumab)
syndrome losses (vomiting and (sorafenib) - Vomiting
- Nausea/vomiting diarrhea) - Diarrhea
- Diarrhea - Drug-induced Fanconi
syndrome
Other causes - Hypothyroidism - Hyperglycemia - Hypercalcemia - Blood transfusions - Hyperalimentation - Rhabdomyolysis - Proton pump
(drug-induced) and osmotic - Hypomagnesemia (refeeding syndrome) - Hypoparathyroidism inhibitors
- Cirrhosis diuresis - Postobstructive diuresis - Vitamin D deficiency (postsurgical) - Aminoglycosides
- Heart failure - Use of growth factors - Malabsorption - Amphotericin B
- Hypercalcemia
EGF, endothelial growth factor; mTOR, mammalian target of rapamycin; PTHrP, parathyroid hormone–related protein; SIADH, syndrome of inappropriate antidiuretic hormone secretion; VEGF, vascular endothelial growth factor.

meeting report
561
meeting report A Capasso et al.: Onco-nephrology: an emerging field in medicine

neoplasms.137,138 Treatment of hyponatremia in patients with importance is pseudohyperkalemia, usually in the setting of
cancer is challenging and often suboptimal.139 Blockade of the marked leukocytosis or thrombocytosis.157
type 2 vasopressin receptor with tolvaptan may be particularly Hypophosphatemia can occur with proximal tubular
useful in cases of refractory syndrome of inappropriate dysfunction (Fanconi syndrome) because of toxic light chains
antidiuretic hormone.140 At least in patients with non–small in multiple myeloma.160 A rare etiology of hypophosphatemia
cell lung cancer, normalization of serum sodium was is the syndrome of tumor-induced osteomalacia, in which
associated with improved prognosis.141 tumor production of phosphaturic factors, such as fibroblast
Hypernatremia is less commonly seen and may be due to growth factor 23, results in decreased renal tubular reab-
malignant involvement of the hypothalamic-pituitary axis or sorption of phosphate.161
hypercalcemia-induced nephrogenic diabetes insipidus.129,142 In patients with multiple myeloma and Waldenström
Hypernatremia is associated with substantial mortality risk.143 macroglobulinemia, circulating monoclonal proteins can
Hypercalcemia is common in multiple myeloma and interfere with the laboratory measurement of phosphate,
advanced stage cancers.144 resulting in spuriously elevated serum phosphate levels
Survival in patients with cancer-associated hypercalcemia (pseudohyperphosphatemia).162
is dismal.145,146 Hypercalcemia is most often caused by the Hypomagnesemia can be due to gastrointestinal losses or
release of parathyroid hormone–related peptide or local renal tubular dysfunction due to chemotherapy.163,164
osteolysis (mediated by cytokines) (Figure 3).147–151 Treat- Chemotherapeutic agents with hypomagnesemia include
ment of cancer-associated hypercalcemia includes intravenous cisplatin and cetuximab.154,164
hydration to increase renal calcium excretion, followed by
either bisphosphonate, calcitonin, or denosumab, to decrease Cannabinoids, endocannabinoids, and cancer
the bone release of calcium.147,152 The psychotropic principle of Cannabis sativa, D9-tetrahydro-
Hypokalemia results from gastrointestinal or renal los- cannabinol, and its synthetic derivative, nabilone, are already
ses (due to ifosfamide, cisplatin, non-Kþ–sparing diuretics, used for nausea induced by chemotherapeutic agents and have
or rarely leukemia-associated lysozymuria).129,133,153,154 also been suggested to reduce cachexia, another possible conse-
Hypokalemia may also be caused by the paraneoplastic quence of cancer.165 However, the discovery of the mechanism of
secretion of ectopic adrenocorticotropin hormone.155,156 action of D9-tetrahydrocannabinol via 2 G protein–coupled re-
Spurious hypokalemia (pseudohypokalemia) may occur ceptors, known as cannabinoid receptor type 1 (CB1) and 2
in patients with acute myeloid leukemia and a marked (CB2), and the fact that these receptors are activated by endog-
leukocytosis.157 enous lipid mediators, known as endocannabinoids, have opened
Hyperkalemia may result from tumor lysis syn- new therapeutic possibilities for several tumors, beyond merely
drome.158,159 Less common causes include adrenal insuffi- palliative treatments.166 In fact, the activation of CB1 and/or CB2
ciency and drugs (calcineurin inhibitors). Of particular by D9-tetrahydrocannabinol, endocannabinoids, and several

Malignancy-Associated Malignancy-Associated
Hypercalcemia Hypocalcemia

Low PTH High PTH Drugs Low Mg2+


Humoral Osteolysis Rare Causes
Hypercalcemia

Postsurgical Tumor lysis Denosumab


PTHrp-related Bony Elevated removal of
paraneoplas c metastases calcitriol levels parathyroid Osteoblas c Cinacalcet
syndromes and local or ectopic PTH glands metastases
cytokine secre on
Radia on
Examples: ac vity damage
-Squamous cell Examples:
carcinoma Examples: Lymphoma Cancer
-Renal cell cancer Breast cancer Ovarian cancer infiltra on of
-Breast cancer Mul ple Thyroid papillary gland
-Ovarian cancer myeloma cancer
-Prostate cancer Lymphoma Pancrea c cancer
-Lymphoma Leukemia
-Leukemia

Figure 3 | Cancer associated with calcium disorders. Both hyper- and hypocalcemia can be associated with an underlying diagnosis of
cancer. In each case, specific mechanisms and etiologies are operative. Although hypercalcemia is typically due to the effects of the underlying
cancer, hypocalcemia is usually the result of therapeutic maneuvers (surgery, chemotherapies, or radiation therapy). PTH, parathyroid
hormone; PTHrP, parathyroid hormone–related protein.

562 Kidney International (2019) 96, 555–567


A Capasso et al.: Onco-nephrology: an emerging field in medicine meeting report

synthetic agonists of such receptors was shown to interfere with Berne, Switzerland; Amit Alahoti, Houston, Texas, USA; Todd R. Alexander,
Toronto, Alberta, Canada; Lucia Altucci, Naples, Italy; Hatem Amer, Rochester,
the growth of several types of solid tumors, including, but not Minnesota, USA; Vincenzo Barone, Pisa, Italy; Ariela Benigni, Bergamo, Italy;
limited to, breast, prostate, and colorectal carcinomas, glioblas- Luigi Biancone, Turin, Italy; Joseph V. Bonventre, Boston, Massachusetts, USA;
toma, as well as skin cancers, including malignant melanomas Giovanni Camussi, Turin, Italy; Anna Capasso, Denver, Colorado, USA; Fortunato
Ciardiello, Naples, Italy; Umberto Capitanio, Milan, Italy; Michele Caraglia,
in vitro and in vivo, in xenograft as well as genetic experimental Naples, Italy; Giacomo Cartenì, Naples, Italy; Andrés Cervantes, Valencia, Spain;
models.167 The anticancer action of CB1 and CB2 agonists occurs Franco Citterio, Rome, Italy; Laura Cosmai, Milan, Italy; Farhad R. Danesh,
through several parallel mechanisms, including inhibition of the Houston, Texas, USA; Bruno Daniele, Benevento, Italy; Antonietta D’Errico,
Bologna, Italy; Ferdinando De Vita, Naples, Italy; Vincenzo Di Marzo, Laval,
cell cycle, induction of apoptosis, inhibition of neo- Quebec, Canada; Antonio Ereditato, Berne, Switzerland; Geppino Falco, Naples,
vascularization, and counteraction of metastatic processes.168 Italy; Denis Fouque, Lyon, France; Renato Franco, Naples, Italy; Maurizio Gal-
Furthermore, inhibitors of endocannabinoid enzymatic inacti- lieni, Milan, Italy; Giovanni Gambaro, Rome, Italy; Loreto Gesualdo, Bari, Italy;
Giuseppe Grandaliano, Foggia, Italy; Calvin Kuo, Stanford, California, USA; Edgar
vation were shown to be effective and possibly devoid of the A. Jaimes, New York, New York, USA; Vincent Launay-Vacher, Paris, France;
unwanted psychotropic effects of CB1 agonists.169 More recently, Evaristo Maiello, San Giovanni Rotondo, Italy; Francesca Mallamaci, Reggio
however, nonpsychotropic cannabinoids that act only in part via Calabria, Italy; Jolanta Malysxko, Bialystok, Poland; Gennaro Marino, Naples,
Italy; Erica Martinelli, Naples, Italy; Giuseppe Matarese, Naples, Italy; Takeshi
the endocannabinoid system and hit several molecular targets Matsubara, Kyoto, Japan; Piergiorgio Messa, Milan, Italy; Carlo Messina, Milan,
have been suggested to be effective anticancer agents.170 Their use Italy; Vincenzo Mirone, Naples, Italy; Floriana Morgillo, Naples, Italy; Alessandro
as add-on therapies to chemotherapeutic or biological drugs Nanni Costa, Rome, Italy; Michele Orditura, Naples, Italy; Antonello Pani,
Cagliari, Italy; Mark Anthony Perazella, New Haven, Connecticut, USA; Ales-
should be considered in future clinical trials. sandra Perna, Naples, Italy; Claudio Pisano, Ariano Irpino, Italy; Todd Pitts,
Denver, Colorado, USA; Camillo Porta, Pavia, Italy; Giuseppe Procopio, Milan,
Future directions and research agenda for onco-nephrology Italy; Qi Qian, Rochester, Minnesota, USA; Giuseppe Remuzzi, Bergamo, Italy;
Pierre Ronco, Paris, France; Mitchell H. Rosner, Charlottesville, Virginia, USA;
Onco-nephrology is an emerging and expanding field and, as Domenico Russo, Naples, Italy; Lilian L. Siu, Toronto, Ontario, Canada; Walter
such, the items to be investigated are numerous. The amount Stadler, Chicago, Illinois; USA; Francesco Trepiccione, Naples, Italy; Teresa
of information is impressive, but this contrasts with our poor Troiani, Naples, Italy; Davide Viggiano, Ariano Irpino, and Alessandro Weisz,
Salerno, Italy; Andrzej Wie˛ cek, Katowice, Poland; Ding Xiaoqiang, Shanghai,
understanding of the phenomenon and our limited ability to China; Ortensio Zecchino, Ariano Irpino, Italy; Carmine Zoccali, Reggio Calabria,
predict and treat nephrotoxicity. Italy.
All nephrologists need to be acquainted with these emerging
issues and need to constantly improve their knowledge of the DISCLOSURE
EAJ is a cofounder, stock holder, and chief of the Medical Advisory Board of
nephrotoxic profile of an increasingly growing number of Goldilocks Inc. MHR is a member of AbbVie Data Safety Monitoring Board,
oncological treatments. In addition, the distinctive issues sur- Retrophin Data Safety Monitoring Board, and Baxter consultant. VDM receives
rounding the diagnosis and treatment of cancer in patients research grants from GW Pharmaceuticals. All the other authors declared no
competing interests.
with kidney disease need additional investigation.
In conclusion, we report a short, nonexhaustive list of
subjects for a research agenda, which mirror the topics ACKNOWLEDGMENTS
The conference was supported by a generous grant from the
addressed in the article:
Menarini International Foundation. EAJ was supported by P30
(i) to explore novel methods to rapidly and easily measure CA008748, RO1 DK114321, and Byrne Research Funds. FRD received
GFR in the field of onco-nephrology; support from National Institutes of Health grants R01 DK078900 and
(ii) to institute an international registry of cancer in trans- R01 DK091310. GC received support from POR CAMPANIA ICURE CUP
planted patients; B21C17000030007.
The conference received the official endorsement of the following
(iii) to understand the higher risk of cancer in CKD, glo-
societies: Italian Society of Nephrology (SIN), Italian Association of
merulopathies, and kidney transplant recipients; Medical Oncology (AIOM), European Renal Association – European
(iv) to verify which subpopulations with TMA might gain Dialysis and Transplant Association (ERA-EDTA), and International
advantage from the use of eculizumab or other novel Society of Nephrology (ISN). The meeting was organized in
therapies; collaboration with the University of Campania “L. Vanvitelli” and
BioGeM.
(v) to study approaches for nephroprotection from che-
motherapies such as with cisplatin treatment (see, e.g.,
inhibitors of dipeptidyl peptidase 4 [DPP-4])171; REFERENCES
(vi) to evaluate new biomarkers for AKI (e.g., neutrophil 1. Xu J, Murphy SL, Kochanek KD, et al. Deaths: final data for 2013. Natl
gelatinase-associated lipocalin [NGAL]) in the field of Vital Stat Rep. 2016;64:1–119.
2. Cosmai L, Porta C, Gallieni M, et al. Onco-nephrology: a decalogue.
drug-induced nephrotoxicity and plan possible follow- Nephrol Dial Transplant. 2016;31:515–519.
up protocols; and 3. Janus N, Launay-Vacher V, Byloos E, et al. Cancer and renal insufficiency
(vii) early identification of immunotherapy-linked results of the BIRMA study. Br J Cancer. 2010;103:1815–1821.
4. Benoit DD, Hoste EA. Acute kidney injury in critically ill patients with
nephrotoxicity. cancer. Crit Care Clin. 2010;26:151–179.
5. Canet E, Zafrani L, Lambert J, et al. Acute kidney injury in patients with
newly diagnosed high-grade hematological malignancies: impact on
APPENDIX remission and survival. PLoS One. 2013;8, e55870.
List of the International Conference on Onco-Nephrology Participants 6. Salahudeen AK, Doshi SM, Pawar T, et al. Incidence rate, clinical
The following people attended the International Conference on Onco- correlates, and outcomes of AKI in patients admitted to a
Nephrology held on December 7 to 9, 2017, in Naples, Italy: Ariga Akitaka, comprehensive cancer center. Clin J Am Soc Nephrol. 2013;8:347–354.

Kidney International (2019) 96, 555–567 563


meeting report A Capasso et al.: Onco-nephrology: an emerging field in medicine

7. Christiansen CF, Johansen MB, Langeberg WJ, et al. Incidence of acute 33. Trobec K, Knez L, Mesko Brguljan P, et al. Estimation of renal function in
kidney injury in cancer patients: a Danish population-based cohort lung cancer patients. Lung Cancer. 2012;76:397–402.
study. Eur J Intern Med. 2011;22:399–406. 34. Quinton A, Lewis P, Ali P, et al. A comparison of measured and
8. Libório AB, Abreu KL, Silva GB Jr, et al. Predicting hospital mortality in estimated glomerular filtration rate for carboplatin dose calculation in
critically ill cancer patients according to acute kidney injury severity. stage I testicular seminoma. Med Oncol. 2013;30:661.
Oncology. 2011;80:160–166. 35. Cathomas R, Klingbiel D, Geldart TR, et al. Relevant risk of carboplatin
9. Darmon M, Ciroldi M, Thiery G, et al. Clinical review: specific aspects of underdosing in cancer patients with normal renal function using
acute renal failure in cancer patients. Crit Care. 2006;10:211. estimated GFR: lessons from a stage I seminoma cohort. Ann Oncol.
10. Launay-Vacher V, Oudard S, Janus N, et al, Renal Insufficiency and 2014;25:1591–1597.
Cancer Medications (IRMA) Study Group. Prevalence of renal 36. Kaag D. Carboplatin dose calculation in lung cancer patients with low
insufficiency in cancer patients and implications for anticancer drug serum creatinine concentrations using CKD-EPI and Cockcroft-Gault
management: the renal insufficiency and anticancer medications with different weight descriptors. Lung Cancer. 2013;79:54–58.
(IRMA) study. Cancer. 2007;110:1376–1384. 37. Ainsworth NL, Marshall A, Hatcher H, et al. Evaluation of glomerular
11. Launay-Vacher V. Epidemiology of chronic kidney disease in cancer filtration rate estimation by Cockcroft-Gault, Jelliffe, Wright and
patients: lessons from the IRMA study group. Semin Nephrol. 2010;30: Modification of Diet in Renal Disease (MDRD) formulae in oncology
548–556. patients. Ann Oncol. 2012;23:1845–1853.
12. Iff S, Craig JC, Turner R, et al. Reduced estimated GFR and cancer 38. Janowitz T, Williams EH, Marshall A, et al. New model for estimating
mortality. Am J Kidney Dis. 2014;63:23–30. glomerular filtration rate in patients with cancer. J Clin Oncol. 2017;35:
13. Na SY, Sung JY, Chang JH, et al. Chronic kidney disease in cancer: an 2798–2805.
independent predictor of cancer-specific mortality. Am J Nephrol. 39. Inker LA, Schmid CH, Tighiouart H, et al. Estimating glomerular filtration
2011;33:121–130. rate from serum creatinine and cystatin C. N Engl J Med. 2012;367:
14. Rosner MH, Perazella MA. Acute kidney injury in patients with cancer. 20–29.
N Engl J Med. 2017;376:1770–1781. 40. Stevens LA, Coresh J, Schmid CH, et al. Estimating GFR using serum
15. Perazella MA, Dreicer R, Rosner MH. Renal cell carcinoma for the cystatin C alone and in combination with serum creatinine: a pooled
nephrologist. Kidney Int. 2018;94:471–483. analysis of 3,418 individuals with CKD. Am J Kidney Dis. 2008;51:
16. Yang Y, Li HY, Zhou Q, et al. Renal function and all-cause mortality risk 395–406.
among cancer patients. Medicine (Baltimore). 2016;95:e3728. 41. Barreto JN, McClanahan AL, Rule AD, et al. Incorporating cystatin C to
17. Byrne AT, Alférez DG, Amant F, et al. Interrogating open issues in cancer predict methotrexate elimination in patients with CNS lymphoma and
precision medicine with patient-derived xenografts. Nat Rev Cancer. suspicious renal function. Case Rep Hematol. 2018;2018:7169897.
2017;17:254–268.  ckusic E, et al. Cystatin C as a potential
42. Mulaomerovic A, Halilbasic A, Ci
18. Gao H, Korn JM, Ferretti S, et al. High-throughput screening using marker for relapse in patients with non-Hodgkin B-cell lymphoma.
patient-derived tumor xenografts to predict clinical trial drug response. Cancer Lett. 2007;248:192–197.
Nat Med. 2015;21:1318–1325. 43. Molitoris BA, Reilly ES. Quantifying glomerular filtration rates in acute
19. Leung KM, Yeoh GP, Chan JK, et al. Ductal type signet ring cell kidney injury: a requirement for translational success. Semin Nephrol.
carcinoma of breast with growth pattern of pure mucinous carcinoma. 2016;36:31–41.
Pathology. 2011;43:282–284. 44. Anastasio P, Viggiano D, Zacchia M, et al. Delay in renal hemodynamic
20. Tentler JJ, Tan AC, Weekes CD, et al. Patient-derived tumour xenografts response to a meat meal in severe obesity. Nephron. 2017;136:151–157.
as models for oncology drug development. Nat Rev Clin Oncol. 2012;9: 45. Vajdic C, van Leeuwen MT. Cancer incidence and risk factors after solid
338–350. organ transplantation. Int J Cancer. 2009;125:1747–1754.
21. Hidalgo M, Amant F, Biankin AV, et al. Patient-derived xenograft 46. Piselli P, Serraino D, Segoloni GP, et al. Risk of de novo cancers after
models: an emerging platform for translational cancer research. Cancer transplantation. Eur J Cancer. 2013;49:336–344.
Discov. 2014;4:998–1013. 47. Piselli P, Busnach G, Fratino L, et al. De novo malignancies after organ
22. Jenkins RW, Aref AR, Lizotte PH, et al. Ex vivo profiling of PD-1 blockade transplantation: focus on viral infections. Curr Mol Med. 2013;13:
using organotypic tumor spheroids. Cancer Discov. 2018;8:196–215. 1217–1222.
23. Mestas J, Hughes CC. Of mice and not men: differences between mouse 48. O’Donovan P, Perrett CM, Zhang X, et al. Azathioprine and UVA light
and human immunology. J Immunol. 2004;172:2731–2738. generate mutagenic oxidative DNA damage. Science. 2005;309:
24. Takao K, Miyakawa T. Genomic responses in mouse models greatly 1871–1874.
mimic human inflammatory diseases. Proc Natl Acad Sci U S A. 2015;112: 49. Farrugia D, Mahboob S, Cheshire J, et al. Malignancy-related mortality
1167–1172. following kidney transplantation is common. Kidney Int. 2014;85:
25. Seok J, Warren HS, Cuenca AG, et al. Genomic responses in mouse 1395–1403.
models poorly mimic human inflammatory diseases. Proc Natl Acad Sci 50. Desai R, Collett D, Watson CJ. Estimated risk of cancer transmission
U S A. 2013;110:3507–3512. from organ donor to graft recipient in a national transplantation
26. Walsh NC, Kenney LL, Jangalwe S, et al. Humanized mouse models of registry. Br J Surg. 2014;101:768–774.
clinical disease. Annu Rev Pathol. 2017;12:187–215. 51. Abramowicz D, Cochat P, Claas FH, et al. European Renal Best Practice
27. Shultz LD, Lyons BL, Burzenski LM, et al. Human lymphoid and myeloid Guideline on kidney donor and recipient evaluation and perioperative
cell development in NOD/LtSz-scid IL2R gamma null mice engrafted care. Nephrol Dial Transplant. 2015;30:1790–1797.
with mobilized human hemopoietic stem cells. J Immunol. 2005;174: 52. Stallone G, Infante B, Grandaliano G. Management and prevention of
6477–6489. post-transplant malignancies in kidney transplant recipients. Clin Kidney
28. Melkus MW, Estes JD, Padgett-Thomas A, et al. Humanized mice mount J. 2015;8:637–642.
specific adaptive and innate immune responses to EBV and TSST-1. Nat 53. Karalliedde J, Viberti G. Microalbuminuria and cardiovascular risk. Am J
Med. 2006;12:1316–1322. Hypertens. 2004;17:986–993.
29. Lan P, Tonomura N, Shimizu A, et al. Reconstitution of a functional 54. Hillege HL, Fidler V, Diercks GF, et al. Urinary albumin excretion predicts
human immune system in immunodeficient mice through combined cardiovascular and noncardiovascular mortality in general population.
human fetal thymus/liver and CD34þ cell transplantation. Blood. Circulation. 2002;106:1777–1782.
2006;108:487–492. 55. Kshirsagar AV, Bomback AS, Bang H, et al. Association of C-reactive
30. Wang M, Yao LC, Cheng M, et al. Humanized mice in studying efficacy protein and microalbuminuria (from the National Health and Nutrition
and mechanisms of PD-1-targeted cancer immunotherapy. FASEB J. Examination Surveys, 1999 to 2004). Am J Cardiol. 2008;101:401–406.
2018;32:1537–1549. 56. Leoncini G, Sacchi G, Ravera M, et al. Microalbuminuria is an integrated
31. Capasso A, Lang J, Pitts TM, et al. Characterization of immune responses marker of subclinical organ damage in primary hypertension. J Hum
to anti-PD-1 mono and combination immunotherapy in hematopoietic Hypertens. 2002;16:399–404.
humanized mice implanted with tumor xenografts. J Immunother 57. Lin YS, Chiu FC, Lin JW, et al. Association of albuminuria and cancer
Cancer. 2019;7:37. mortality. Cancer Epidemiol Biomarkers Prev. 2010;19:2950–2957.
32. Canadian Cancer Association. Glomerular filtration rate study. Available 58. Jorgensen L, Heuch I, Jenssen T, et al. Association of albuminuria and
at: http://www.cancer.ca. Accessed March 5, 2019. cancer incidence. J Am Soc Nephrol. 2008;19:992–998.

564 Kidney International (2019) 96, 555–567


A Capasso et al.: Onco-nephrology: an emerging field in medicine meeting report

59. Wen CP, Matsushita K, Coresh J, et al. Relative risks of chronic kidney 84. Jachiet V, Mekinian A, Carrat F, et al, French Network of systemic and
disease for mortality and end-stage renal disease across races are immune disorders associated with hemopathies and cancer
similar. Kidney Int. 2014;86:819–827. (MINHEMON). Autoimmune manifestations associated with lymphoma:
60. Tonelli M, Wiebe N, Culleton B, et al. Chronic kidney disease and characteristics and outcome in a multicenter retrospective cohort
mortality risk: a systematic review. J Am Soc Nephrol. 2006;17: study. Leuk Lymphoma. 2018;59:1399–1405.
2034–2047. 85. Birkeland SA, Storm HH. Glomerulonephritis and malignancy: a
61. Wong G, Hayen A, Chapman JR, et al. Association of CKD and cancer population-based analysis. Kidney Int. 2003;63:716–721.
risk in older people. J Am Soc Nephrol. 2009;20:1341–1350. 86. Lefaucheur C, Stengel B, Nochy D, et al., GN-PROGRESS Study Group.
62. Weng PH, Hung KY, Huang HL, et al. Cancer-specific mortality in chronic Membranous nephropathy and cancer: epidemiologic evidence and
kidney disease: longitudinal follow-up of a large cohort. Clin J Am Soc determinants of high-risk cancer association. Kidney Int. 2006;70:
Nephrol. 2011;6:1121–1128. 1510–1517.
63. James L, Wong G, Craig J, et al. Nephrologists’ perspectives on cancer 87. Beaufils H, Jouanneau C, Chomette G. Kidney and cancer: results of
screening in patients with chronic kidney disease: an interview study. immunofluorescence microscopy. Nephron. 1985;40:303–308.
Nephrology (Carlton). 2019;24:414–421. 88. Pascal RR, Iannaccone PM, Rollwagen FM, et al. Electron microscopy
64. Assayag M, Rouvier P, Gauthier M, et al. Renal failure during and immunofluorescence of glomerular immune complex deposits in
chemotherapy: renal biopsy for assessing subacute nephrotoxicity of cancer patients. Cancer Res. 1976;36:43–47.
pemetrexed. BMC Cancer. 2017;17:770. 89. Qu Z, Liu G, Li J, et al. Absence of glomerular IgG4 deposition in
65. Robson CJ, Churchill BM, Anderson W. The results of radical patients with membranous nephropathy may indicate malignancy.
nephrectomy for renal cell carcinoma. J Urol. 2017;197:S111–S113. Nephrol Dial Transplant. 2012;27:1931–1937.
66. Capitanio U, Montorsi F. Renal cancer. Lancet. 2016;387:894–906. 90. Lai KW, Wei CL, Tan LK, et al. Overexpression of interleukin-13 induces
67. Scosyrev E, Messing EM, Sylvester R, et al. Renal function after nephron- minimal-change-like nephropathy in rats. J Am Soc Nephrol. 2007;18:
sparing surgery versus radical nephrectomy: results from EORTC 1476–1485.
randomized trial 30904. Eur Urol. 2014;65:372–377. 91. Taniguchi K, Fujioka H, Torashima Y, et al. Rectal cancer with
68. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa paraneoplastic nephropathy: association of vascular endothelial growth
in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115–124. factor. Dig Surg. 2004;21:455–457.
69. Escudier B, Pluzanska A, Koralewski P, et al. Bevacizumab plus 92. Tatsis E, Reinhold-Keller E, Steindorf K, et al. Wegener’s granulomatosis
interferon alfa-2a for treatment of metastatic renal cell carcinoma: associated with renal cell carcinoma. Arthritis Rheum. 1999;42:751–756.
a randomised, double-blind phase III trial. Lancet. 2007;370: 93. Ahmed M, Solangi K, Abbi R, et al. Nephrotic syndrome, renal failure,
2103–2111. and renal malignancy: an unusual tumor-associated
70. Hudes G, Carducci M, Tomczak P, et al. Temsirolimus, interferon alfa, or glomerulonephritis. J Am Soc Nephrol. 1997;8:848–852.
both for advanced renal-cell carcinoma. N Engl J Med. 2007;356:2271– 94. Mustonen J, Pasternack A, Helin H. IgA mesangial nephropathy in
2281. neoplastic diseases. Contrib Nephrol. 1984;40:283–291.
71. Rini BI, Halabi S, Rosenberg JE, et al. Bevacizumab plus interferon alfa 95. Perazella MA. Onco-nephrology: renal toxicities of chemotherapeutic
compared with interferon alfa monotherapy in patients with metastatic agents. Clin J Am Soc Nephrol. 2012;7:1713–1721.
renal cell carcinoma: CALGB 90206. J Clin Oncol. 2008;26:5422–5428. 96. Lesesne JB, Rothschild N, Erickson B, et al. Cancer-associated hemolytic-
72. Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced uremic syndrome: analysis of 85 cases from a national registry. J Clin
or metastatic renal cell carcinoma: results of a randomized phase III Oncol. 1989;7:781–789.
trial. J Clin Oncol. 2010;28:1061–1068. 97. Medina PJ, Sipols JM, George JN. Drug-associated thrombotic
73. Motzer RJ, Hutson TE, Cella D, et al. Pazopanib versus sunitinib in thrombocytopenic purpura-hemolytic uremic syndrome. Curr Opin
metastatic renal-cell carcinoma. N Engl J Med. 2013;369:722–731. Hematol. 2001;8:286–293.
74. Motzer RJ, Nosov D, Eisen T, et al. Tivozanib versus sorafenib as initial 98. Izzedine H, Isnard-Bagnis C, Launay-Vacher V, et al. Gemcitabine-
targeted therapy for patients with metastatic renal cell carcinoma: induced thrombotic microangiopathy: a systematic review. Nephrol Dial
results from a phase III trial. J Clin Oncol. 2013;31:3791–3799. Transplant. 2006;21:3038–3045.
75. Choueiri TK, Hessel C, Halabi S, et al. Cabozantinib versus sunitinib as 99. Eremina V, Jefferson JA, Kowalewska J, et al. VEGF inhibition and renal
initial therapy for metastatic renal cell carcinoma of intermediate or thrombotic microangiopathy. N Engl J Med. 2008;358:1129–1136.
poor risk (Alliance A031203 CABOSUN randomised trial): progression- 100. Touyz RM, Herrmann SMS, Herrmann J. Vascular toxicities with VEGF
free survival by independent review and overall survival update. Eur J inhibitor therapies—focus on hypertension and arterial thrombotic
Cancer. 2018;94:115–125. events. J Am Soc Hypertens. 2018;12:409–425.
76. Escudier B, Motzer RJ, Sharma P, et al. Treatment beyond progression in 101. Bellomo R, Kellum JA, Ronco C. Acute kidney injury. Lancet. 2012;380:
patients with advanced renal cell carcinoma treated with Nivolumab in 756–766.
CheckMate 025. Eur Urol. 2017;72:368–376. 102. Miller RP, Tadagavadi RK, Ramesh G, et al. Mechanisms of cisplatin
77. McDermott DF, Huseni MA, Atkins MB, et al. Clinical activity and nephrotoxicity. Toxins (Basel). 2010;2:2490–2518.
molecular correlates of response to atezolizumab alone or in 103. Capasso G, Rosati C, Ciani F, et al. The beneficial effect of atrial
combination with bevacizumab versus sunitinib in renal cell carcinoma. natriuretic peptide on cyclosporine nephrotoxicity. Am J Hypertens.
Nat Med. 2018;24:749–757. 1990;3:204–210.
78. Lalani A-KA, McGregor BA, Albiges L, et al. Systemic treatment of 104. Capasso G, Giordano DR, de Tommaso G, et al. Parathyroidectomy has a
metastatic clear cell renal cell carcinoma in 2018: current paradigms, beneficial effect on experimental cisplatin nephrotoxicity. Clin Nephrol.
use of immunotherapy, and future directions. Eur Urol. 2019;75: 1990;33:184–191.
100–110. 105. Capasso G, Giordano DR, De Santo NG, et al. The effect of parathyroid
79. Antonelli A, Minervini A, Sandri M, et al. Below safety limits, every unit hormone on cisplatin nephrotoxicity. Adv Exp Med Biol. 1989;252:
of glomerular filtration rate counts: assessing the relationship between 325–329.
renal function and cancer-specific mortality in renal cell carcinoma. Eur 106. Sengupta P, Basu S, Soni S, et al. Cholesterol-tethered platinum II-based
Urol. 2018;74:661–667. supramolecular nanoparticle increases antitumor efficacy and reduces
80. Capitanio U, Larcher A, Kriegmair MC, et al. Do we truly care about the nephrotoxicity. Proc Natl Acad Sci U S A. 2012;109:11294–11299.
functional outcomes for renal cancer patients? Multidisciplinarity is still 107. Brooks C, Wei Q, Cho SG, et al. Regulation of mitochondrial dynamics in
far away. Eur Urol. 2019;75:349–350. acute kidney injury in cell culture and rodent models. J Clin Invest.
81. Trevisani F, Ghidini M, Larcher A, et al. MicroRNA 193b-3p as a 2009;119:1275–1285.
predictive biomarker of chronic kidney disease in patients undergoing 108. Marina M, Luca P, Cinzia R, et al. Sirtuin 3–dependent mitochondrial
radical nephrectomy for renal cell carcinoma. Br J Cancer. 2016;115: dynamic improvements protect against acute kidney injury. J Clin
1343–1350. Invest. 2015;125:715–726.
82. Zacchia M, Vilasi A, Capasso A, et al. Genomic and proteomic 109. Yang Z, Schumaker LM, Egorin MJ, et al. Cisplatin preferentially binds
approaches to renal cell carcinoma. J Nephrol. 2011;24:155–164. mitochondrial DNA and voltage-dependent anion channel protein in the
83. Cambier JF, Ronco P. Onco-nephrology: glomerular diseases with mitochondrial membrane of head and neck squamous cell carcinoma:
cancer. Clin J Am Soc Nephrol. 2012;7:1701–1712. possible role in apoptosis. Clin Cancer Res. 2006;12:5817–5825.

Kidney International (2019) 96, 555–567 565


meeting report A Capasso et al.: Onco-nephrology: an emerging field in medicine

110. Zhan M, Brooks C, Liu F, et al. Mitochondrial dynamics: regulatory 136. Berardi R, Antonuzzo A, Blasi L, et al. Practical issues for the
mechanisms and emerging role in renal pathophysiology. Kidney Int. management of hyponatremia in oncology. Endocrine. 2018;61:
2013;83:568–581. 158–164.
111. Ralto KM, Parikh SM. Mitochondria in acute kidney injury. Semin 137. Holland-Bill L, Christiansen CF, Farkas DK, et al. Diagnosis of
Nephrol. 2016;36:8–16. hyponatremia and increased risk of a subsequent cancer diagnosis:
112. Perico L, Morigi M, Benigni A. Mitochondrial sirtuin 3 and renal diseases. results from a nationwide population-based cohort study. Acta Oncol.
Nephron. 2016;134:14–19. 2018;57:522–527.
113. Morigi M, Perico L, Benigni A. Sirtuins in renal health and disease. J Am 138. Selmer C, Madsen JC, Toro-Pedersen C, et al. Hyponatremia, all-cause
Soc Nephrol. 2018;29:1799–1809. mortality, and risk of cancer diagnosis in the primary care setting: a
114. Morigi M, Perico L, Rota C, et al. Sirtuin 3-dependent mitochondrial large population study. Eur J Intern Med. 2016;36:36–43.
dynamic improvements protect against acute kidney injury. J Clin 139. Burst V, Grundmann F, Kubacki T, et al. Euvolemic hyponatremia in
Invest. 2015;125:715–726. cancer patients. Report of the Hyponatremia Registry: an observational
115. Perico L, Morigi M, Rota C, et al. Human mesenchymal stromal cells multicenter international study. Support Care Cancer. 2017;25:
transplanted into mice stimulate renal tubular cells and enhance 2275–2283.
mitochondrial function. Nat Commun. 2017;8:983. 140. Gralla RJ, Ahmad F, Blais JD, et al. Tolvaptan use in cancer patients with
116. Pillai VB, Samant S, Sundaresan NR, et al. Honokiol blocks and reverses hyponatremia due to the syndrome of inappropriate antidiuretic
cardiac hypertrophy in mice by activating mitochondrial Sirt3. Nat hormone: a post hoc analysis of the SALT-1 and SALT-2 trials. Cancer
Commun. 2015;6:6656. Med. 2017;6:723–729.
117. Li N, Xie H, Li L, et al. Effects of honokiol on sepsis-induced acute kidney 141. Beradi R, Santoni M, Newsom-Davis T, et al. Hyponatremia
injury in an experimental model of sepsis in rats. Inflammation. 2014;37: normalization as an independent prognostic factor in patients with
1191–1199. advanced non-small cell lung cancer treated with first-line therapy.
118. Li N, Zhang J, Yan X, et al. SIRT3-KLF15 signaling ameliorates kidney Oncotarget. 2017;8:23871–23879.
injury induced by hypertension. Oncotarget. 2017;8:39592–39604. 142. Khositseth S, Charngkaew K, Boonkrai C, et al. Hypercalcemia induces
119. Hershberger KA, Martin AS, Hirschey MD. Role of NADþ and targeted autophagic degradation of aquaporin-2 at the onset of
mitochondrial sirtuins in cardiac and renal diseases. Nat Rev Nephrol. nephrogenic diabetes insipidus. Kidney Int. 2017;91:1070–1087.
2017;13:213–225. 143. Salahudeen AK, Doshi SM, Shah P. The frequency, cost and clinical
120. Bonkowski MS, Sinclair DA. Slowing ageing by design: the rise of NADþ outcomes of hypernatremia in patients hospitalized to a
and sirtuin-activating compounds. Nat Rev Mol Cell Biol. 2016;17: comprehensive cancer center. Support Care Cancer. 2013;21:1871–1878.
679–690. 144. Gastanaga VM, Schwartzberg LS, Jain RK, et al. Prevalence of
121. Poyan Mehr A, Tran MT, Ralto KM, et al. De novo NADþ biosynthetic hypercalcemia among cancer patients in the United States. Cancer Med.
impairment in acute kidney injury in humans. Nat Med. 2018;24: 2016;5:2091–2100.
1351–1359. 145. Ramos REO, Perez Mak M, Alves MFS, et al. Malignancy-related
122. Hammers HJ, Plimack ER, Infante JR, et al. Immune-related adverse hypercalcemia in advanced solid tumors: survival outcomes. J Glob
events (irAEs) following CTLA-4 and PD-1/PD-L1 blockade in advanced Oncol. 2017;3:728–733.
melanoma: a comprehensive rapid autopsy study. Mod Pathol. 2016; 146. Zagouri F, Kastritis E, Zomas A, et al. Hypercalcemia remains an adverse
29:4A. prognostic factor for newly diagnosed multiple myeloma in the era of
123. Troxell ML, Higgins JP, Kambham N. Antineoplastic treatment and renal novel antimyeloma therapies. Eur J Hematol. 2017;99:409–414.
injury: an update on renal pathology due to cytotoxic and targeted 147. Rosner MH, Dalkin AC. Onco-nephrology: the pathophysiology and
therapies. Adv Anat Pathol. 2016;23:310–329. treatment of malignancy-associated hypercalcemia. Clin J Am Soc
124. Rosner MH, Capasso G, Perazella MA. Acute kidney injury and Nephrol. 2012;7:1722–1729.
electrolyte disorders in the critically ill patient with cancer. Curr Opin 148. Soki FN, Park SI, McCauley LK. The multifaceted actions of PTHrP in
Crit Care. 2017;23:475–483. skeletal metastasis. Future Oncol. 2012;8:803–817.
125. Doshi SM, Shah P, Lei X, et al. Hyponatremia in hospitalized cancer 149. Donovan PJ, Sundac L, Pretorius CJ, et al. Calcitriol-mediated
patients and its impact on clinical outcomes. Am J Kidney Dis. 2012;59: hypercalcemia: causes and course in 101 patients. J Clin Endocrinol
222–228. Metab. 2013;98:4023–4029.
126. Berghmans T, Paesmans M, Body JJ. A prospective study on 150. Wysolmerski JJ. Parathyroid hormone-related protein: an update. J Clin
hyponatraemia in medical cancer patients: epidemiology, aetiology and Endocrinol Metab. 2012;97:2947–2956.
differential diagnosis. Support Care Cancer. 2000;8:192–197. 151. O’Donnell EK, Raje NS. Myeloma bone disease: pathogenesis and
127. Gill G, Huda B, Boyd A, et al. Characteristics and mortality of severe treatment. Clin Adv Hematol Oncol. 2017;15:285–295.
hyponatraemia—a hospital-based study. Clin Endocrinol (Oxf). 2006;65: 152. Thosani S, Hu MI. Denosumab: a new agent in the management of
246–249. hypercalcemia of malignancy. Future Oncol. 2015;11:2865–2871.
128. Ingles Garces AH, Ang JE, Ameratunga M, et al. A study of 1088 153. Oronsky B, Caroen S, Oronsky A, et al. Electrolyte disorders with
consecutive cases of electrolyte abnormalities in oncology phase I trials. platinum-based chemotherapy: mechanisms, manifestations and
Eur J Cancer. 2018;104:32–38. management. Cancer Chemother Pharmacol. 2017;80:895–907.
129. Rosner MH, Dalkin AC. Electrolyte disorders associated with cancer. Adv 154. Evans JJ, Bozdech MJ. Hypokalemia in nonblastic chronic myelogenous
Chronic Kidney Dis. 2014;21:7–17. leukemia. Arch Intern Med. 1981;141:786.
130. Penttilä P, Bono P, Peltola K, et al. Hyponatremia associated with poor 155. Torpy DJ, Mullen N, Ilias I, et al. Association of hypertension and
outcome in metastatic renal cell carcinoma patients treated with hypokalemia with Cushing’s syndrome caused by ectopic ACTH
everolimus: prognostic impact. Acta Oncol. 2018;4:1–6. secretion: a series of 58 cases. Ann N Y Acad Sci. 2002;970:134.
131. Castillo JJ, Glezerman IG, Boklage SH, et al. The occurrence of 156. Zhang HY, Zhao J. Ectopic Cushing syndrome in small cell lung
hyponatremia and its importance as a prognostic factor in a cross- cancer: a case report and literature review. Thorac Cancer. 2017;8:
section of cancer patients. BMC Cancer. 2016;16:564. 114–117.
132. Mrowczynski OD, Bourcier AJ, Liao J, et al. The predictive potential of 157. Liamis G, Liberopoulos E, Barkas F, et al. Spurious electrolyte disorders:
hyponatremia for glioblastoma patient survival. J Neurooncol. 2018;138: a diagnostic challenge for clinicians. Am J Nephrol. 2013;38:50–57.
99–104. 158. Lameire N, Vanholder R, Van Biesen W, et al. Acute kidney injury in
133. Liamis G, Filippatos TD, Elisaf MS. Electrolyte disorders associated with critically ill cancer patients: an update. Crit Care. 2016;20:209.
the use of anticancer drugs. Eur J Pharmacol. 2016;777:78–87. 159. Criscuolo M, Fianchi L, Dragonetti G, et al. Tumor lysis syndrome: review
134. Beradi R, Santoni M, Rinaldi S, et al. Risk of hyponatremia in cancer of pathogenesis, risk factors and management of a medical emergency.
patients treated with targeted therapies: a systematic review and meta- Expert Rev Hematol. 2016;9:197–208.
analysis of clinical trials. PLoS One. 2016;11:e0152079. 160. Lacy MQ, Gertz MA. Acquired Fanconi’s syndrome associated with
135. Enzoe Y, Mizusawa J, Katayama H, et al. An integrated analysis of monoclonal gammopathies. Hematol Oncol Clin North Am. 1999;13:
hyponatremia in cancer patients receiving platinum-based or 1273–1280.
nonplatinum-based chemotherapy in clinical trials (JCOG1405-A). 161. Minisola S, Peacock M, Fukumoto S, et al. Tumor-induced osteomalacia.
Oncotarget. 2017;9:6595–6606. Nat Rev Dis Primers. 2017;13:17044.

566 Kidney International (2019) 96, 555–567


A Capasso et al.: Onco-nephrology: an emerging field in medicine meeting report

162. Larner AJ. Pseudohyperphosphatemia. Clin Biochem. 1985;28:391–393. 167. Velasco G, Sánchez C, Guzmán M. Endocannabinoids and cancer.
163. Cheminet G, Clain G, Jannot AS, et al. Extreme hypomagnesemia: Handb Exp Pharmacol. 2015;231:449–472.
characteristics of 119 consecutive inpatients. Intern Emerg Med. 2018;13: 168. Velasco G, Sánchez C, Guzmán M. Anticancer mechanisms of
1201–1209. cannabinoids. Curr Oncol. 2016;23:S23–S32.
164. Izzedine H, Perazella MA. Adverse kidney effects of epidermal 169. Petrosino S, Di Marzo V. FAAH and MAGL inhibitors: therapeutic
growth factor receptor inhibitors. Nephrol Dial Transplant. 2017;32: opportunities from regulating endocannabinoid levels. Curr Opin
1089–1097. Investig Drugs. 2010;11:51–62.
165. Davis MP. Oral nabilone capsules in the treatment of chemotherapy- 170. Hinz B, Ramer R. Anti-tumour actions of cannabinoids. Br J Pharmacol.
induced nausea and vomiting and pain. Expert Opin Investig Drugs. 2019;176:1384–1394.
2008;17:85–95. 171. Viggiano D, Capasso A, Capasso G. A quest for protecting kidneys from
166. Bifulco M, Di Marzo V. Targeting the endocannabinoid system in cancer cisplatin toxicity [e-pub ahead of print]. Nephrol Dial Transplant. https://
therapy: a call for further research. Nat Med. 2002;8:547–550. doi.org/10.1093/ndt/gfz029. Accessed February 27, 2019.

Kidney International (2019) 96, 555–567 567

You might also like