Você está na página 1de 122

Interconsulta

de Nefrologia

Programa (não-oficial) do R1 de Clinica Médica

Teoria Básica

1) Lesão Renal Aguda (texto + artigo)

2) Emergências Dialíticas (texto + artigo)

3) Síndromes Glomerulares (artigo)

4) Acesso Vascular Guiado por Ultrassom (artigo)

5) Diagnósticos Diferenciais e Manejo das Hipercalemias (artigo)

6) Diagnósticos Diferenciais e Manejo das Hipercalcemias (artigo)

7) Síndromes Cardiorrenais (artigo)

8) Teste de Estresse com Furosemida (artigo)

9) Gasometria Arterial (artigo)

10) Tipos de Acesso Vascular (artigo)

11) Princípios de Hemodiálise (artigo)

Lucas Theotonio
2

Teoria Básica
A idéia desse caderno não é de esgotar o assunto de forma alguma, mas sim dar um
norte para as discussões diárias.

1. Lesão Renal Aguda


a. Definição
Lesão renal aguda é definida pela KDIGO como:
• Aumento da Creatinina ≥ 0,3 mg/dL dentro de 48h; ou
• Aumento da Creatinina para ≥ 1,5 vezes a de base dentro de 7 dias; ou
• Débito urinário < 0,5 mL/kg/hora em um período de 6 horas.

Podemos ainda classificar a lesão renal aguda em 3 estágios diferentes,


progressivos em gravidade, conforme creatinina e débito urinário;

Estadio Creatinina Débito Urinário

1,5 a 1,9 x Creatinina de Base < 0,5 mL/kg/hora


1
Ou em 6 a 12 horas
Aumento ≥ 0,3 mg/dL
2,0 a 2,9 x Creatinina de Base < 0,5 mL/kg/hora
2
por mais de 12 horas
≥ 3 x Creatinina de Base < 0,3 mL/kg/hora
3
Ou por mais de 24 horas
Aumento da Creatinina para níveis ≥ 4,0 OU
mg/dL Anúria
Ou por mais de 12 horas
Necessidade de Terapia Renal Substitutiva

 Atualmente não utilizamos mais as antigas escalas AKIN e RIFLE para


classificar o quadro de lesão renal aguda.
3

b. Creatinina
A creatinina é um bom marcador de função renal pela sua simplicidade e
ampla disponibilidade, porém está longe de ser o método ideal de
avaliação, podendo sofrer interferência em qualquer fase da sua síntese,
como exposto abaixo:

Deste esquema podemos inferior que:


• Pacientes sarcopênicos (pouca musculatura), como idosos e
desnutridos, podem apresentar níveis falsamente baixos de creatinina;
• Pacientes cirróticos também podem apresentar níveis falsamente baixos
de creatinina pois há menor síntese da Creatina, sua precursora.

c. Classificação cronológica
A alteração aguda da função renal ainda pode ser classificada quanto a
sua evolução em:
• Lesão Renal Aguda – se durar até 7 dias entre o início do quadro e
sua completa resolução
• Doença Renal aguda – se o quadro durar de 7 a 89 dias até sua
resolução.
4

d. Investigação etiológica
Para fins didáticos, podemos dividir as causas de LRA em três
categorias, a saber:
• LRA pré-renal (desidratação/hipoperfusão)
• LRA pós-renal (obstrução de trato urinário, por exemplo HPB)
• LRA renal (glomerulopatias, nefrotóxicos, nefrite intersticial, etc)

Algumas dicas práticas para avaliação de cada uma das três


possibilidades:
• LRA pré-renal
o Avaliar grau de hidratação pelo exame de mucosas é válido,
porém pouco sensível para a real detecção de hipovolemia –
principalmente em idosos.
o Procure dados na história que possam te fazer pensar nesta
hipótese, como: diarréia, vômitos, exposição solar
prolongada, baixa ingesta alimentar/hídrica e uso de
diuréticos.
• LRA pós-renal
o Tanto homens quanto mulheres podem apresentar obstrução
do trato urinário, de diversas causas, as quais devem ser
devidamente excluídas com cateterismo vesical e exame de
imagem (a palpação de bexigoma é pouco sensível):
§ Obstrução baixa (uretra/bexiga): bexiga neurogênica,
hiperplasia prostática benigna, câncer de próstata,
câncer de reto, câncer de bexiga, câncer de colo
uterino, nefrolitíase bilateral, disfunção de assoalho
pélvico.
5

• LRA renal
o Glomerulopatias
Síndrome Achados Clínicos Patologias
Alterações 1. GESF, Membranosa
1. Proteinúria Isolada
Urinárias 2. IgA, Alport,
2. Hematúria Isolada
Assintomáticas GNMP,DMBF
*Proteinúria > 3,0-3,5g
Em 24 horas
*Hipoalbuminemia
*Edema GESF, Membranosa,
Nefrótica
Pode ter: Doença de Lesão Mínina
Hipercoagulabilidade
Infecções de repetição
Desnutrição
* Hematúria dismórfica
*Hipertensão
GNDA (pós-
*Disfunção Renal
estreptocócica), GNMP
Nefrítica *Edema
(Lupus, nefropatia por
*Proteinúria
IgA, vasculite por IgA)
usualmente não
nefrótica
Síndrome Nefrítica com
Três grandes grupos:
perda rápida de função
1. por Imunocomplexos
GNRP renal (dias a semanas),
2. Relacionadas ao ANCA
caracterizada por
3. Anti-MBG
crescentes glomerulares
Doença renal crônica Qualquer glomerulopatia
GNC secundária a que evolua para doença
glomerulopatias renal crônica
GESF: Glomeruloesclerose Segmentar e Focal
GNMP: Glomerulonefrite Membranoproliferativa
DMBF: Doença da Membrana Basal Fina
GNDA: Glomerulonefrite Difusa Aguda
GNRP: Glomerulonefrite Rapidamente Progressiva
GNC: Glomerulonefrite Crônica
 Síndromes Relacionadas a LRA: Nefrótica, Nefrítica e GNRP.
 Exames complementares: Urina I, Frações do Complement0 (C3,
C4, C2, CH50), ANCA, FAN, Biópsia Renal.
 Suspeitar de glomerulopatias em pacientes com suposta infecção
urinária de repetição com urocultura sempre negativa.
6

• LRA renal
o Nefrotóxicos
§ Diversos são os agentes nefrotóxicos. Em pacientes
internados, lembrar sempre dos principais
antimicrobianos com potencial nefrotóxico, a saber:
• Bactrim
• Polimixina
• Vancomicina
• Amicacina/Gentamicina
• Anfotericina B
• Aciclovir/Ganciclovir
• Piperacilina-Tazobactam (controverso)

o Nefrite Intersticial Aguda


§ Várias causas, dentre elas medicações e infecções;
§ Não há um período típicos de apresentação após a
exposição ao fator causal, podendo se apresentar de 3
a 5 dias após, ou até mesmo semanas a meses após;
§ A NIA relacionada a Rimfapicina, por exemplo, pode
se instalar em 24 horas;
§ Drogas: Beta-lactâmicos, Bactrim, Rimfampicina,
Furosemida, Ciprofloxacina, Alopurinol, IBPs
(Omeprazol por exemplo) e Claritromicina
§ Patógenos: Legionella spp., Leptospira spp., CMV e
Streptococcus spp.
§ Tríade clássica: rash cutâneo, febre e eosinofilia
§ Outros sintomas possíveis: dor lombar bilateral,
hematuria microscópica (podendo ser confundida com
nefrolitíase no pronto-socorro).
7

2. Emergências Dialíticas

a. Princípios Gerais
• A hemodiálise de urgência é sempre do tipo intermitente (duração usual de
4 a 6 horas).
• Usualmente, não utilizamos a diálise peritoneal para tratamento de
emergências dialíticas.
• Sempre que pensar em caracterizar uma situação como emergência
dialítica, sempre tenha em mente a refratariedade a outros tratamentos
menos invasivos (por exemplo, hipercalemia refratária a furosemida, ou
hipercalemia em paciente anúrico)

b. As Emergências (AEIOU)

A (Acidose Metabólica): usualmente acidose metabólica não relacionada a cetonemia ou


hiperlactatemia.

• Usualmente consideramos emergência se Bic < 15 ou pH < 7,15, após outras medidas)
E (Eletrólitos): K+ > 5,0-5,5 mEq/L com repercussão no ECG ou K+ ≥ 6,5 refratário a medidas
clínicas que reduzam o pool (diuréticos) ou redistribuam definitivamente (corrigir acidose) o K+.

• Glico-insulina, B2 agonista e Gluconato de Cálcio são medidas temporárias.


I (Intoxicações exógenas): alguns drogas são dialisáveis, como por exemplo Lítio, Atenolol,
Metanol, Metformina, Acido Valpróico, Carbamazepina e Paracetamol.

• Os critérios para indicar TRS para cada uma das intoxicações fogem ao escopo desse guia.
As drogas em negrito são as que tem maior indicação.
O (Overload): sobrecarga de volume, principalmente edema agudo de pulmão ou IC
descompensada refratários a diuréticoterapia, ou com boa resposta porém com efeitos colaterais
importantes dos diuréticos (citados abaixo).

• Hiper/Hiponatremia, Hipocalemia, Alcalose Metabólica com BIC > 35 mEq/L


U (Uremia): coma urêmico, sangramento urêmico (TGI), pericardite/pleurite urêmica,
convulsão secundária a uremia

• Crise usualmente do tipo mioclônica – excluir outras causas como sangramento de SNC
ARTIGO 1

LESÃO RENAL AGUDA


Annals of Internal Medicine!

In the Clinic®

Acute Kidney Injury


A
cute kidney injury is a heterogeneous group
Screening and Prevention
of conditions characterized by a sudden de-
crease in glomerular filtration rate, mani-
fested by an increase in serum creatinine concentra-
tion or oliguria, and classified by stage and cause. Diagnosis
This type of injury occurs in approximately 20% of
hospitalized patients, with major complications in-
cluding volume overload, electrolyte disorders, ure- Treatment
mic complications, and drug toxicity. Management
includes specific treatments according to the under-
lying cause and supportive treatment to prevent and
manage complications. Kidney replacement therapy
is used when complications cannot be managed
with medical therapy alone. Despite advances in
care, the mortality rate in patients requiring kidney
replacement therapy remains approximately 50%.

CME/MOC activity available at Annals.org.

Physician Writers doi:10.7326/AITC201711070


Andrew S. Levey, MD
Matthew T. James, MD CME Objective: To review current evidence for screening, prevention, diagnosis, and
From Tufts Medical Center, treatment of acute kidney injury.
Boston, Massachusetts; and Funding Source: American College of Physicians.
the University of Calgary,
Calgary, Alberta. Disclosures: Dr. Levey, ACP Contributing Author, reports that he was a member of the KDIGO
Workgroup for the Clinical Practice Guideline on Acute Kidney Injury (published 2012). Dr.
James, ACP Contributing Author, reports grants from Amgen Canada outside the submitted
work. Disclosures can also be viewed at www.acponline.org/authors/icmje
/ConflictOfInterestForms.do?msNum=M17-2010.
Acknowledgment: The authors thank Sara Couture, BS, and Bryan Ma for assistance with
manuscript preparation.
With the assistance of additional physician writers, the editors of Annals of Internal Medi-
cine develop In the Clinic using MKSAP and other resources of the American College of
Physicians.
In the Clinic does not necessarily represent official ACP clinical policy. For ACP clinical
guidelines, please go to https://www.acponline.org/clinical_information/guidelines/.

© 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


Acute kidney inj ry (AKI) is not a mately 50%. AKI may occur in pa-
single disease entity. It is a hetero- tients with or without underlying
geneous group of conditions char- chronic kidney disease (CKD). In-
acterized by a sudden decrease in complete recovery may lead to
glomerular filtration rate (GFR) fol- new onset or worsening of CKD.
lowed by an increase in serum cre- Evaluation and management of
1. Kellum JA, Lameire N, atinine concentration (SCC) or oli- AKI proceed in parallel rather than
Aspelin P. Kidney Disease:
Improving Global Out-
guria. AKI generally occurs in the sequentially. The goals are to ap-
comes (KDIGO) Acute setting of acute or chronic illness. It ply specific treatments according
Kidney Injury Work Group.
KDIGO clinical practice affects approximately 20% of hos- to the underlying cause and pro-
guideline for acute kidney pitalized patients, of whom 10% vide supportive care to prevent
injury. Kidney Interna-
tional Supplement 1s. require kidney replacement ther- and treat complications. KRT is
2012;2:1-138.
2. Mehta R, Bagga A, Pati-
apy (KRT). Recent clinical guide- used when complications of kid-
bandla R, Chakravarthi R. lines from Kidney Disease Improv- ney failure cannot be managed
Detection and manage-
ment of AKI in the devel- ing Global Outcomes (KDIGO) with medical therapy alone. The
oping world: The 18th define AKI as a subgroup of acute need for nephrologist consultation
Acute Disease Quality
Initiative (ADQI) Interna- kidney diseases and disorders or comanagement depends on
tional Consensus Confer-
ence. Kidney International
(AKD), and classify AKI according the stage, cause, and severity of
Reports. 2017;2:515-8. to severity (stages) and cause, complications. This review focuses
3. Susantitaphong P, Cruz
DN, Cerda J, et al; Acute which influence prognosis and on general features of AKI in adults
Kidney Injury Advisory management (1). Major complica- in developed countries, not includ-
Group of the American
Society of Nephrology. tions include volume overload, ing pregnant women or kidney
World incidence of AKI: a
meta-analysis. Clin J Am
electrolyte disorders, uremic com- transplant recipients. AKI is a more
Soc Nephrol. 2013;8: plications, and drug toxicity. De- serious problem in developing
1482-93. [PMID:
23744003] spite advances in prevention and countries due to risk factors re-
4. Wonnacott A, Meran S, treatment, the mortality in patients lated to underdevelopment and
Amphlett B, Talabani B,
Phillips A. Epidemiology requiring KRT remains approxi- lack of availability of KRT (2).
and outcomes in
community-acquired ver-
sus hospital-acquired AKI.
Clin J Am Soc Nephrol.
2014;9:1007-14. [PMID: Screening and Prevention
24677557]
5. Wang Y, Wang J, Su T, Which patients are at increased required dialysis. Rates were substantially higher
et al; ISN AKF 0by25 risk for AKI, and how should for patients with CKD. Decreased GFR and pro-
China Consortium.
Community-acquired clinicians identify them? teinuria were independent risk factors. Adjusted
acute kidney injury: a rate ratios were 2.5– 4.4 for patients with GFR
nationwide survey in AKI generally occurs in the set-
China. Am J Kidney Dis. >60 mL/min/1.73 m2 (CKD stages 1–2); 2.3– 8.2
2017;69:647-657. [PMID:
ting of acute and chronic illness, for GFR 45–59 mL/min/1.73 m2 (CKD stage 3a);
28117208] and is common among hospital- 5.6 –13 for GFR 30-44 mL/min/1.73 m2 (CKD
6. James MT, Hemmelgarn
BR, Wiebe N, et al; Alberta ized patients. stage 3b); and 13–19 for GFR 15–29 mL/min/
Kidney Disease Network.
1.73 m2 (CKD stage 4).
Glomerular filtration rate, A systematic review of large cohort studies, pri-
proteinuria, and the inci-
dence and consequences marily among hospitalized patients, con- The Box shows risk factors for AKI.
of acute kidney injury: a ducted between 2004 and 2012, showed sig-
cohort study. Lancet. Acute illness, complications of medi-
2010;376:2096-103. nificant heterogeneity in estimates among
[PMID: 21094997] studies, countries, and clinical settings, and
cations, and medical procedures are
7. Tsai TT, Patel UD, Chang
between adults and children (3). In studies of the most common. Older age and
TI, et al. Validated contem-
porary risk model of acute adults that identified AKI according to the preexisting CKD are the main sus-
kidney injury in patients
undergoing percutaneous KDIGO criteria and staging system, the pooled ceptibility factors. Risk prediction
coronary interventions: incidence rate of AKI was 21.6% (95% CI, instruments are available for some
insights from the National
Cardiovascular Data Regis- 19.3%–24.1%). Approximately 10% required high-risk settings, such as percuta-
try Cath-PCI Registry. J Am dialysis. The highest pooled AKI rate was ob- neous coronary intervention, cardiac
Heart Assoc. 2014;3:
e001380. [PMID: served in critical care settings (32%). In other surgery, liver surgery, and vascular
25516439] studies, among hospitalized patients approxi-
8. Allen DW, Ma B, Leung surgery (7–11).
KC, et al. Risk prediction
mately two thirds of AKI episodes were com-
models for contrast- munity-acquired—the remaining one third AKI is generally asymptomatic, so
induced acute kidney
injury accompanying
were hospital-acquired (4, 5). screening is usually required for
cardiac catheterization:
systematic review and In 1 population-based study (6), the rate of adults detection. The U.S. Preventive Ser-
meta-analysis. Can J Car-
diol. 2017;33:724-736.
with AKI but not CKD who required hospital ad- vices Task Force does not have
[PMID: 28545621] mission was 0.1% per year. Approximately 10% recommendations regarding

" 2017 American College of Physicians ITC66 In the Clinic Annals of Internal Medicine 7 November 2017

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


screening for AKI. KDIGO recom- drugs in patients with CKD, such
mends screening based on stratifi- as nonsteroidal anti-inflammatory Risk Factors for Acute Kidney
cation by risk according to expo- drugs (NSAIDs) and iodinated ra- Injury
sures and susceptibility. Because diocontrast media (contrast-
Exposures
there are few risk prediction instru- induced AKI) (15–17).
ments, we suggest the following Critical illness
approach guided by the clinical Risk for contrast-induced AKI Sepsis
setting. For outpatients with acute seems to be greater after arterial Circulatory shock
illness, measure SCC and calculate than venous administration of
Burns
estimated GFR (eGFR) and com- contrast media. For patients at
increased risk for this disorder, Trauma
pare to previous (“baseline”) val-
ues (12). Remeasure if SCC or KDIGO recommends using either Cardiac surgery (especially
eGFR are abnormal or worse than an iso-osmolal or a low-osmolal with cardiopulmonary bypass)
previous values. The urgency of medium (osmolality 2–3 times Major noncardiac surgery
repeated measurement depends that of plasma) rather than a
Nephrotoxic drugs
on the severity of illness and the high-osmolal contrast medium
(osmolality >4 times that of Iodinated radiocontrast
level of SCC and eGFR; hospital-
agents
ization should be considered. For plasma). IV volume expansion
hospitalized patients, SCC and with either an isotonic sodium chlo- Poisonous plants and animals
eGFR are generally measured on ride or a sodium bicarbonate solu- Susceptibility factors
admission and should be mea- tion should be done rather than no
Volume depletion
sured daily or every other day. For IV volume expansion. Several proto-
cols are available for intra-arterial Older age
patients with critical illness, SCC
and eGFR, as well as urine output, contrast administration, and these Female sex
should be measured at least daily. can be used for high-risk patients Black race
In our experience, accurate mea- receiving IV contrast administration Chronic kidney disease
surement of urine output is diffi- (18–22). Tailoring administration to
Other chronic diseases
cult, unless the patient is in an left ventricular filling pressure can
(heart, lung, liver)
intensive care unit (ICU). In all set- safely facilitate more volume expan-
tings, urinalysis should be done for sion and reduce the incidence of Diabetes mellitus
detection of AKD and CKD. contrast-induced AKI during cardiac Cancer
catheterization (23, 24). A recent Anemia
Increased SCC and oliguria may comparative effectiveness review From reference 1.
not occur for several hours after compares other strategies for pre-
the onset of an acute decline in venting this complication (25, 26).
GFR. Novel biomarkers are under
investigation to determine We also recommend volume ex-
whether they may enable earlier pansion with isotonic sodium chlo-
detection of decreased GFR and ride for other high-risk conditions,
complications of AKI (13, 14). such as cardiac surgery; hemolysis;
rhabdomyolysis; tumor lysis; and
Which measures are useful for
administration of cisplatinum, car-
preventing AKI, and when boplatin, ifosphamide, and am-
9. Wijeysundera DN, Kark-
outi K, Dupuis JY, et al.
should they be used? photericin B. Caution is warranted Derivation and validation
of a simplified predictive
We recommend general measures in patients with volume overload, index for renal replace-
to reduce exposures and suscepti- and IV fluids should be discontin- ment therapy after cardiac
surgery. JAMA. 2007;297:
bility when possible—for example, ued if symptoms of volume over- 1801-9. [PMID:
correcting volume depletion by load develop (see below).
17456822]
10. Wilson T, Quan S,
increasing oral salt and fluid intake Cheema K, et al. Risk
or intravenous (IV) isotonic saline. Monitoring therapeutic levels of prediction models for
acute kidney injury fol-
Other examples are avoiding di- nephrotoxic drugs, such as van- lowing major noncardiac
surgery: systematic re-
uretics and angiotensin-converting comycin, aminoglycosides, and view. Nephrol Dial Trans-
enzyme inhibitors (ACEIs) and calcineurin inhibitors, can reduce plant. 2016;31:231-40.
[PMID: 26705194]
angiotensin-receptor blockers risk for AKI. KDIGO suggests ad- 11. Borthwick E, Ferguson A.
(ARBs) during acute illness to pre- ditional measures to reduce the Perioperative acute kid-
ney injury: risk factors,
vent volume depletion and hypo- risk for nephrotoxicity of amino- recognition, manage-
ment, and outcomes.
tension, and avoiding nephrotoxic glycosides and amphotericin B. BMJ. 2010;341:c3365.
[PMID: 20603317]

7 November 2017 Annals of Internal Medicine In the Clinic ITC67 " 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


Screening and Prevention... AKI generally occurs in the setting of
acute and chronic illness and is common among hospitalized patients.
Older age and CKD are the main susceptibility factors. Measurement of
SCC and eGFR and monitoring during hospitalization are essential to
detect AKI. Urine output should be monitored in patients with critical
illness. Urinalysis is helpful to detect AKD and CKD. General measures
to reduce risk include prevention and treatment of volume depletion
and avoidance of nephrotoxic drugs. IV isotonic fluids before, during,
and after intra-arterial administration of iodinated radiocontrast media
can reduce risk for contrast-induced AKI.

CLINICAL BOTTOM LINE

Diagnosis
What criteria should clinicians tion based on severity (stages)
use to define and classify AKI? and cause (Figure 2 ). KDIGO
AKI is a heterogeneous group of definition and staging are
conditions, with a common defi- based on the Risk, Injury, Fail-
nition (Figure 1 ) and classifica- ure, Loss, End-Stage Renal Dis-
ease (RIFLE) and AKI Network
(AKIN) criteria and studies on
Figure 1. Relationships and definitions of kidney diseases and disorders. risk relationships. The rationale
for defining AKI separately from
other acute kidney diseases and
disorders was to provide a
more rigorous basis for re-
search studies, clinical practice
guidelines, and public health
efforts.
AKD AKI CKD
The KDIGO definition of AKI in-
cludes a change in SCC within
2–7 days and oliguria for 6 or
more hours. The stage is defined
by the peak rise in SCC com-
pared with previous values and
nadir in urine output and is re-
Variable AKI AKD CKD NKD*
Duration Within 7 d ≤3 mo >3 mo lated to risk for complications
Functional Increase in serum AKI GFR <60 GFR ≥60 and prognosis (Figure 2 ). eGFR
criteria creatinine clearance OR mL/min/1.73 m2 mL/min/1.73 m2 is preferred to SCC for assessing
by ≥50% within 7 d GFR <60
OR mL/min/1.73 m 2 GFR in the steady state (i.e.,
Increase in serum OR when GFR is stable) because the
creatinine clearance Decrease in GFR by
by ≥0.3 mg/dL ≥35% times baseline
coefficients for age, sex, and race
within 2 d OR in the estimating equation take
OR Increase in serum into account variation in creati-
Oliguria for ≥6 h creatinine clearance by
≥50% times baseline nine generation by muscle, inde-
Structural Not defined Marker of kidney Marker of kidney No marker of pendent of GFR (12). A 1.5-, 2.0-,
criteria damage damage for >3 mo kidney damage and 3.0-fold increase in SCC dur-
(albuminuria, (albuminuria is most
hematuria, or pyuria common)
ing steady-state conditions re-
is most common) flect a 39%, 57%, and 74% de-
crease in eGFR, respectively.
AKI = acute kidney injury; AKD = acute kidney diseases and disorders; CKD = chronic kidney However, during AKI, SCC may
disease; GFR = glomerular filtration rate; NKD = no kidney disease. be in the nonsteady state, so
* Implies no functional or structural criteria according to the definitions for AKI, AKD, or CKD.
Clinical judgment required for individual patient decision making. changes in SCC and eGFR lag

" 2017 American College of Physicians ITC68 In the Clinic Annals of Internal Medicine 7 November 2017

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


Figure 2. Overview of acute kidney injury.

At Risk Complications
Older age, comorbid conditions, CKD (decreased GFR, albuminuria) Volume overload
Electrolyte disorders
Stage 1 (hyperkalemia, metabolic acidosis,
Serum creatinine: 1.5–1.9 times baseline, or ≥0.3 mg/dL increase, or hyponatremia and hypernatremia,
KDIGO Staging and

urine output: <0.5 mL/kg/h for 6–12 h hypocalcemia and hypercalcemia,


Complications

hyperphosphatemia, hypermagnesemia)
Stage 2
Uremic complications
Serum creatinine: 2.0–2.9 times baseline, or urine output: <0.5 mL/kg/h for ≥12 h
(encephalopathy, pericarditis, pruritus, bleeding)
Stage 3 Drug toxicity
Initiation or renal replacement therapy, or serum creatinine: 3.0 times baseline, or
≥4.0 mg/dL, or urine output: <0.3 mL/kg/h for ≥24 h, or anuria for ≥12 h

Decreased Kidney Obstruction of the Urinary Parenchymal Kidney ATN


Perfusion Tract Diseases Other Than ATN
Causes
Causes Causes Causes Toxic ATN; ischemic ATN
Volume depletion; heart, Obstructive nephropathy Acute glomerulonephritis;
Causes, Clinical Setting and

lung, or liver disease; acute interstitial nephritis; Clinical Setting


Urinary Tract Findings

sepsis; increased intra- Clinical Setting pyelonephritis; thrombotic Circulatory shock; sepsis;
abdominal pressure; renal Urinary tract symptoms; microangiopathy; cast neph- drug exposure; transient
artery stenosis; NSAID history of urolithiasis, ropathy; infarction; hypotension; hemolysis;
toxicity genitourinary tract neoplasia, atheroembolism rhabdomyolysis; tumor lysis
or retroperitoneal disease
Clinical Setting Clinical Setting Urinary Tract Findings
Signs of volume depletion Urinary Tract Findings Systemic diseases; Urine not concentrated
or overload; SIRS; severe Hydronephrosis; relief with microangiopathic hemolysis urine; RTE cells; granular
hypertension urinary catheter casts
Urinary Tract Findings
Urinary Tract Findings Hematuria with RBC casts;
Concentrated urine; no pyuria with WBC casts; RTE
RTE cells or casts cells

Prevention Strategies Nephrologist


Intravenous fluid volume expansion, nephrotoxic medication avoidance, minimization Consultation and Comanagement
of radiocontrast media, hemodynamic monitoring and management Uncertainty about diagnosis
Management

Uncertainty about cause


Early-Stage Management Treatment of parenchymal diseases
Specific therapies and interventions that are dependent on the cause of acute kidney injury. Need for kidney replacement therapy
Intravenous fluid volume resuscitation, vasopressors, therapeutic drug level monitoring,
and adjustment of medication dosing Kidney Replacement Therapy
Volume overload, electrolyte disorders, uremic
Late-Stage Management complications (refractory to medical management)
Supportive care (maintenance of nutrition, electrolyte, and volume balance) and
assessment for complications requiring kidney replacement therapy

ATN = acute tubular necrosis; CKD = chronic kidney disease; GFR = glomerular filtration rate; NSAID = nonsteroidal anti-inflammatory drug;
RBC = red blood cell; RTE = renal tubular epithelial; SIRS = systemic inflammatory response syndrome; WBC = white blood cell.

12. Levey AS, Becker C, Inker


LA. Glomerular filtration
behind changes in GFR, and than 1 cause. Identifying causes rate and albuminuria for
detection and staging of
eGFR may be a less accurate esti- other than ATN is important be- acute and chronic kidney
mate of measured GFR. None- cause specific treatment of other disease in adults: a sys-
tematic review. JAMA.
theless, reporting eGFR in AKI causes can reverse the decline in 2015;313:837-46.
may be useful because changes GFR, whereas treatment for ATN [PMID: 25710660]
13. Kimmel M, Shi J, Latus J,
in eGFR show the direction and is supportive. The Box discusses et al. Association of renal
stress/damage and filtra-
estimate the magnitude of features that distinguish de- tion biomarkers with
changes in GFR. creased kidney perfusion from subsequent AKI during
hospitalization among
ATN. patients presenting to
The causes of AKI, which are cat- the emergency depart-
egorized according to underlying Complications of AKI result from ment. Clin J Am Soc
Nephrol. 2016;11:938-
pathophysiology, are decreased impaired excretory, endocrine, 46. [PMID: 27026519]
14. Perazella MA, Coca SG.
kidney perfusion, obstruction of and metabolic kidney functions. Traditional urinary bio-
the urinary tract, parenchymal Decreased GFR and tubular func- markers in the assess-
ment of hospital-
kidney diseases other than acute tion lead to retained water and acquired AKI. Clin J Am
tubular necrosis (ATN), and ATN solutes, manifested by volume Soc Nephrol. 2012;7:
167-74. [PMID:
(Figure 2 ). AKI often has more overload, hyperkalemia, high an- 22096038]

7 November 2017 Annals of Internal Medicine In the Clinic ITC69 " 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


ion gap metabolic acidosis, hy- were due to decreased kidney perfusion, 12%
Features Distinguishing ponatremia, hyperphosphatemia, were due to obstruction, 27% had “intrinsic
Decreased Kidney Perfusion hypermagnesemia, encephalop- kidney diseases” (parenchymal diseases, in-
From ATN athy, pericarditis, pruritus, and cluding ATN), and 12% were not classified (5).
Causes of hospital-acquired AKI in patients
Extracellular fluid volume depletion bleeding due to platelet dysfunc-
or circulatory disorders were described in patients admitted to general
tion. Deficiencies of erythropoie- medical or surgical services of an urban teach-
associated with volume
expansion are common in
tin and decreased synthesis of ing hospital. Of 380 episodes, 39% were due
decreased kidney perfusion, active vitamin D lead to anemia to decreased kidney perfusion, 2% to obstruc-
whereas recent exposure to and hypocalcemia. During recov- tion, 3% to parenchymal kidney diseases other
radiographic contrast or ery from AKI, persistent impair- than ATN, and 55% to causes associated with
nephrotoxic drugs or ATN; 3% were not classified (27).
ment in tubular function despite
hypotension is more prominent
in ATN. increasing GFR may give rise to
Decreased kidney perfusion de-
Decreased kidney perfusion excessive water and solute loss,
creases GFR and increases tubu-
improves rapidly after leading to volume depletion, hy-
lar reabsorption of sodium, chlo-
measures to increase kidney pernatremia, hypokalemia, non–
perfusion (e.g., intravenous ride, urea, and water, leading to
anion gap metabolic acidosis,
fluid in volume depletion), but urine concentration. Volume de-
no such response is observed
hypophosphatemia, hypomag- pletion as the cause of decreased
in ATN. nesemia, and hypercalcemia in kidney perfusion should be con-
Decreased kidney perfusion usually some cases. Drug toxicity is com- sidered in patients with a history
has concentrated urine and no mon because of altered pharma- of decreased oral intake, vomit-
RTE cells or granular casts in cokinetics and pharmacodynam-
urine; ATN usually has ing, diarrhea, increased ostomy
ics. Complications may occur in output, excessive sweating, per-
nonconcentrated urine, RTE
cells, and granular casts. other organ systems throughout cutaneous fluid drainage, bleed-
ATN = acute tubular necrosis; the course of disease; multiple ing, dizziness, light-headedness,
RTE = renal tubular epithelial. organ failure is associated with fainting, or recent weight loss,
the highest mortality. especially in the setting of dietary
What clinical manifestations
15. Huerta C, Castellsague J, should clinicians look for?
Varas-Lorenzo C, Garcı́a
Rodrı́guez LA. Nonsteroi- AKI is generally asymptomatic
Combined Diagnostic and
dal anti-inflammatory until the onset of kidney failure,
drugs and risk of ARF in Therapeutic Approach to AKI
the general population. emphasizing the importance of
Am J Kidney Dis. 2005; Assess volume status; administer
45:531-9. [PMID: monitoring SCC and eGFR and intravenous fluid if volume
15754275] urine output in patients at in- depleted or volume status is
16. Lapi F, Azoulay L, Yin H,
Nessim SJ, Suissa S. creased risk. However, clinical uncertain
Concurrent use of diuret-
ics, angiotensin convert-
manifestations are useful to de- Assess for indications for urgent
ing enzyme inhibitors, termine the cause of AKI and de- kidney replacement therapy
and angiotensin receptor (volume overload, uremic
blockers with non- tect its complications. After de-
complications, electrolyte
steroidal anti-
inflammatory drugs and
tection, whether rapid evaluation disorders, drug toxicity)
risk of acute kidney in- is necessary depends on the clin- Conduct urine studies
jury: nested case-control
study. BMJ. 2013;346: ical setting and severity of AKI. Consider additional laboratory
e8525. [PMID: We recommend a combined di- tests and imaging
23299844]
17. Dreischulte T, Morales agnostic and therapeutic ap- In all cases, thoroughly review the
DR, Bell S, Guthrie B.
proach (see the Box). If AKI is history of present illness,
Combined use of non-
medical history, medications
steroidal anti- severe, treatment must begin and exposure to radiocontrast
inflammatory drugs with
diuretics and/or renin- concurrently with evaluation. media, recent surgery and
angiotensin system in- other procedures, recent travel
hibitors in the commu- Evaluating Causes
nity increases the risk of or exposure to infectious
acute kidney injury. Decreased kidney perfusion is diseases, fluid intake and
Kidney Int. 2015;88: output, and laboratory tests and
396-403. [PMID:
the most common cause of
imaging and conduct a careful
25874600] community-acquired AKI, whereas
18. Trivedi HS, Moore H, physical examination. In
Nasr S, et al. A random- ATN is the most common cause of particular, assessment for
ized prospective trial to
assess the role of saline
hospital-acquired AKI. complications should be done
hydration on the devel- promptly to determine the
opment of contrast neph- Causes of community-acquired AKI were de- need for urgent therapy.
rotoxicity. Nephron Clin
Pract. 2003;93:C29-34.
scribed in a large survey of hospital discharges AKI = acute kidney injury.
[PMID: 12411756] in China. Of 4136 patients, 49% of AKI cases

" 2017 American College of Physicians ITC70 In the Clinic Annals of Internal Medicine 7 November 2017

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


salt restriction or diuretic therapy. gests lesions in the urinary tract.
Increased GFR within hours of Complete obstruction causes Drugs That Contribute to
volume repletion with oral or IV anuria, but partial obstruction may Acute Kidney Injury and the
fluids confirms the diagnosis. De- cause polyuria due to impairment Pathophysiologic Mechanism
creased kidney perfusion due to of tubular function. Immediate im- of Injury*
exacerbations of chronic heart, provement in GFR is expected af- Iodinated radiocontrast media
(acute tubular necrosis)
lung, or liver disease manifests as ter relief of acute obstruction, but
Nonsteroidal anti-inflammatory
weight gain and edema (car- improvement after relief of chronic
drugs (decreased kidney
diorenal and hepatorenal syn- obstruction may be slow or perfusion and tubular
dromes), often with low blood incomplete. toxicity)
pressure. Sepsis may also be as- Aminoglycosides (acute tubular
Parenchymal kidney diseases necrosis)
sociated with low blood pressure
other than ATN are frequent Amphotericin B (acute tubular
and other features of the sys-
causes of AKI. Many of these necrosis)
temic inflammatory response
diseases occur in the setting of !-Lactam antibiotics (interstitial
syndrome. Intra-abdominal pres-
systemic disorders (infections; nephritis)
sure may increase in critically ill
vasculitis; inflammatory, my- Sulfonamides (interstitial
patients with abdominal or pelvic nephrtitis)
eloproliferative, or lymphoprolif-
disorders (trauma, hemoperito- Vancomycin (acute tubular
erative diseases; or drug toxicity)
neum, pancreatitis, surgery, ra- necrosis)
and may be associated with fe-
diologic procedures) or in condi- Acyclovir (crystal nephropathy)
ver, night sweats, arthralgias,
tions that do not originate in that Tenofovir (tubular toxicity)
mononeuropathy, or skin rash.
region (fluid resuscitation, sepsis, Glomerulonephritis may cause Methotrexate (crystal
burns). Bilateral renal artery ste- nephropathy)
gross hematuria (without clots).
nosis may occur in patients with Cisplatinum and carboplatin
Acute interstitial nephritis is gen- (acute tubular necrosis)
aortic aneurysm or diffuse ath- erally due to an allergic or toxic Ifosphamide (acute tubular
erosclerotic cardiovascular dis- drug reaction (see the Box). Bac- necrosis)
ease and causes severe hyper- terial pyelonephritis must be se- Vascular endothelial growth factor
tension. Use of NSAIDs can vere and bilateral to cause AKI inhibitors (thrombotic
decrease kidney perfusion, even and often causes flank pain with microangiopathy)
without volume or blood pres- high fever. Urinary symptoms are Calcineurin inhibitors (e.g.,
sure abnormalities. ACEIs and cyclosporine, tacrolimus)
prominent when pyelonephritis is (decreased kidney perfusion
ARBs reduce GFR in patients with caused by ascending infection and tubular toxicity)
acute and chronic kidney disease from the lower urinary tract but Herbal and dietary supplements
and may cause severe AKI in the may be absent in the presence of (e.g., aristolochic acid;
setting of decreased kidney hematogeneous dissemination of creatine; vitamins A, C, and
perfusion. bacteremia. Thrombotic microan- D; germanium; star fruit)
(interstitial nephritis)
giopathy is accompanied by mir-
Obstruction of both kidneys or Proton-pump inhibitors (interstitial
coangiopathic hemolysis and
obstruction of a solitary kidney nephritis)
thrombocytopenia with schisto-
may cause AKI. Obstruction may Angiotensin-converting enzyme
cytes. Cast nephropathy in my- inhibitors, angiotensin-receptor
be acute or chronic, complete or
eloma is typically associated with blockers, direct renin inhibitors
partial, due to upper or lower (decreased kidney perfusion)
a high tumor burden and large
urinary tract disease, and due to * Many drugs have several
amounts of paraprotein in the
lesions within or outside the uri- pathyphysiologic
urine and frequently with hyper-
nary tract. Flank pain or a history mechanisms.
calcemia. Renal infarction is asso-
of urolithiasis, genitourinary tract
ciated with flank pain and abrupt
neoplasia, or retroperitoneal dis-
onset of severe hypertension.
ease should raise suspicion for
Atheroembolism may occur af- 19. Taylor AJ, Hotchkiss D,
obstruction (28). Symptoms of
ter percutaneous intra-arterial Morse RW, McCabe J.
lower urinary tract disease in- procedures or surgery involving
PREPARED: Preparation
for Angiography in Renal
clude dysuria, suprapubic pain, the aorta or initiation of Dysfunction: a random-
ized trial of inpatient vs
slow urine stream, and increased anticoagulation. outpatient hydration
frequency of urination. Bladder protocols for cardiac
catheterization in mild-
distention may be detectable by The pathology of ATN is charac- to-moderate renal dys-
physical examination. Gross he- terized by necrosis of tubular function. Chest. 1998;
114:1570-4. [PMID:
maturia (with blood clots) sug- epithelial cells diffusely or local- 9872190]

7 November 2017 Annals of Internal Medicine In the Clinic ITC71 " 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


ized to nephron segments, with- of intermittently administered
out involvement of the glomeruli, medication.
20. Mueller C, Buerkle G,
Buettner HJ, et al. Pre-
accompanied by variable intersti-
Evaluating Complications
vention of contrast tial inflammation. However, clini-
media-associated ne- Irrespective of cause, the stage
phropathy: randomized cal pathologic correlations are
(severity) of AKI is related to the
comparison of 2 hydra- imprecise, and renal biopsy is
tion regimens in 1620 risk for complications.
patients undergoing rarely done in AKI. As a result,
coronary angioplasty.
Arch Intern Med. 2002; the diagnosis of ATN in AKI is The frequency of AKI-related complications was
162:329-36. [PMID: generally presumed in patients studied in 18 410 patients in ICUs in 4 hospitals
11822926]
21. Krasuski RA, Beard BM, with a typical clinical history and (29). The risk for volume overload increased
Geoghagan JD, Thomp-
urinary tract findings (as de- from 41% in patients without AKI to 58% for
son CM, Guidera SA.
Optimal timing of hydra- scribed below) after exclusion of stage 1, 77% for stage 2, and 83% for stage
tion to erase contrast-
other causes of AKI. Despite this 3 AKI. Other comparisons found respective
associated nephropathy:
increases in hyperkalemia from 3% to 9%,
the OTHER CAN study. J uncertainty, we will retain use of
Invasive Cardiol. 2003; 17%, and 32%; in metabolic acidosis from
15:699-702. [PMID: the term ATN as a cause of AKI 59% to 74%, 86%, and 91%; in hyponatre-
14660821]
22. Bader BD, Berger ED, because it remains a useful clini- mia from 19% to 30%, 46%, and 60%; and
Heede MB, et al. What is cal concept in diagnosis and azotemia (blood urea nitrogen >60 mg/dL)
the best hydration regi-
men to prevent contrast treatment. Circulatory shock and from 1% to 10%, 20%, and 49%.
media-induced nephro-
toxicity? Clin Nephrol.
sepsis are the most common
2004;62:1-7. [PMID: causes of ATN. Other common Physical examination can detect
15267006]
causes include recent exposure to volume overload (dyspnea, jugu-
23. Brar SS, Aharonian V,
Mansukhani P, et al. nephrotoxic drugs (see the Box) or lar venous distention, rales, as-
Haemodynamic-guided
fluid administration for radiocontrast media, transient hy- cites, lower extremity edema),
the prevention of
potension after surgery or a proce- uremic encephalopathy (leth-
contrast-induced acute
kidney injury: the POSEI- dure, and rapid cell necrosis (he- argy, asterixis, hyperreflexia, and
DON randomised con-
molysis, rhabdomyolysis, and myoclonus), and pericarditis
trolled trial. Lancet.
2014;383:1814-23.
tumor lysis). Pigment-associated (pericardial friction rub), which
[PMID: 24856027]
24. Qian G, Fu Z, Guo J, Cao ATN may cause urine discolor- require urgent therapy. Detec-
F, Chen Y. Prevention of
ation. Tumor lysis can occur before tion of other complications re-
contrast-induced ne-
quires laboratory testing.
phropathy by central or after chemotherapy.
venous pressure-guided
fluid administration in What laboratory tests and
chronic kidney disease It is essential to review the history
and congestive heart imaging should clinicians use?
failure patients. JACC and medical record to determine
The need for laboratory tests and
Cardiovasc Interv. 2016; whether the patient has had CKD
9:89-96. [PMID: imaging depends on the clinical
26685074] or past episodes of AKD. Com-
25. Subramaniam RM, setting. No further evaluation
parison of current values of SCC
Suarez-Cuervo C, Wilson may be necessary for outpatients
RF, et al. Effectiveness of and eGFR to previous levels is
prevention strategies for with AKI and decreased kidney
contrast-induced ne- helpful for early detection of AKI.
perfusion due to volume deple-
phropathy: a systematic Past urinalyses, measures of albu-
review and meta- tion and rapid resolution of AKI
analysis. Ann Intern minuria (albumin-to-creatinine
Med. 2016;164:406-16. after oral or IV volume repletion.
ratio, protein-to-creatinine ratio),
[PMID: 26830221] If volume status is not clear, we rec-
26. Eng J, Wilson RF, Subra- and imaging studies of the kid-
maniam RM, et al. Com- ommend an early therapeutic trial of
parative effect of contrast neys (abdominal ultrasonogra-
media type on the inci- withholding diuretics and adminis-
phy, computed tomography,
dence of contrast- tering an IV fluid bolus of 500 mL
induced nephropathy: a magnetic resonance imaging,
systematic review and isotonic saline over 4–6 hours with
meta-analysis. Ann Intern and angiography) may also be
assessment of volume status, urine
Med. 2016;164:417-24. used. AKI superimposed on CKD
[PMID: 26830055] output, and SCC and eGFR within
27. Nash K, Hafeez A, Hou S. may be due to an exacerbation
Hospital-acquired renal 8–12 hours. Improved urine output,
insufficiency. Am J Kid- of the underlying disease, which SCC, and eGFR suggests AKI due to
ney Dis. 2002;39:930-6. is common in chronic diseases volume depletion, whereas no im-
[PMID: 11979336]
28. Licurse A, Kim MC, Dz- causing decreased kidney perfu- provement suggests some other
iura J, et al. Renal ultra-
sonography in the evalu-
sion, or onset of a new condition. cause.
ation of acute kidney A history of AKD may provide a
injury: developing a risk
stratification framework. clue to the cause of a current epi- In patients with AKI from some
Arch Intern Med. 2010;
170:1900-7. [PMID:
sode, such as a flare of autoim- other cause, urine appearance,
21098348] mune disease or nephrotoxicity dipstick, sediment, and chemis-

" 2017 American College of Physicians ITC72 In the Clinic Annals of Internal Medicine 7 November 2017

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


tries (osmolality, sodium, urea ated with preserved urine concen-
nitrogen, creatinine, albumin, tration, despite the presence of
and total protein) must be as- urine sediment findings suggestive
sessed to ascertain concentra- of ATN.
29. Libório AB, Leite TT,
tion; albuminuria and total pro- Neves FM, Teles F, Be-
teinuria; and the presence or Tests for albumin rather than to- zerra CT. AKI complica-

absence of hematuria, pyuria, tal protein are preferred for eval- tions in critically ill pa-
tients: association with
uation of kidney disease in adults mortality rates and RRT.
renal tubular epithelial cells, and Clin J Am Soc Nephrol.
(12), but both may be helpful in
granular and cellular casts (Ap- 2015;10:21-8. [PMID:
the evaluation of AKI. Loss of al- 25376761]
pendix Table 1 , available at 30. Miller TR, Anderson RJ,
bumin in the urine is a marker of Linas SL, et al. Urinary
Annals.org). If obstruction is sus-
glomerular damage and occurs diagnostic indices in
pected, ultrasonography of the acute renal failure: a
in most parenchymal kidney dis- prospective study. Ann
kidneys is warranted, with a Intern Med. 1978;89:47-
eases other than ATN. Total pro-
postvoiding image of the blad- 50. [PMID: 666184]
teinuria in the absence of albu- 31. Muriithi AK, Nasr SH,
der if symptoms occur during Leung N. Utility of urine
minuria is a marker of increased eosinophils in the diag-
urination (Appendix Table 1 )
production or impaired tubular nosis of acute interstitial
(28). We generally obtain a urine nephritis. Clin J Am Soc
reabsorption of low-molecular- Nephrol. 2013;8:1857-
culture, since urinary tract infec- 62. [PMID: 24052222]
weight serum proteins (light- 32. Wald R, Bell CM, Nisen-
tion can be a cause of AKI (pye-
chain proteinuria or tubular pro- baum R, et al. Interob-
lonephritis or ATN associated server reliability of urine
teinuria, respectively). The urine sediment interpretation.
with sepsis) or may complicate
dipstick is more sensitive to albu- Clin J Am Soc Nephrol.
other causes. Additional tests min than other serum proteins;
2009;4:567-71. [PMID:
19261816]
are required to assess complica- the albumin-to-creatinine ratio 33. Perazella MA, Coca SG,
Kanbay M, Brewster UC,
tions, systemic diseases or dis- and protein-to-creatinine ratio Parikh CR. Diagnostic
eases in other organ systems, provide a quantitative assess-
value of urine micros-
copy for differential diag-
and hemodynamic status in criti- ment, but urine creatinine excre- nosis of acute kidney
injury in hospitalized
cally ill patients. tion decreases when SCC in- patients. Clin J Am Soc
Nephrol. 2008;3:1615-9.
Tests of urine concentration were creases, which may cause a [PMID: 18784207]

initially proposed for the evalua- falsely elevated albumin-to- 34. Chawla LS, Bellomo R,
Bihorac A, et al; Acute
tion of oliguric AKI to distinguish creatinine or protein-to- Disease Quality Initiative

decreased kidney perfusion from creatinine ratio. Workgroup 16. Acute


kidney disease and renal
recovery: consensus
ATN (14, 30). They are also useful Red blood cells and white blood report of the Acute Dis-
in nonoliguric AKI due to these cells may be detected in unspun
ease Quality Initiative
(ADQI) 16 Workgroup.
and other causes. The glomerular urine with a dipstick (heme or Nat Rev Nephrol. 2017;
13:241-257. [PMID:
filtrate is isotonic with plasma; con- leukocyte esterase reagent pads, 28239173]
centration of the urine requires in- respectively) and quantified with 35. Bellomo R, Vaara ST,
Kellum JA. How to im-
tact tubular function. Concentration a manual or automated cell coun- prove the care of patients
of the urine in the setting of AKI indi- ter. We do not recommend rou-
with acute kidney injury
[Editorial]. Intensive Care
cates decreased kidney perfusion tine testing for urine eosinophils Med. 2017;43:727-729.
[PMID: 28600756]
and preserved tubular function. Ab- (31). Detection of renal tubular 36. Davenport A, Anker SD,
sence of urine concentration indi- epithelial cells and granular and Mebazaa A, et al; Acute
Dialysis Quality Initiative
cates impaired tubular function. The cellular casts requires micro- (ADQI) consensus group.
ADQI 7: the clinical man-
fractional excretion of sodium and scopic examination of urine sedi- agement of the Cardio-
urea (FENa and FEurea) can be ment. Increased red or white Renal syndromes: work
group statements from
computed easily from simultaneous blood cells in the urine indicates the 7th ADQI consensus
serum and spot urine samples (Ap- a urinary tract lesion, but the conference. Nephrol Dial
Transplant. 2010;25:
pendix Table 1 ). Diuretic therapy presence of renal tubular epithe- 2077-89. [PMID:
20494894]
impairs sodium reabsorption more lial cells or granular or cellular 37. Facciorusso A, Chandar
than urea reabsorption, so a low casts in the sediment localizes AK, Murad MH, et al.
Comparative efficacy of
FEurea may be a more reliable test the lesion to the kidney. The di- pharmacological strate-
gies for management of
for distinguishing decreased kidney agnostic accuracy of these find- type 1 hepatorenal syn-
perfusion from ATN than a low ings has not been well-studied drome: a systematic
review and network
FENa in patients with recent diuretic (32, 33). Our interpretation is that meta-analysis. Lancet
therapy. Exacerbations of heart, the presence of renal tubular epi- Gastroenterol Hepatol.
2017;2:94-102. [PMID:
lung, or liver disease may be associ- thelial cells and granular and cel- 28403995]

7 November 2017 Annals of Internal Medicine In the Clinic ITC73 " 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


lular casts for the conditions in bodies, complement compo-
Appendix Table 2 (available at nents, and inflammatory markers
38. Dellinger RP, Levy MM,
Rhodes A, et al; Surviv-
Annals.org) is more specific than may be indicated.
ing Sepsis Campaign sensitive—in other words, paren-
Guidelines Committee
chymal kidney disease is more Hemodynamic monitoring (jugular
including the Pediatric
Subgroup. Surviving likely when these findings are venous pressure or pulmonary capil-
sepsis campaign: inter-
present but is not ruled out lary wedge pressure) may be re-
national guidelines for
management of severe
when they are absent. Kidney quired to assess cardiac filling and
sepsis and septic shock:
2012. Crit Care Med. biopsy may be performed when guide volume management in pa-
2013;41:580-637.
suspicion for a parenchymal dis- tients with hypotension. Dynamic
[PMID: 23353941]
39. Cattran DC, Feehally J, ease other than ATN is high. variables, such as pulse–pressure
Cook TH, et al. Kidney variation, inferior vena cava filling on
disease: Improving
global outcomes (KDIGO) Imaging studies are usually done ultrasonography, and echocardiog-
glomerulonephritis work
group. KDIGO clinical
to assess hydronephrosis, de- raphy may be useful. Intra-
practice guideline for fined as dilatation of the renal abdominal pressure can be as-
glomerulonephritis.
Kidney International collecting system due to obstruc- sessed by measuring bladder
Supplement 1s. 2012;2): tion. However, they may also be pressure. Subclavian vein catheters
135.
40. González E, Gutiérrez E, performed to assess kidney should be avoided in patients with
Galeano C, et al; Grupo
Madrileño De Nefritis
shape and size in patients with CKD stages 4–5 (GFR <30 mL/min/
Intersticiales. Early ste- AKI superimposed on CKD or in 1.73 m2) to avoid venous stenosis
roid treatment improves
the recovery of renal patients who have not had previ- that may preclude later vascular ac-
function in patients with ous imaging studies. Renal ultra- cess for hemodialysis.
drug-induced acute inter-
stitial nephritis. Kidney sonography is preferred because
Int. 2008;73:940-6. What other diagnoses should
it has >90% sensitivity for detect-
[PMID: 18185501]
clinicians consider in patients
41. Koyner JL, Davison DL, ing hydronephrosis and is not
Brasha-Mitchell E, et al.
associated with radiation expo- with possible AKI?
Furosemide Stress Test
and Biomarkers for the sure or contrast administration. Decreased GFR is classified as
Prediction of AKI Sever-
ity. J Am Soc Nephrol. Causes of obstruction of the urinary CKD, AKD, or AKI, depending on
2015;26:2023-31.
tract without hydronephrosis in- severity and duration; AKD and
[PMID: 25655065]
42. Chawla LS, Davison DL, clude massive bleeding into the AKI can be superimposed on
Brasha-Mitchell E, et al.
Development and stan- urinary tract or extensive retroperito- CKD (Figure 1 ). Distinguishing
dardization of a furo- neal fibrosis. Dilatation of the urinary among these conditions is impor-
semide stress test to
predict the severity of tract in the absence of obstruction tant for determining the cause of
acute kidney injury. Crit
may be observed after relief of ob- kidney disease and for determin-
Care. 2013;17:R207.
[PMID: 24053972] struction in vesicoureteral reflux, ing the urgency of evaluation and
43. Ho KM, Sheridan DJ.
Meta-analysis of during massive diuresis, and in treatment. Many causes of kidney
frusemide to prevent or
pregnancy. A combination of clinical disease may have an acute or
treat acute renal failure.
BMJ. 2006;333:420. characteristics can identify patients chronic presentation, although
[PMID: 16861256]
in which ultrasonography could be the most common causes of CKD
44. Cantarovich F, Rangoon-
wala B, Lorenz H, et al; omitted (28). (diabetic glomerulosclerosis and
High-Dose Flurosemide
in Acute Renal Failure
hypertension nephrosclerosis) do
Study Group. High-dose Other tests are required to assess not have an acute presentation,
furosemide for estab-
lished ARF: a prospec-
causes and complications of AKI. and ATN does not have a chronic
tive, randomized, Serum urea nitrogen and electro- presentation. Due to the non-
double-blind, placebo-
controlled, multicenter lyte (sodium, potassium, chlo- steady state, changes in SCC and
trial. Am J Kidney Dis.
2004;44:402-9. [PMID:
ride, bicarbonate, calcium, phos- eGFR lag behind those in GFR,
15332212] phorus, magnesium) levels causing delayed recognition of
45. Finfer S, Bellomo R,
Boyce N, et al; SAFE
should be measured. Venous or AKI. The rise in SCC (and de-
Study Investigators. A arterial blood gasses may be re- crease in eGFR) may be slower in
comparison of albumin
and saline for fluid resus- quired for interpretation of acid– patients with low muscle mass or
citation in the intensive
care unit. N Engl J Med.
base disorders. Complete blood volume overload and faster in
2004;350:2247-56. count; liver function tests; muscle patients with high muscle mass
[PMID: 15163774]
46. Perel P, Roberts I. Col- enzymes; and imaging for heart, or volume depletion. When base-
loids versus crystalloids lung, and liver diseases should line GFR is low, minor fluctua-
for fluid resuscitation in
critically ill patients. be obtained. Blood and body tions in GFR can cause a rise in
Cochrane Database Syst
Rev. 2007:CD000567.
fluid cultures and serologic tests SCC by 0.3 mg/dL in the absence
[PMID: 17943746] for infectious diseases, autoanti- of acute kidney disease. Adher-

" 2017 American College of Physicians ITC74 In the Clinic Annals of Internal Medicine 7 November 2017

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


ence to the time requirement for less than 20 mL/min/1.73 m2, it can
diagnosis of AKI (48 hours) can be assessed as the mean of urine
minimize overdiagnosis. An in- clearance of urea and creatinine
crease in SCC (and decrease in during a timed urine collection.
eGFR) in the absence of decline 47. Young P, Bailey M, Beas-
in GFR may occur after a medica-
When should clinicians consider ley R, et al; SPLIT Investi-

tion is started that inhibits creati- consulting a specialist? gators. Effect of a buff-
ered crystalloid solution
nine secretion (trimethoprim or Consultation with a nephrologist vs saline on acute kidney
injury among patients in
cimetidine) or interferes with the is often unnecessary for detect- the intensive care unit:
assay for creatinine (flucytosine ing AKI. However, it should usu- The SPLIT randomized
clinical trial. JAMA.
for the creatinine iminohydrolase ally be requested for identifying 2015;314:1701-10.
[PMID: 26444692]
assay). Serum ketones interfere with the cause of AKI resulting from 48. Hewitt J, Uniacke M,
the widely used colorimetric assay something other than volume Hansi NK, Venkat-Raman
G, McCarthy K. Sodium
for creatinine. GFR, measured using depletion that resolves promptly bicarbonate supplements
for treating acute kidney
clearance of an exogenous filtration with volume repletion (Figure 2 ). injury. Cochrane Data-
marker, or creatinine clearance, can Consultation may also be helpful base Syst Rev. 2012:
CD009204. [PMID:
be assessed to identify misleading for identifying the cause of 22696382]
alterations in SCC or eGFR. If GFR is CKD. 49. Bellomo R, Cass A, Cole
L, et al; RENAL Study
Investigators. Calorie
intake and patient out-
comes in severe acute
Diagnosis... Decreased GFR may be due to AKI, AKD, or CKD. KDIGO kidney injury: findings
guidelines define AKI as an increase in SCC by ≥50% within 7 days or from The Randomized
Evaluation of Normal vs.
≥0.3 mg/dL (26.5 μmol/L) within 2 days, or oliguria for ≥6 hours. The Augmented Level of
stage (severity) is defined by the peak increase in SCC compared with Replacement Therapy
(RENAL) study trial. Crit
previous values and the nadir in urine output, and is related to the risk Care. 2014;18:R45.
for complications and the prognosis. The causes of AKI— decreased kid- [PMID: 24629036]
ney perfusion, obstruction of the urinary tract, parenchymal kidney dis- 50. Bellomo R, Cass A, Cole
L, et al; RENAL Study
eases other than ATN, and ATN—are grouped according to underlying Investigators. Daily pro-
pathophysiology and are the basis for specific therapy. The clinical set- tein intake and patient
outcomes in severe acute
ting, including response to IV fluid, and urinary tract findings are helpful kidney injury: findings of
in determining the cause of AKI. the randomized evalua-
tion of normal versus
augmented level of
replacement therapy
CLINICAL BOTTOM LINE (RENAL) trial. Blood Purif.
2014;37:325-34. [PMID:
25171270]
51. Cox ZL, McCoy AB, Ma-
theny ME, et al. Adverse
drug events during AKI

Treatment and its recovery. Clin J


Am Soc Nephrol. 2013;
8:1070-8. [PMID:
The goals of management of AKI ther diagnostic evaluation is re- 23539228]
include use of specific treatments quired, if AKI is severe or not 52. Matzke GR, Aronoff GR,
Atkinson AJ Jr, et al.
according to the underlying rapidly reversible, or when Drug dosing consider-
ation in patients with
cause and providing supportive complications are present. Man- acute and chronic kidney
care to prevent and manage agement in the ICU should be disease-a clinical update
from Kidney Disease:
complications. KRT is used when considered for patients with AKI Improving Global Out-
comes (KDIGO). Kidney
complications develop that can- and serious illness (35). Int. 2011;80:1122-37.
not be managed with medical [PMID: 21918498]
What pharmacologic therapies 53. McCoy AB, Waitman LR,
therapy alone (Figure 2 ). Gadd CS, et al. A com-
should be used? puterized provider order
When should patients be The use of pharmacologic thera- entry intervention for
medication safety during
hospitalized? pies in the setting of AKI is spe- acute kidney injury: a
quality improvement
For outpatients with new-onset cific to the underlying cause report. Am J Kidney Dis.
or worsening GFR decline, early (Appendix Table 2 ). IV fluids are 2010;56:832-41. [PMID:
20709437]
follow-up is required to distin- recommended to correct volume 54. Bagshaw SM, Wald R.
Strategies for the optimal
guish AKI from AKD and CKD depletion. Afterload reducing timing to start renal
(1, 34). Although evidence to sup- agents are appropriate in cases replacement therapy in
critically ill patients with
port specific criteria for hospitaliza- of acute heart failure (36), acute kidney injury.
tion is not available, hospitalization whereas midodrine, octreotide, Kidney Int. 2017;91:
1022-1032. [PMID:
is generally recommended if fur- and albumin can be used in 28222898]

7 November 2017 Annals of Internal Medicine In the Clinic ITC75 " 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


cases of liver failure (37). IV fluid ceiving dialysis, or time to achieve a serum cre-
55. Gaudry S, Hajage D,
Schortgen F, et al; AKIKI and early antibiotic therapy is atinine level < 200 umol/L was noted (44).
Study Group. Initiation important for treating infections
strategies for renal- KDIGO guidelines recommend
replacement therapy in (38). Withdrawal of NSAIDs,
the intensive care unit. N using vasopressors (e.g., norepi-
Engl J Med. 2016;375: ACEIs, and ARBs is recom-
nephrine or vasopressin) in con-
122-33. [PMID: mended. Immunosuppressive
27181456] junction with fluids in patients
56. Zarbock A, Kellum JA, therapies are recommended for
Schmidt C, et al. Effect of with vasomotor shock accompa-
many causes of acute glomerulo-
early vs delayed initia- nying AKI (1). Low-quality evi-
tion of renal replacement nephritis (39). Although high-
therapy on mortality in dence suggests using a protocol-
critically ill patients with quality evidence is lacking, many
based approach to achieve
acute kidney injury: the experts recommend corticoste-
ELAIN randomized clini- specific mean arterial pressure and
cal trial. JAMA. 2016; roids to treat acute interstitial ne-
315:2190-9. [PMID: other physiologic targets (1). On
27209269]
phritis when AKI is severe or in
the basis of moderate-quality evi-
57. Bellomo R, Cass A, Cole cases of drug-induced interstitial
L, et al; RENAL Replace- dence, KDIGO guidelines recom-
ment Therapy Study nephritis if AKI does not resolve
mend against dopamine, fenoldo-
Investigators. Intensity of after the causative medication is
continuous renal- pam, atrial natriuretic peptides,
replacement therapy in discontinued (40). Plasma ex-
critically ill patients. N insulin-like growth factor, or
Engl J Med. 2009;361: change is recommended for some
N-acetylcysteine to treat AKI (1).
1627-38. [PMID: causes of thrombotic microangi-
19846848]
58. Palevsky PM, O’Connor opathy. High-dose chemotherapy How should clinicians manage
TZ, Chertow GM, et al;
US Department of Veter-
is used in multiple myeloma. There volume problems?
ans Affairs/National Insti- are currently no effective pharma- AKI may develop in the setting of
tutes of Health Acute
Renal Failure Trial Net- cotherapies for treating ATN. The volume depletion or overload,
work. Intensity of renal KIDGO guidelines recommend and either condition may occur
replacement therapy in
acute kidney injury: against diuretics to treat AKI ex- during the course of AKI. Volume
perspective from within
the Acute Renal Failure
cept for management of volume overload is more likely to occur
Trial Network Study. Crit overload. However, a furosemide in the setting of oliguria, is asso-
Care. 2009;13:310.
[PMID: 19678919] “stress test” (administration of 1 ciated with poor outcomes in
59. Pannu N, Klarenbach S,
Wiebe N, Manns B, To-
mg/kg of IV furosemide with 1:1 AKI, and should be avoided to
nelli M; Alberta Kidney replacement of urine output with prevent life-threatening pulmo-
Disease Network. Renal
replacement therapy in saline) can be used to assess prog- nary edema. Volume depletion
patients with acute renal nosis: Patients with <200 mL of should also be avoided because
failure: a systematic
review. JAMA. 2008; urine output over the subsequent it can delay recovery of AKI due
299:793-805. [PMID:
18285591]
2 hours are at greater risk for pro- to other conditions. Frequent
60. Vinsonneau C, Camus C, gression to a higher AKI stage or monitoring of fluid intake and
Combes A, et al; Hemo-
diafe Study Group. Con- to the need for KRT (41, 42). Al- output, body weight, and volume
tinuous venovenous though high-dose loop diuretics status and administering or re-
haemodiafiltration versus
intermittent haemodialy- can increase urine output in AKI, stricting fluid depending on the
sis for acute renal failure
in patients with multiple-
they do not seem to reduce mor- results are important.
organ dysfunction syn- tality or the need for dialysis.
drome: a multicentre
randomised trial. Lancet. IV fluids are recommended for
2006;368:379-85. A systematic review of 6 moderate- to low- correcting AKI with volume de-
[PMID: 16876666] quality randomized controlled trials of furo-
61. Coca SG, Yusuf B, Sh- pletion and for intravascular vol-
lipak MG, Garg AX, semide to treat AKI reported that it did not re-
Parikh CR. Long-term risk duce in-hospital mortality, requirement for ume expansion in AKI with sepsis
of mortality and other
dialysis, or the number of dialysis treatments (1). Isotonic crystalloids (e.g., nor-
adverse outcomes after
acute kidney injury: a required until recovery of kidney function. In mal saline, Ringer lactate, or
systematic review and
meta-analysis. Am J addition, higher doses were associated with in- other balanced crystalloid solu-
Kidney Dis. 2009;53: creased toxicity (43). tions) are recommended rather
961-73. [PMID:
19346042] than colloids (albumin or
In a moderate-quality randomized trial of 338
62. James MT, Samuel SM, starches) as initial management
Manning MA, et al. patients from ICUs and nephrology wards who
Contrast-induced acute on the basis of moderate-quality
kidney injury and risk of had AKI requiring KRT, patients who were ran-
adverse clinical out- domly assigned to daily furosemide 25 mg/kg evidence (45– 47). Colloids may
comes after coronary
IV or 35 mg/kg orally more rapidly achieved be used for patients with liver
angiography: a system-
atic review and meta- urine output >2 L per day (5.7 days with furo- failure or burns. Treatment of vol-
analysis. Circ Cardiovasc
Interv. 2013;6:37-43.
semide vs. 7.8 days with placebo). However, ume overload in patients with
[PMID: 23322741] no significant differences in mortality, time re- AKI can sometimes be accom-

" 2017 American College of Physicians ITC76 In the Clinic Annals of Internal Medicine 7 November 2017

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


plished using high doses of IV erally appropriate. In the presence
loop diuretics, given as multiple of volume overload, water restric-
doses throughout the day or as tion and loop diuretics should be
an infusion and often in conjunc- used. Hypercalcemia may be en-
tion with IV thiazide diuretics. countered in AKI accompanying
multiple myeloma and can be cor-
How should clinicians manage rected by volume expansion and
electrolyte problems? loop diuretics in patients who are
Initial management of electrolyte not oliguric or by promoting bone
abnormalities should start with uptake with administration of an IV
medical management and pro- bisphosphate. Hyperphos-
ceed to KRT when medical man- phatemia can be managed by di-
agement is no longer satisfac- etary phosphate restriction and
tory. Dietary potassium should oral phosphate binders. Hyperma- 63. Coca SG, Singanamala S,
be restricted, and medications gnesemia may occur after magne- Parikh CR. Chronic kid-
ney disease after acute
that cause hyperkalemia should sium infusion and can be man- kidney injury: a system-
atic review and meta-
be used cautiously. Treatment of aged by discontinuing the infusion analysis. Kidney Int.
severe hyperkalemia (serum po- and administering loop diuretics. 2012;81:442-8. [PMID:
22113526]
tassium > 6.5 mmol/L or with 64. Hsu CY, Hsu RK, Yang J,
electrocardiographic changes)
How should clinicians manage et al. Elevated BP after
AKI. J Am Soc Nephrol.
includes administration of cal- nutrition, drug dosing, and 2016;27:914-23. [PMID:

cium gluconate to reduce the risk anticoagulation? 26134154]


65. Sawhney S, Marks A,
for arrhythmia, followed by insu- Nutrition should provide ade- Fluck N, et al. Post-
discharge kidney func-
lin plus dextrose, !-agonists, or quate calories with restricted po- tion is associated with
sodium bicarbonate to shift po- tassium and phosphate. KDIGO subsequent ten-year
renal progression risk
tassium from the extracellular to guidelines for patients with AKI among survivors of acute
kidney injury. Kidney Int.
the intracellular compartment. recommend a total energy intake 2017;92:440-452.
These treatments are temporary of 20 –30 kcal/kg per day, prefer- [PMID: 28416224]
66. Schmitt R, Coca S, Kan-
and must be accompanied by ably provided via the enteral bay M, et al. Recovery of

measures to remove potassium route (49). Minimal nitrogenous kidney function after
acute kidney injury in
from the body. For patients with- waste production is desirable in the elderly: a systematic
review and meta-
out oliguria, high-dose loop di- AKI; however, protein restriction analysis. Am J Kidney
uretics can be used to increase is not suggested as a means to Dis. 2008;52:262-71.
[PMID: 18511164]
urine output and potassium ex- avoid KRT. On the basis of low- 67. Heung M, Steffick DE,

cretion. For patients with oliguria, quality evidence, KDIGO guide- Zivin K, et al; Centers for
Disease Control and
sorbitol with sodium polystyrene lines recommend protein goals Prevention CKD Surveil-
lance Team. Acute kidney
sulfonate or calcium polystyrene of 0.8 –1.0 g/kg per day in non- injury recovery pattern

sulfonate resins can be used to catabolic patients, 1.1–1.5 g/kg and subsequent risk of
CKD: an analysis of Vet-
per day in patients receiving re-
induce osmotic diarrhea and fe- erans Health Administra-
nal replacement therapy, and a tion data. Am J Kidney
cal potassium losses. Most ex- Dis. 2016;67:742-52.
maximum 1.7 g/kg per day in [PMID: 26690912]
perts suggest using supplemen- 68. Wu VC, Wu CH, Huang
hypercatabolic patients or those
tal sodium bicarbonate when TM, et al; NSARF Group.
requiring continuous KRT (1, 50). Long-term risk of coro-
metabolic acidosis is severe, al- nary events after AKI. J
In critically ill patients, KDIGO Am Soc Nephrol. 2014;
though there is no high-quality
guidelines suggest insulin ther- 25:595-605. [PMID:
evidence (48). Hypernatremia 24503241]
apy targeting plasma glucose 69. Ftouh S, Thomas M;
may be encountered in AKI with Acute Kidney Injury
110 –149 mg/dL.
dehydration, after normal saline Guideline Development
Group. Acute kidney
resuscitation, or when access to Patients with AKI require special injury: summary of NICE
guidance. BMJ. 2013;
water is restricted. It can usually drug dosing due to buildup from 347:f4930. [PMID:
be corrected by providing water decreased excretion and metab- 23985310]
70. Soares DM, Pessanha JF,
via enteral routes or IV hypnatric olism by the kidney as well as the Sharma A, Brocca A,
Ronco C. Delayed ne-
solutions. The management of effects of kidney failure on other phrology consultation
hyponatremia in the setting of routes of drug excretion and me- and high mortality on
acute kidney injury: a
AKI depends on its cause. In tabolism (51, 52). Estimated GFR meta-analysis. Blood
states of volume depletion, admin- is less accurate to guide dosing Purif. 2017;43:57-67.
[PMID: 27915348] doi:
istration of isotonic IV fluids is gen- in the nonsteady state than in the 10.1159/000452316

7 November 2017 Annals of Internal Medicine ITC77 " 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


steady state. Expert consensus What is the role of KRT? Significant differences between
recommends closely monitoring KRT is used to manage the com- the 2 methods in terms of mor-
drug response in patients with plications of severe AKI. Com- tality, length of hospitalization,
known nephrotoxicity or other mon indications include life- and long-term requirements for
toxicities, using therapeutic drug threatening changes in fluid, dialysis have not been found
monitoring when clinically useful electrolyte, and acid– base bal- (59, 60).
assays are available, and being ance that require emergent cor- What is the prognosis?
cognizant of residual effects of rection (Figure 2 ). KRT may be
AKI is associated with high mor-
medicines that are excreted by started before these complications
tality rates that range from 16%–
the kidney even after they have develop, although evidence con-
50% according to stage (Appen-
been discontinued (e.g., oral flicts about whether preemptive
dix Figure, available at Annals.
hypoglycemic agents and opi- initiation at an earlier stage of AKI
org) and vary according to the
oids) (52, 53). The assistance of a is more beneficial than when com-
cause, coexisting conditions, and
pharmacist may be helpful, par- plications occur (54 –56). KRT
availability of KRT (3). Survivors
ticularly in determining dosing should be discontinued as soon as
of AKI are at increased risk for
and dosing intervals in patients it is no longer required, either be-
hypertension and progressive
receiving dialysis and those re- cause kidney function has recov-
CKD, including end-stage kidney
quiring antibiotics for AKI ac- ered enough to meet the patient's
disease (61– 65). KDIGO guide-
companying sepsis. needs or life-sustaining therapy is
lines thus recommend evaluat-
no longer the goal of the patient's
ing patients after AKI for recov-
Certain anticoagulants should be care.
ery of kidney function, new
used with caution in patients with
Several types of KRT can be onset, or worsening of preexist-
AKI due to the increased bleeding
used, and the type varies by loca- ing CKD. Older age, lower base-
risk attributed to uremic platelet
tion of care and available equip- line eGFR, higher baseline albu-
dysfunction and to decreased minuria, and AKI severity
excretion of low-molecular-weight ment and expertise (Appendix
Table 3 , available at Annals.org). (KDIGO stage) are predictors of
heparins and direct oral anticoag- CKD after AKI and should
Peritoneal dialysis is rarely used
ulants. Although some manufac- prompt postdischarge follow-up
for KRT in adults with AKI in
turers provide altered dosing (6, 66, 67). Referral to commu-
North America, although it is
schedules based on creatinine nity nephrology services may be
more commonly used in areas
clearance for some low- appropriate for patients who do
with limited resources. Continu-
molecular-weight heparins (e.g., not recover kidney function
ous KRT is a slow, continuous
enoxaparin) and direct oral anti- given the associated long-term
form of therapy delivered 24
coagulants (e.g., dabigatran), de- risks of kidney failure, cardiovas-
hours a day in ICUs and is usu-
termining safe dosing schedules ally used to treat hemodynami- cular events, and mortality for
when GFR is changing, especially cally unstable patients (57). several years after hospital dis-
during dialysis, is difficult. Al- Conventional intermittent hemo- charge (62, 68).
though low-molecular-weight dialysis is used in AKI for hemo-
heparins are used intermittently in In a systematic review, the pooled mortality
dynamically stable patients; it is rate was 23.0% overall and 49.4% in patients
patients receiving long-term dialy- similar to the in-center long-term needing dialysis (Appendix Figure) (3). The
sis and some organizations have hemodialysis used for end-stage pooled unadjusted odds of death were 3.4-
suggested monitoring anti-factor kidney disease (58). More fre- fold higher for patients with KDIGO stage 1
X activity with continuous use, quent sessions may be required AKI, 7.5-fold higher for stage 2, 13.2-fold
many experts prefer to use unfrac- to manage fluid and electrolyte higher for stage 3, and 24-fold higher in pa-
tionated heparin in severe AKI. abnormalities for some patients tients needing dialysis compared with patients
For patients with active bleeding with AKI. Prolonged intermittent without AKI. The AKI-associated mortality rate
in the presence of uremia, desmo- KRT uses the same equipment declined over 8 years and was inversely related
pressin (0.3 mcg/kg IV, subcuta- to the percentage of a country's gross domes-
as conventional dialysis but pro-
tic product spent on total health
neous, or intranasal) and cryopre- vides dialysis using lower blood expenditure.
cipitate can be used to rapidly flow rates over longer sessions
diminish bleeding time, whereas (usually ≥6 hours/session). Sev- When should clinicians
conjugated estrogens and dialysis eral randomized trials have com- consider consulting a specialist?
(without systemic anticoagulation) pared outcomes with continuous Because AKI is a common com-
can be used for more prolonged KRT versus intermittent hemodi- plication of many medical and
bleeding control. alysis in critically ill patients (59). surgical illnesses, most patients

" 2017 American College of Physicians ITC78 Annals of Internal Medicine 7 November 2017

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


with AKI are cared for by general tion have been excluded or cor-
internists, hospitalists, surgeons, rected. Clinicians should discuss
and critical care and primary care management with a nephrologist if
physicians. These clinicians a diagnosis that may need specific
should consult a nephrologist or treatments is suspected (e.g., paren-
critical care specialist for patients chymal kidney diseases other than
who have AKI with any indication ATN), response to treatment has
for KRT (69). Nephrology referral been inadequate, there are associ-
and comanagement is also appro- ated complications, AKI is severe
priate if the cause of AKI is uncertain, (KDIGO stage 3), or it is superim-
particularly when decreased kidney posed on CKD stages 4–5 (baseline
perfusion and urinary tract obstruc- GFR < 30 mL/min/1.73 m2) (70).

Treatment... The main goals of managing AKI are specific pharmaco-


logic therapy addressing underlying causes and providing supportive
care to prevent and manage complications. Supportive care includes
maintenance of hemodynamic stability and kidney perfusion using IV
crystalloids when volume expansion is needed and the use of diuretics
in states of volume overload, medical management of electrolyte disor-
ders, provision of adequate nutrition and glycemic control, and cau-
tious use of medications to avoid adverse drug events. When AKI is se-
vere, KRT may be required to manage complications. Survivors of AKI
should be assessed for recovery of kidney function and receive subse-
quent follow-up to identify and subsequently manage CKD.

CLINICAL BOTTOM LINE

In the Clinic Clinical Guidelines

Tool Kit
www.kdigo.org/clinical_practice_guidelines/pdf
/KDIGO%20AKI%20Guideline.pdf
Guidelines published by the International Society of
Nephrology.
www.clinicalguidelines.gov.au/portal/2481/clinical
IntheClinic
-practice-guideline-acute-kidney-injury
2014 guideline from the National Health and Medical

Acute Kidney Research Council of Australia.


www.nice.org.uk/guidance/cg169
Injury Guidance from the National Institute for Health and Care
Excellence.

Patient Information
www.kidney.org/atoz/content/AcuteKidneyInjury
Information presented by the National Kidney foundation
for both patients and caregivers.
www.thinkkidneys.nhs.uk/aki/wp-content/uploads
/sites/2/2015/11/BKPA-RCGP-A4-Printout-Leaflet_v4
.pdf
Printable leaflet of the British Kidney Patient Association.
www.ncbi.nlm.nih.gov/pubmedhealth/PMH0071507/
Information for patients and caregivers from PubMed
Health.
www.nhs.uk/conditions/acute-kidney-injury/Pages
/Introduction.aspx
Information from the National Health Service.

7 November 2017 Annals of Internal Medicine ITC79 " 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


WHAT YOU SHOULD KNOW In the Clinic
Annals of Internal Medicine
ABOUT ACUTE KIDNEY INJURY
What Is Acute Kidney Injury?
Doctors diagnose acute kidney injury (AKI) when
your kidneys stop working properly and the
change happens over a few hours or a few days.
It can cause a buildup of waste products in your
blood and make it hard for your kidneys to bal-
ance the fluids in your body. It can lead to seri-
ous health problems, such as permanent kidney
damage and even death.
AKI usually happens to people who are sick, espe-
cially when they are in the hospital. AKI can be a
complication of the following conditions:
• Sepsis (a type of infection)
• Shock
• Accidents
• Burns • Swelling or retaining fluid
• Heart attack or other heart disease • Shortness of breath
• Blockage of the urinary tract • Feeling dizzy or lightheaded
• Infection of the urinary tract
• Surgery How Is It Diagnosed?
• Dyes for x-rays • Your doctor will ask you questions about your
• Certain over-the-counter and prescription health history and take blood and urine
medicines samples.
• Contact with a poisonous plant or animal • If there is something wrong with your kidneys,
• Severe allergic reaction you may have an imaging test. This could
include an ultrasound.
What Are the Risk Factors?
You are more susceptible to AKI if you: How Is It Treated?
• Are an older adult • Patients with AKI are usually treated in a
• Are female hospital. Your health care provider will give
• Are African American you intravenous fluids to bring your levels

Patient Information
• Have chronic kidney disease back to normal. You may also take medicines
• Have chronic heart, lung, or liver disease to help balance your fluid levels.
• Have diabetes • In more serious cases, you may need dialysis
• Have cancer while your kidneys recover. Dialysis filters the
• Are anemic waste in your body, just as healthy kidneys do.
What Are the Symptoms? Questions for My Doctor
AKI usually has no symptoms until your kidneys • What caused my AKI?
start to fail. Kidney failure means that your kid- • Will I need dialysis?
neys can no longer properly remove waste from • Will I have permanent damage?
your body. These symptoms may occur if you • If I recover, what are the chances I'll get it
have kidney failure: again in the future?
• Feeling sleepy • Should I eat a special diet?
• Feeling sick to your stomach • Should I avoid any medications?

For More Information


National Kidney Foundation
www.kidney.org/atoz/content/AcuteKidneyInjury
Medline Plus
https://medlineplus.gov/ency/article/000501.htm

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


Appendix Table 1. Findings of Urine Testing in Acute Kidney Injury
Variable Normal Decreased Obstruction of the Parenchymal Kidney ATN
Kidney Perfusion Urinary Tract Diseases Other Than ATN
Concentration* Varies, depending Concentrated, Concentrated, not if Not concentrated, may Not concentrated;
on water intake possibly not if chronic be concentrated in may be concen-
recent glomerulonephritis trated in heart
diuretic failure or liver
administration failure
Albumin/total Normal to mildly Normal to mildly Normal to mildly Increased albumin Normal to mildly
protein levels† increased increased increased (unless and total protein in increased
chronic) glomerular disease;
normal albumin and
increased total protein
in cast nephropathy
(immunoglobulin light
chains) and in tubular
diseases (!2-micro-
globulin)
Hematuria (RBCs)‡ No No Yes, with instrinsic Yes, with dysmorphic No, heme-positive
disease RBCs and RBC casts in dipstick in
glomerulonephritis; yes, hemolysis and
without RBC casts in rhabdomyolysis
thrombotic
microangiopathy
Pyuria (WBCs)‡ No No Yes, if superimposed Yes, with WBC casts in No
urinary tract interstitial nephritis
infection
Renal tubular No No (may contain No (except if Yes Yes
epithelial cells hyaline casts) chronic)
and granular
casts
Hydronephrosis, No No Hydronephrosis, No hydronephrosis, No
kidney shape hydronephrosis, hydronephrosis, enlarged, may be normal size, may be hydronephrosis,
and size§ normal size, normal size, small if chronic, large in infiltrative normal size
symmetrical symmetrical, may be diseases, symmetrical,
may be asymmetrical if may be small if chronic
asymmetrical unilateral,
if renal artery nephrolithiasis
stenosis may be present

RBC = red blood cell; HPF = high-power field; WBC = white blood cell.
* Urine concentration assessed from urine specific gravity (concentrated > 1.020); osmolality (concentrated >500 mosm/kg);
fractional excretion of sodium (FENa) (concentrated <1%); fractional excretion of urea (FEurea) (concentrated <35%); ratio of SUN
to serum creatinine clearance (Scr) >20:1. FENa is calculated from UNa x Scr/SNa x Ucr. FEurea is calculated from UUN x Scr/SUN
x Ucr.
† Albumin and total protein assessed from dipstick (more sensitive to albumin than other proteins); albumin-to-creatinine ratio
(ACR), moderately increased 30 –300 mg/g, severely increased >300 mg/g (nephrotic range >2200 mg/g); and total protein-to-
creatinine ratio (PCR), moderately increased 150 –500 mg/g, severely increased >500 mg/g (nephrotic range >3500 mg/g).
Ranges for ACR and PCR are defined for steady-state conditions; values may be higher in acute kidney injury because urine
creatinine excretion decreases with increased Scr.
‡ Normal urine contains <5 RBC/HPF and <5 WBC/HPF.
§ Renal ultrasonography is the preferred method for evaluating hydronephrosis. Hydronephrosis may be absent in cases of
massive bleeding into the urinary tract or extensive retroperitoneal fibrosis.

" 2017 American College of Physicians Annals of Internal Medicine 7 November 2017

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


Appendix Table 2. Treatment According to Specific Causes of AKI Other Than Acute Tubular Necrosis
Cause of AKI Treatment/Intervention Comment
Decreased kidney perfusion
Volume depletion Intravenous fluid Isotonic crystalloids (normal saline, Ringer lactate, or
balanced crystalloid solutions) are recommended
rather than colloids as initial management for
intravascular volume expansion; improvement in Scr
and eGFR and urine output after intravenous fluid
administration confirms the diagnosis of decreased
kidney perfusion due to volume depletion
Heart failure, left- or right-sided Afterload reduction and Cardiac output may be improved with ACE/ARB,
(with lung disease) (cardiorenal diuretics if volume hydralazine or nitrates; diuresis can improve GFR when
syndrome) overloaded left ventricular filling pressure exceeds that for
optimum cardiac output or when systemic venous
pressure is elevated so as to reduce the afferent versus
efferent arteriolar pressure gradient across the
glomerulus
Liver failure (hepatorenal syndrome) Albumin, midodrine, and Volume may be expanded with albumin, especially in
octreotide setting of bacterial peritonitis or paracentesis;
midodrine and octreotide are used to raise blood
pressure and promote splanchnic vasoconstriction;
terlipressin may improve outcomes (but is not currently
available in the United States or Canada); kidney failure
can be an indication for liver transplant in eligible
patients
Sepsis Intravenous fluid and Intravenous volume expansion and early broad-spectrum
antibiotics antibiotics, subsequently tailored to culture and
sensitivity results
Increased intra-abdominal pressure Intra-abdominal Nasogastric and rectal drainage, and evacuation of
decompression intra-abdominal space-occupying lesions (e.g., ascites,
fluid collections, hematomas); may require surgical
decompression (open abdomen) when
intra-abdominal pressure is ≥20 mm Hg
Renovascular disease Antihypertensive therapy; Angioplasty with or without stenting or surgical bypass of
revascularization the renal artery can be considered in select situations
of AKI (e.g., bilateral renal artery stenosis or stenosis to
a solitary kidney)
Nonsteroidal anti-inflammatory Withdrawal of medications Isotonic crystalloid is a useful adjunct
drugs
ACE inhibitors and ARBs Temporary medication Consider educating patients about tablet holidays
withdrawal (withholding ACE inhibitors or ARBs, especially when
combined with diuretics); during periods of potential
volume depletion from acute illness, to avoid recurrent
AKI episodes
Urinary tract obstruction
Obstructive nephropathy Nephrostomy tube or Percutaneous nephrostomy tubes or ureteric stenting for
urinary catheter relief of upper tract obstruction or Foley catheter or
definitive surgical intervention for lower tract
obstruction
Parenchymal kidney disease
Acute glomerulonephritis Immunosuppressive therapy Dependent on histopathologic findings on kidney biopsy
and underlying cause; crescentic glomerulonephritis
(e.g., anti-neutrophil cytoplasmic antibody and
anti-GBM disease) is usually treated with induction
therapy using steroids and cyclophosphamide;
proliferative lupus nephritis is treated with steroids and
cyclophosphamide or mycophenolate mofetil for
induction therapy; corticosteroids or other
immunosuppressive regimens may be used to treat IgA
nephropathy; when glomerulonephritis is due to an
infection or cancer, these secondary causes should be
treated

Continued on following page

7 November 2017 Annals of Internal Medicine " 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


Appendix Table 2—Continued
Cause of AKI Treatment/Intervention Comment
Acute interstitial nephritis Withdrawal of culprit Stop culprit drug when medication-related; may consider
medication and corticosteroids if AKI is severe or patient is not
corticosteroids recovering
Acute pyelonephritis Antibiotics Intravenous antibiotics when accompanied by AKI;
consider investigations for anatomical urinary tract
abnormalities, obstruction or stones, or sources of
hematogenous dissemination
Thrombotic microangiopathy Plasma exchange Used for thrombotic thromobocytopenia purpura and
some secondary (immune-mediated) causes of
thrombotic microangiopathy.
Cast nephropathy (multiple Chemotherapy High-dose chemotherapy provided through consultation
myeloma) with a hematologist, which may include steroids,
cyclophosphamide, immunomodulatory drugs (e.g.,
lenolidamide, or proteasome inhibitors (e.g.,
bortezemab) for initial treatement, followed by
autologous hematopoietic cell transplantation for
eligible patients
Renal infarction Revascularization or Consider revascularization for arterial fibromuscular
anticoagulation dysplasia of the renal arteries or anticoagulation for
arterial embolism or venous thrombosis to prevent
recurrence; not indicated for treatment of acute events
Atheroembolism Avoidance of intra-arterial Avoidance of intra-arterial procedures to prevent
procedures recurrence; consider withdrawal of anticoagulation (if
recently initiated) and atheroembolism is spontaneous
(not following intra-arterial procedures)

ACE = angiotensin-converting enzyme; AKI = acute kidney injury; ARB = angiotensin-receptor blocker; eGFR = estimated glo-
merular filration rate; GBM = glomerular basement membrane; GFR = glomerular filration rate; Scr = serum creatinine.

Appendix Table 3. Kidney Replacement Therapies for Hemodialysis and Hemofiltration in Acute Kidney Injury
Example Solute Removal Blood Flow Rate Duration Recommended Dose
Continuous kidney
replacement
therapies
Continuous Convective 150–250 mL/min Daily for 24 h/d (minus To deliver an effluent volume
venovenous interruptions) (replacement for fluid
hemofiltration removed by ultrafiltration)
of 20–25 mL/kg per h
Continuous Diffusive
venovenous
hemodialysis
Continuous Diffusive and
venovenous convective
hemodiafiltration
Intermittent kidney
replacement
therapies
Intermittent Diffusive 200–350 mL/min Typically 3–4 times/wk, To deliver a weekly Kt/V (a
hemodialysis 4 h/session measure of urea clearance)
of 3.9
Prolonged intermittent
kidney replacement
therapies
Sustained low Diffusive 100–300 mL/min Typically daily for ≥6 h To deliver a weekly Kt/V (a
efficiency dialysis measure of urea clearance)
of 3.9
Sustained low Diffusive and
efficiency convective
diafiltration
Sustained continuous Convective
ultrafiltration

" 2017 American College of Physicians Annals of Internal Medicine 7 November 2017

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


Appendix Figure. Pooled AKI-associated mortality rate from studies based on KDIGO creatinine-based staging.

70

60 49.4
47.8

Pooled AKI-Associated
50

Mortality Rate 40
28.5
30 23.0
15.9
20

10

Overall Stage 1 Stage 2 Stage 3 Dialysis


(KDIGO- (Risk) (Injury) (Failure) Requirement
Equivalent)

Studies, n 110 26 25 25 31
Patients With AKI, n 429 535 8226 42 354 42 354 6534

Error bars represent 95% CIs. AKI = acute kidney injury; KDIGO = Kidney Disease Improving Global Outcomes.

7 November 2017 Annals of Internal Medicine " 2017 American College of Physicians

Downloaded From: https://annals.org/ by a Scott Memorial Library User on 12/03/2017


ARTIGO 2

EMERGÊNCIAS DIALÍTICAS
ACUTE KIDNEY INJURY

The management of acute Outcomes (KDIGO) Work Group has integrated the RIFLE and
AKIN criteria, including patients under 18 with a decrease in
eGFR to less than 35 ml/min in the stage 3 category.5 (see Table 1
kidney injury of Assessment and initial management of acute kidney injury on
pages 000e000 of this issue).
Carolyn E Amery
Annette Davies General management of AKI
Lui G Forni Early recognition and treatment of AKI saves nephrons and
prevents further decline in GFR. It is important to remember that
Abstract measured serum creatinine may not rise appreciably until GFR
Acute kidney injury affects up to 15% of in patients in the acute hospital has fallen significantly. This is of particular relevance in in-
setting. Although accurate history-taking, careful physical examination dividuals of small build, vegetarians and the undernourished,
and meticulous monitoring of volume balance are essential, there is, such as patients with hepatic failure, in whom a serum creatinine
to-date, little evidence supporting any intervention that may reverse in the normal range can be misleading. Tubular secretion of
this process. Acute kidney injury presents a unique set of metabolic de- creatinine is increased as GFR falls, and this may also lead to an
rangements that, if untreated, will result in death. We outline the initial overestimation of renal function in AKI.6 Treatment in AKI is
management of acute kidney injury as well as specific treatments that aimed at minimizing further damage to the kidney while
may be required. Some consideration is also given to the use of renal providing support until there are signs of functional recovery.
replacement therapies. This includes restoration of the circulating volume, relief of
outflow obstruction if present, removal of tubular toxins and
Keywords Acute kidney injury; glomerular filtration rate; haemofiltra-
specific treatment of glomerular disease. Early restoration of
tion; hyperkalaemia; metabolic acidosis; uraemic encephalopathy; urae-
renal perfusion in precipitant AKI due to presumed acute tubular
mic pericarditis
necrosis is essential. Recovery of GFR depends on the number of
remaining functional nephrons that will increase their filtration
to maintain GFR. However, continued hyperfiltration may result
Introduction
in progressive glomerular sclerosis and nephron death, leading to
In 2004, the Acute Dialysis Quality Initiative group (ADQI) pro- end-stage renal failure. (See Assessment and initial management
posed the RIFLE classification of acute kidney injury (AKI) of acute kidney injury on pp xxexx of this issue.)
encompassing two separate criteria, the calculated glomerular
filtration rate (GFR) and urine output.1 The previously adopted Specific problems
acronym RIFLE provides diagnostic definitions for the three
Volume resuscitation
grades of increasing severity of AKI and the two outcome vari-
In hypovolaemia, volume expansion is recommended. However,
ables of loss (L) and end-stage renal disease (E). The grades of
uncontrolled volume substitution may result in clinical deterio-
severity of injury include the stage at which injury can be pre-
ration with an increased risk of morbidity and mortality in pa-
vented (risk, R), when the kidney has already been damaged
tients with AKI and should be avoided. Isotonic crystalloids
(injury, I) and when renal failure has occurred (failure, F). In
remain the mainstay of volume replacement therapy. Crystalloids
2007, the Acute Kidney Injury Network (AKIN) modified the
expand plasma volume by about 25% of the infused volume,
RIFLE criteria.2 The AKIN criteria refer to the same stages (risk,
correcting sodium depletion as well as restoring solute and water
injury and failure) but the time frame for diagnosis of AKI is
diuresis. However, large-volume infusion of sodium chloride can
reduced to 48 hours, and a lower threshold for the rise of serum
lead to so-called ‘hyperchloraemic acidosis’, which may be
creatinine from baseline to peak value is used. Both sets of
associated with renal vasoconstriction and gut hypoperfusion.7
criteria have been validated for in-hospital mortality in numerous
Colloids, such as albumin, gelatins and hydroxyethyl starch,
studies and can offer prognostic information based on the stages
result in volume expansion approximate to the infused volume
of AKI.3,4 More recently, the Kidney Disease: Improving Global
but, if administered in isolation in AKI, can lead to osmotic
nephrosis (osmotic tubular damage). Hydroxyethyl starches are
highly polymerized sugars characterized by their molecular
Carolyn E Amery MBChB MRCP is a Specialist Registrar in Renal Medicine weight, grade of substitution and concentration. They are
at Brighton and Sussex University Hospitals Trust, Brighton, East degraded through hydrolytic cleavage, the remnants of which are
Sussex, UK. Competing interests: none. excreted by the kidney, and should be avoided in AKI particularly
when this has resulted from sepsis.
Annette Davies RN BSc (Hons) MSc is Teaching Fellow at School of Health
Sciences, University of Surrey, UK. Competing interests: none.
Volume overload
Lui G Forni MBBS BSc PhD MRCPI is visiting Professor in Intensive Care The volume status of a patient with AKI should be assessed
Medicine at the School of Health Sciences, University of Surrey and carefully. Although somewhat unfashionable, careful bedside
Consultant Intensivist, Department of Intensive Care Medicine, Surrey examination including assessment of the venous pressure,
Peri-Operative Anaesthesia Critical Care Collaborative Research Group capillary refill time, pulse and postural blood pressure changes
(SPACeR), Royal Surrey County Hospital NHS Foundation Trust, UK. are elementary tools for assessing volume status. Hourly urine-
Competing interests: none. output and accurate fluid-input charts need to include all fluid

MEDICINE --:- 1 Crown Copyright ! 2015 Published by Elsevier Ltd. All rights reserved.

Please cite this article in press as: Amery CE, et al., The management of acute kidney injury, Medicine (2015), http://dx.doi.org/10.1016/
j.mpmed.2015.05.015
ACUTE KIDNEY INJURY

losses, including estimated insensible losses, drain/stoma output Uraemic pericarditis


and nasogastric losses where appropriate. If possible, patients Uraemic pericarditis is observed in 6e10% of patients with
should be weighed daily. A central venous catheter and an advanced renal failure, resulting from inflammation of both the
arterial cannula can be helpful where multi-organ failure (MOF) visceral and parietal membranes of the pericardial sac. Pericar-
is imminent. In established MOF, absolute measurement of ditis in AKI presents with fever and pleuritic chest pain, char-
central venous pressure can be misleading, especially in the acteristically worse in the recumbent position. A pericardial rub
ventilated patient in whom high intrathoracic pressures may be may be heard on auscultation, although the ECG does not show
present, and further invasive monitoring may be required. the typical diffuse ST and T wave elevations observed with other
Where volume overload is encountered in the setting of AKI, causes of acute pericarditis. The development of pericarditis in a
symptoms can be alleviated by high-flow oxygen. Where pul- patient with AKI is an indication to institute RRT, unless there are
monary oedema is refractory to pharmacological management signs of cardiac tamponade due to a pericardial effusion. Under
while preparing the patient for urgent RRT, continuous positive such conditions, heparin-free haemodialysis or haemofiltration
airway pressure ventilation (CPAP) should be considered. Trials should be used because of the risk of increased bleeding into the
using diuretics, such as furosemide and mannitol, and ‘low-dose’ pericardial sac with anticoagulation.
dopamine have yielded inconsistent results in AKI.8,9 Although
loop diuretics may promote diuresis in oliguric renal failure, there Uraemic encephalopathy
is little evidence that they influence outcome,10 and the large Like uraemic pericarditis, uraemic encephalopathy tends to be
doses of furosemide often required for diuresis in AKI can result related to the degree of uraemia. Early clinical features include
in ototoxicity. Similarly, the use of dopamine does not reduce rambling speech, disorientation, lethargy, irritability, hallucina-
mortality or accelerate the recovery of renal function and, in the tions and, more rarely, coma. Commonly encountered signs
critically ill, dopamine use can lead to peripheral vasoconstric- include tremor, myoclonus and asterixis, which tend to occur with
tion, causing gut ischaemia, tissue necrosis and digital gangrene. deterioration in mental status. Transient focal signs, such as hem-
iparesis or reflex asymmetry, occur rarely but resolve with treat-
Hyperkalaemia ment, although mental state may not improve for up to 48 hours. If
This life-threatening complication arises through cellular shifts of the signs do not resolve, other pathologies should be excluded.
potassium or release from lysed cells, together with decreased
renal excretion of potassium, and requires immediate treatment. Renal replacement therapy
Acidosis, hyponatraemia and hypocalcaemia all potentiate the
Renal replacement therapy (RRT) does not cure acute kidney
harmful effects of hyperkalaemia on cardiac function and must
injury but it is a safe and efficient way of replacing renal function
also be corrected. Muscle weakness may be present and, if se-
while the kidneys recover from disease or injury. Historically,
vere, can lead to flaccid paralysis, although symptoms are rarely
single-organ AKI has been treated with either peritoneal or
apparent until the serum potassium exceeds 7.0 mmol/litre. The
intermittent haemodialysis, which are the mainstay of chronic
most serious effect is on cardiac conduction, which classically
renal replacement therapy. Hospitals with acute nephrology
presents as a shortened QT interval with a tall peaked T wave on
services will perform haemodialysis in haemodynamically stable
the electrocardiogram (ECG). Without treatment, progressive
patients, although RRT is most commonly performed acutely in a
lengthening of QRS duration and PR interval ultimately lead to a
level 2 or 3 facility employing haemodialysis or more commonly
‘sine wave’ appearance, followed by ventricular standstill or
continuous therapies such as haemofiltration or haemodiafiltra-
fibrillation. A variety of other conduction disturbances may
tion. However, more recently there has been a trend towards
occur, including bundle-branch block, bifascicular block and
using hybrid therapies such as prolonged intermittent renal
advanced atrioventricular block. Asymptomatic patients with
replacement therapy (PIRRT). Although superficially similar, the
serum potassium <6.5 mmol/litre, or whose ECG does not
mechanisms by which the composition of the blood is altered
manifest signs of hyperkalemia, should be treated with a low
differ markedly.11,12 In haemodialysis, blood flows along one
potassium diet; additional sources of potassium intake should be
side of a semipermeable membrane as a solution of crystalloids
withheld and any potentiating drugs discontinued. (Specific
(the dialysis fluid) is pumped against the direction of the blood
treatment of hyperkalaemia is set out in the article Assessment
flow along the other side of the membrane. Molecules diffuse
and initial management of acute kidney injury (Table 3) on pages
across the membrane from higher to lower concentrations driven
000e000 of this issue.)
by the law of mass action. The composition of the dialysis fluid
Acidosis allows as near normalization of the plasma as possible. The
Metabolic acidosis in AKI results from increased acid production, dialysis-fluid compartment is under lower pressure, generating a
increased acid retention and decreased renal reabsorption of bi- transmembrane pressure gradient enabling the removal of salt
carbonate. Acidosis is exacerbated by sepsis, malnutrition and and water. Haemofiltration in its simplest form involves the
some drugs, and is very common among critically ill patients and passage of blood under pressure passing down one side of a
acute admissions. Metabolic acidosis is easily detected by highly permeable membrane, allowing water and other mole-
measuring venous serum bicarbonate in the above patient groups cules up to a size of about 20 kDa to pass through the membrane
when requesting routine blood tests. (Specific treatment of by convection. Thus, filtrate is discarded and replaced by an
acidosis is set out is set out in the article Assessment and initial idealized buffered replacement fluid, the crystalloid components
management of acute kidney injury (Table 3) on pages 000e000 of which are at physiological concentration. Haemodiafiltration
of this issue.) as the name implies is a combination of the two therapies

MEDICINE --:- 2 Crown Copyright ! 2015 Published by Elsevier Ltd. All rights reserved.

Please cite this article in press as: Amery CE, et al., The management of acute kidney injury, Medicine (2015), http://dx.doi.org/10.1016/
j.mpmed.2015.05.015
ACUTE KIDNEY INJURY

offering theoretically enhanced clearances of some middle mol- mortality in patients with both AKI complicated by sepsis.
ecules. Indications for renal replacement depend on the clinical However, the IVOIRE (Impact of High-volume Venovenous
and biochemical derangements outlined above and, although Continuous Haemofiltration in the Early Management of Septic
these are general indications, use will be guided by the clinical Shock Patients with Acute Renal Failure) study, which compared
situation. In the intensive care unit, for example, treatment may patients treated with 70 ml/kg/h or 35 ml/kg/h, found no evi-
begin before the absolute indications are reached, as there is dence that HVHF leads to a reduction in mortality at 28 days or
uncertainty surrounding the optimal timing of the initiation of contributes to early improvements in organ function.20 A
renal support. There is some evidence that, in the critically ill
intensively managed patient, early treatment may result in
REFERENCES
improved outcome. However, a randomized controlled trial from
1 Bellomo R, Ronco C, Kellum JA, Mehta R, Palevsky P. the ADQI
India in 2013, using intermittent therapies in a non-ITU popula-
workgroup. Acute renal failureedefinition, outcome measures, ani-
tion, found that time to recovery of renal function was longer in
mal models, fluid therapy and information technology needs: the
the group with early initiation of dialysis.13 A large multicentre
Second International Consensus Conference of the Acute Dialysis
randomized controlled trial in critically ill patients is currently
Quality Initiative (ADQI) Group. Crit Care 2004; 8: R204e12.
recruiting participants.14
2 Mehta RL, Kellum JA, Shah SV, et al. Acute kidney injury network:
Practical aspects of renal replacement therapy report of an initiative to improve outcomes in acute kidney injury. Crit
RRT requires access to the circulation. Various double-lumen Care 2007; 11: R31.
vascular catheters are commercially available, allowing blood 3 Bellomo R, Kellum JA, Ronco C. Defining and classifying acute renal
flow rates up to 200 ml/minute. Adequate venous access is critical failure: from advocacy to consensus and validation of the RIFLE
in preventing technical difficulties with blood flow from the pa- criteria. Intensive Care Med 2007; 33: 409e13.
tient and potential problems with clotting, leading to a loss of the 4 Haase M, Bellomo R, Matalanis G, Calzavacca P, Dragun D, Haase-
extracorporeal circuit. In most cases anticoagulation is required, Fielitz A. A comparison of the RIFLE and acute kidney injury, network
with unfractionated heparin being commonly used to prime the classifications for cardiac surgery-associated acute kidney injury: a
circuits, and a continuous infusion is maintained through the prospective cohort study. J Thorac Cardiovasc Surg 2009; 138:
inflow side of the circuit. If the patient is thrombocytopenic, 1370e6.
alternative anticoagulants can be used, including sodium chlo- 5 KDIGO clinical practice guideline for acute kidney injury. Kidney Int
ride, citrate or prostacyclin, but care must be taken with the latter Supplements 2012; 2.
given its vasodilator effects. More recently, regional anti- 6 Shemesh O, Golbetz H, Kriss JP, et al. Limitations of creatinine as a
coagulation with citrate has increased in popularity with citrate filtration marker in glomerulopathic patients. Kidney Int 1985; 28:
acting as both an anticoagulant and a buffer. Citrate is adminis- 830e8.
tered as sodium citrate before the filter and chelates calcium ions. 7 Wilkes NJ, Woolf R, Mutch M, et al. The effects of balanced versus
The associated regional hypocalcemia in the filter inhibits the saline-based hetastarch and crystalloid solutions on acid base and
generation of thrombin and thereby the clotting process. Citrate is electrolyte status and gastric mucosal perfusion in elderly surgical
partially removed by filtration or dialysis and the remainder is patients. Anesth Analg 2001; 93: 811e6.
rapidly metabolized in the citric acid (Krebs) cycle e especially in 8 Uchino S, Doig GS, Bellomo R, et al. Diuretics and mortality in acute
the liver, muscle and renal cortex. The chelated calcium is renal failure. Crit Care Med 2004; 32: 1669e77.
released and the lost calcium replaced after the filter. Systemic 9 Kellum JA, Decker M. Use of dopamine in acute renal failure: a met-
coagulation is therefore unaffected. For adequate anticoagulation, aanalysis. Crit Care Med 2001; 29: 1526e31.
the citrate dose is adjusted to blood flow to attain an ionized 10 Mehta RL, Chertow GM. Diuretics in critically ill patients with acute
calcium concentration <0.4 mmol/litre in the filter; it follows that renal failure. JAMA 2003; 289: 1379e81.
the lower the calcium concentration, the higher the degree of 11 Forni LG, Hilton PJ. Continuous haemofiltration in the treatment of
anticoagulation, with various protocols being employed.15,16 acute renal failure. N Engl J Med 1997; 336: 1303e9.
In haemofiltration the flow rate, which reflects the rate of 12 D’Intini V, Ronco C, Bonello M, Bellomo R. Renal replacement therapy
ultrafiltrate produced and solute clearance, is taken as a surro- in acute renal failure. Best Pract Res Clin Anaesthesiol 2004; 18:
gate for the dose. In 2000 Ronco et al.17 demonstrated survival 145e57.
rates to be significantly lower in patients treated with ultrafil- 13 Jamale TE, Hase NK, Kulkarni M, et al. Earlier-start versus usual-start
tration rates of 20 mL/kg/h compared to 35 mL/kg/h. However dialysis in patients with community-acquired acute kidney injury: a
more recently the Randomised Evaluation of Normal versus randomized controlled trial. Am J Kidney Dis 2013; 62: 1116e21.
Augmented Level of renal replacement therapy in ICU (RENAL)18 14 Smith ON, Wald R, Adhikari N, et al. Standard versus accelerated
studied a dose of 35 mL/kg/h compared to 20 ml/kg/h and found initiation of renal replacement therapy in acute kidney injury
there to be no difference in mortality between the two groups at (STARRT-AKI): study protocol for a randomized controlled trial. Trials
90 days. The Acute Renal Failure Trial Network (ATN) demon- 2013; 14: 320.
strated similar findings when comparing the two doses and, 15 Joannidis Michael, Oudemans-van Straaten HM. Clinical review:
interestingly, recorded more hypotensive episodes in those patency of the circuit in continuous renal replacement therapy. Crit
receiving the higher dose.19 Care 2007; 11: 218.
High-volume haemofiltration (HVHF) defined as a treatment 16 Oudemans-van Straaten HM, Osterman M. Bench-to-bedside review:
dose of greater then 50 ml/kg/h has been cited as a potential citrate for continuous renal replacement therapy, from science to
method of reducing cytokine levels and thereby reducing practice. Crit Care 2012; 16: 249.

MEDICINE --:- 3 Crown Copyright ! 2015 Published by Elsevier Ltd. All rights reserved.

Please cite this article in press as: Amery CE, et al., The management of acute kidney injury, Medicine (2015), http://dx.doi.org/10.1016/
j.mpmed.2015.05.015
ACUTE KIDNEY INJURY

17 Ronco C, Bellomo R, Homel P, et al. Effects of different doses in 19 The VA/NIH Acute Renal Failure Trial Network. Intensity of renal
continuous veno-venous haemofiltration on outcomes of acute support in critically ill patients with acute kidney injury. N Engl J Med
renal failure: a prospective randomised trial. Lancet 2000; 356: 2008; 359: 7e20.
26e30. 20 The IVORE study group. High-volume versus standard-volume hae-
18 Bellomo R, Cass A, Cole L, et al. Intensity of continuous renal- mofiltration for septic shock patients with acute kidney injury (IVOIRE
replacement therapy in critically ill patients (RENAL study). N Engl J study): a multicentre randomized controlled trial. Intensive Care Med
Med 2009; 361: 1627e38. 2013; 39: 1535e46.

MEDICINE --:- 4 Crown Copyright ! 2015 Published by Elsevier Ltd. All rights reserved.

Please cite this article in press as: Amery CE, et al., The management of acute kidney injury, Medicine (2015), http://dx.doi.org/10.1016/
j.mpmed.2015.05.015
ARTIGO 3

SÍNDROMES GLOMERULARES
1 Síndromes
glomerulares
Juliano Sacramento Mundim ® Viktoria Woronik

} Introdução
O acometimento glomerular pode ocorrer tanto em doenças sistêmicas,
situação na qual a glomerulopatia é dita secundária (nefrite lúpica, nefro-
esclerose diabética, etc.), como em doenças originárias do rim, situação
na qual a glomerulopatia é dita primária.
Independentemente do mecanismo da lesão envolvido, a lesão glomeru-
lar é acompanhada por alterações na função, estrutura e permeabilidade do
glomérulo, levando à passagem anormal de proteínas e de outros elementos
e à redução variável na função de filtração glomerular, além de ocasionar
distúrbios na excreção de sódio e água.
Em qualquer uma das formas, primária ou secundária, a lesão ao gloméru-
lo se manifesta por meio de alguns sintomas e sinais clínicos, listados a seguir:

} proteinúria: decorrente do aumento de permeabilidade glomerular;


} hematúria: decorrente da inflamação da parede capilar;
} elevação de ureia e creatinina: decorrente da queda no ritmo de filtração
glomerular;
} oligúria ou anúria: a depender da intensidade da inflamação glomerular
e queda do ritmo de filtração glomerular;
} hipertensão: decorrente, habitualmente, da retenção de fluido (sal e
água) pelo rim.

De forma geral, o achado de sinais e sintomas sugestivos de doença


glomerular requer, do ponto de vista diagnóstico, a realização de biópsia
renal. Há algumas poucas situações, entretanto, nas quais a biópsia renal
não é necessária. A síndrome nefrótica em crianças pode prescindir da re-
alização inicial de biópsia renal, uma vez que a doença de lesões mínimas
14 Silvia Titan

é preponderante nesta faixa etária, permitindo o início de tratamento com


corticoide mesmo sem a realização da biópsia. Na síndrome nefrítica aguda
com características clínicas e laboratoriais muito sugestivas de glomerulo-
nefrite pós-estreptocócica, particularmente se em situação de epidemia, a
biópsia pode ser indicada apenas naqueles casos de evolução desfavorável. A
biópsia renal também não costuma ser necessária em pacientes com quadro
clínico sugestivo de nefropatia diabética, ou seja, naqueles com diabetes de
longa data e com sinais de lesão em outros órgãos-alvo. A biópsia também
não está rotineiramente indicada em pacientes assintomáticos com doença
glomerular leve (p. ex., hematúria isolada), dado o prognóstico favorável
desse tipo de lesão.
Em todas as outras situações, a biópsia renal é imperativa. Vale notar
que as doenças glomerulares apresentam, muitas vezes, curso insidioso, o
que dificulta o diagnóstico precoce. Assim, o médico deve estar atento ao
achado de sinais sugestivos de glomerulopatias, indicando prontamente a
avaliação diagnóstica e a conduta terapêutica pertinentes.
As síndromes clínicas glomerulares podem ser classificadas como mos-
tra a Tabela 1.1. Cada uma das síndromes glomerulares será abordada em
capítulo específico neste livro.

} Achados clínicos
Hematúria microscópica assintomática
Caracteriza-se pelo achado de hematúria isolada ao exame de urina,
portanto sem a presença de proteinúria, alteração da função renal ou
manifestações sistêmicas de edema e hipertensão. É um achado comum,
pois ocorre em 5 a 10% da população, sendo, em sua maioria, de causa
urológica. Em pacientes acima de 40 anos de idade com micro-hematúria
persistente isolada sem evidência de origem glomerular, a cistoscopia é
obrigatória para excluir a malignidade uroepitelial.
Entre as hematúrias isoladas, apenas 10% ou menos são causadas
por glomerulopatias. Portanto, a investigação urológica é obrigatória, e
o achado de hemácias dismórficas, quando presente, sugere o diagnóstico
de glomerulopatia. O protocolo clínico de avaliação da biópsia renal em
adultos com hematúria microscópica assintomática (excluída causa uro-
lógica) mostrou rim normal em 30%; doença de membrana fina em 26%;
Princípios Básicos de Nefrologia 15

glomerulopatia por IgA em 28%; e, no restante, glomerulonefrite membra-


noproliferativa e síndrome de Alport.

TABELA 1.1. SÍNDROMES CLÍNICAS E DOENÇAS (HISTOPATOLOGIA)

Hematúria microscópica assinto-


mática

Hematúria macroscópica recor-


rente

Síndrome nefrítica
-

GN rapidamente progressiva (GN


crescêntica)

Proteinúria assintomática

Síndrome nefrótica

Doença renal crônica estágios IV e V

Hematúria macroscópica recorrente


Nesta circunstância, o paciente relata o achado de urina de tom mar-
rom, sendo rara a presença de coágulos. A hematúria macroscópica deve
ser distinguida de outras causas de urina marrom ou vermelha, incluindo
hemoglobinúria, mioglobinúria, porfiria, consumo de comidas com coran-
tes, particularmente beterraba, e consumo de medicamentos ou outras
substâncias, em particular a rifampicina. A hematúria macroscópica requer
avaliação urológica, incluindo cistoscopia em qualquer idade, a menos que
a história seja característica de hematúria glomerular.
16 Silvia Titan

A hematúria recorrente é, em geral, episódica, relacionada a infecções


do trato respiratório e/ou exercício físico, autolimitada, ocorrendo predo-
minantemente em adolescentes e adultos jovens. As causas mais frequentes
são nefropatia por IgA, glomerulopatia da membrana basal fina, assim como
síndrome de Alport e suas variantes.
Na nefropatia por IgA, a hematúria é franca, episódica e ocorre geral-
mente um dia após infecção respiratória de vias áreas superiores. Difere,
portanto, da manifestação de glomerulonefrite difusa aguda (GNDA), na
qual a hematúria macroscópica ocorre duas a três semanas após o quadro
infeccioso, geralmente de etiologia estreptocócica.

Glomerulonefrite aguda ou síndrome nefrítica


As doenças que produzem inflamação glomerular aguda são aquelas que
se exteriorizam de forma mais exuberante com síndrome nefrítica, carac-
terizada por edema, hipertensão, hematúria e graus variáveis de insufici-
ência renal, além de proteinúria pouco intensa (< 3 g/dia). São chamadas
de difusas quando o acometimento glomerular ocorre em mais de 50% do
total de glomérulos da biópsia, e denominadas focais quando há menos de
50% de glomérulos acometidos. Neste grande grupo de doenças, as causas
imunológicas são as mais comuns, como nefrite lúpica, nefropatia por IgA
e glomerulonefrite difusa aguda pós-infecciosa. Para o diagnóstico das glo-
merulopatias mais comuns que se exteriorizam sob a forma nefrítica, alguns
aspectos clínico-sorológicos mais importantes estão listados na Tabela 1.2.

TABELA 1.2. TESTES SOROLÓGICOS E ASSOCIAÇÕES COM SÍNDROME NEFRÍTICA


Doença Associação Teste sorológico
Glomerulonefrite pós-estreptocócica ASLO
Endocardite
Shunt Hidrocefalia tratada
História
Glomerulopatia por IgA
Lúpus eritematoso sistêmico rash malar

A apresentação clássica da síndrome nefrítica é vista na glomerulone-


frite difusa aguda pós-estreptocócica em crianças. As crianças geralmente
apresentam oligúria, ganho de peso e edema generalizado de apresentação
Princípios Básicos de Nefrologia 17

aguda (poucos dias). O exame de urina revela proteinúria, hematúria e, co-


mumente, a presença de cilindros hemáticos. A proteinúria costuma ocorrer
em valor não nefrótico e a albumina sérica é geralmente normal. O volume
circulante está aumentado, causando hipertensão e, eventualmente, edema
pulmonar sem evidência de doença cardíaca primária.

Glomerulonefrite rapidamente progressiva


Aqui as manifestações são as de síndrome nefrítica, mas associadas
à perda rápida da função renal (dias ou semanas). A lesão histológica
responsável pela natureza agressiva da lesão é a crescente glomerular.
A crescente decorre da proliferação das células epiteliais da cápsula de
Bowman, promovida pelo intenso processo inflamatório que ocorre no
glomérulo, sendo o macrófago uma célula particularmente importante em
sua gênese. Quando há mais de 50% de glomérulos acometidos por cres-
centes, o diagnóstico histopatológico é de glomerulonefrite crescêntica,
que se expressa clinicamente por perda rápida e intensa da função renal.
Portanto, existe correlação clínica entre número de crescentes e gravidade
da doença. Assim, doenças com mais de 80% de crescentes se exteriorizam,
geralmente, por insuficiência renal dialítica. A causa mais comum dessa
forma de glomerulonefrite crescêntica é a vasculite. Doenças de imuno-
complexos, como nefropatia por IgA e nefrite lúpica, expressam-se mais
comumente com número menor de crescentes e, portanto, quadro clínico
renal mais brando. A resolução (involução) dos crescentes depende, entre
outros fatores, da sua idade histológica, ou seja, sua natureza epitelial ou
fibroblástica. No entanto, outros fatores também participam desse processo,
já que crescentes da mesma “idade” histológica em nefrite lúpica revertem
mais facilmente ao tratamento do que os de nefropatia por IgA.
São três os grupos mais frequentes de doenças que se apresentam como
glomerulonefrite rapidamente progressiva: glomerulonefrite antimembrana
basal glomerular, glomerulonefrite por imunocomplexos e glomerulonefrite
relacionada a ANCA. As formas relacionadas a ANCA podem se apresentar
com manifestações sistêmicas de Wegener, poliangeíte microscópica e
Churg-Strauss, ou ter manifestação exclusivamente renal. A forma relacio-
nada ao anticorpo antimembrana basal glomerular pode se apresentar como
doença de Goodpasture ou sem sinais de comprometimento pulmonar. As
18 Silvia Titan

doenças de imunocomplexos mais comumente encontradas são a nefrite


lúpica, a crioglobulinemia (sendo a forma mais comum à secundária ao
vírus da hepatite C) e a nefropatia por IgA. Para o diagnóstico das doenças
glomerulares que se apresentam sob a forma rapidamente progressiva, os
aspectos clínico e sorológico mais importantes estão na Tabela 1.3.

TABELA 1.3. DOENÇAS GLOMERULARES QUE SE APRESENTAM COMO GLOMERULONEFRITE RAPIDAMENTE PROGRESSIVA
Doença Associação Teste sorológico
Doença de Goodpasture Hemorragia alveolar
Granulomatose de Wegener ANCA C (citoplasmático)
Poliangeíte microscópica Envolvimento multissistêmico ANCA P (perinuclear)
Vasculite pauci-imune Somente envolvimento renal ANCA P (perinuclear)
Lúpus eritematoso sistêmico Envolvimento sistêmico
Glomerulonefrite pós-estreptocócica Faringite/impetigo
Nefropatia por IgA/púrpura de
Henoch-Schönlein
Endocardite

Proteinúria assintomática
É definida quando existe proteinúria isolada em valores acima de 150 mg/dia
e abaixo de 3 g/dia na ausência de outros achados urinários, como hematúria,
e, também, sem sinais ou sintomas sistêmicos, como edema e/ou hipertensão.
É, portanto, um diagnóstico feito por exame de urina. As doenças mais
frequentes neste grupo são a glomeruloesclerose segmentar e focal (GESF)
e a glomerulonefrite membranosa (Tab. 1.1), de evolução benigna, a me-
nos que mudem suas características clínicas com o desenvolvimento de
hipertensão ou proteinúrias nefróticas.
A microalbuminúria é definida como a excreção de 30 a 300 mg de
albumina/dia (equivalente à relação albumina/creatinina urinária entre
0,03 e 0,3). Este parâmetro também é utilizado para identificar o risco de
desenvolvimento de nefropatia em pacientes diabéticos, assim como risco
cardiovascular em pacientes hipertensos.
O achado de outras proteínas urinárias, que não a albumina, tem signi-
ficado fisiopatológico próprio. Assim, a proteinúria tubular, constatada pelo
achado urinário de beta-2-microglobulina, retinol binding protein (RBP) ou
de outras proteínas de baixo peso molecular, é característica de doenças
Princípios Básicos de Nefrologia 19

com comprometimento tubulointersticial, seja de natureza primária ou


secundária à glomerulopatias (p. ex., GESF). Além de seu papel diagnóstico
em relação ao tipo de proteinúria presente, os marcadores de proteinúria
tubular são clinicamente úteis como marcadores de mau prognóstico em
diversas doenças, como ocorre na própria GESF e no transplante renal.
A proteinúria encontrada em estados de hiperprodução de proteínas, parti-
cularmente cadeias leves de globulinas, filtradas pelo glomérulo, é característica
de paraproteinemias e deve ser pesquisada ativamente, já que as proteínas
não são detectadas por testes laboratoriais que detectam somente albumina.
O achado de pequenas quantidades de albumina também pode ocorrer
em certas situações funcionais, não patológicas, como febre ou estados hi-
peradrenérgicos, sendo denominada de proteinúria funcional, que também
pode ser desencadeada pela posição ortostática e deambulação, quando é
chamada de ortostática.
Quanto à indicação de biópsia renal em proteinúria assintomática, geral-
mente, ela não está indicada, sobretudo se a proteinúria for inferior a 1 g/dia.

Síndrome nefrótica
É uma síndrome clínico-laboratorial decorrente do aumento de per-
meabilidade às proteínas plasmáticas, caracterizando-se por proteinúria
acima de 3,5 g/1,73 m2 de superfície corpórea/dia, com consequente hi-
poalbuminemia e edema.
O achado de hiperlipidemia não é obrigatório, porém é muito comum,
assim como os distúrbios relacionados à hipercoagulabilidade por perda
de fatores inibidores de coagulação, a desnutrição proteica e a suscetibili-
dade às infecções. Dentre as glomerulopatias que mais causam síndrome
nefrótica estão, entre as primárias, a glomerulopatia de lesões mínimas,
GESF e glomerulonefrite membranosa, e, entre as secundárias, a glome-
ruloesclerose diabética (Tab. 1.1). As doenças clínicas mais comuns que
se apresentam como síndrome nefrótica estão na Tabela 1.4.

Doença renal crônica (estágios IV e V)


A maioria das glomerulopatias, com exceção da glomerulopatia de lesões
mínimas e da nefropatia da membrana basal fina, pode evoluir para lesão
glomerular crônica esclerosante, com manifestações clínicas de insuficiência
20 Silvia Titan

renal. Estatísticas brasileiras do Ministério da Saúde mostram que as glo-


merulonefrites são, no momento, a segunda causa de doença renal crônica
terminal no país, atrás apenas da hipertensão arterial.

Doença Teste sorológico


Nenhum
GESF Sorologia para HIV
-
nosa neoplasia FAN/anti-DNA
-
proliferativa ou normais
- -

Amiloidose Imunoeletroforese no soro e na urina


Nenhum

AINH = anti-inflamatório não hormonal; GESF = glomeruloesclerose segmentar e focal;


LES = lúpus eritematoso sistêmico.

} Diferenças clínico-laboratoriais entre


síndromes nefrítica e nefrótica
O diagnóstico clínico das síndromes nefrítica e nefrótica frequentemente
não é fácil, pela superposição de sintomas e sinais entre as duas síndromes.
No entanto, como orientação diagnóstica, algumas diferenças podem ser
encontradas na Tabela 1.5.

Sintoma/sinal Síndrome nefrótica Síndrome nefrítica


Instalação Insidiosa
Edema ++++ ++
Pressão arterial Normal Alta
Proteinúria ++++ + ou ++
Hematúria - ou + +++
Normal

} Glomerulopatias e síndromes nefrítica e


nefrótica
É sabido que o diagnóstico histológico de uma glomerulopatia não
permite, via de regra, inferir quanto à sua exteriorização nefrítica ou ne-
Princípios Básicos de Nefrologia 21

frótica. Assim, apenas a glomerulopatia de lesões mínimas se exterioriza na


forma nefrótica sempre. As doenças glomerulares que mais se aproximam
de formas nefríticas puras são a GNDA e a glomerulonefrite crescêntica,
como descrito na Tabela 1.6.

TABELA 1.6. ASPECTOS NEFRÍTICOS E NEFRÓTICOS EM DOENÇAS GLOMERULARES

Doenças glomerulares Aspectos nefróticos Aspectos nefríticos


Glomerulopatia de lesão mínima ++++ -
++++ +
GESF +++ ++
+++ ++
Glomerulopatia proliferativa mesangial ++ ++
++ +++
GNDA + ++++
Glomerulonefrite crescêntica + ++++

} Glomerulopatias primárias e frequência de


distribuição
Dados provenientes do Registro Paulista de Glomerulopatias (RPG)
mostram que, no estado de São Paulo, as biópsias renais de 1.844 pacientes
foram indicadas por:

} síndrome nefrótica em 41,5% dos casos;


} hematúria associada a proteinúria em 27,2%;
} doença renal crônica em 8,9%;
} síndrome nefrítica-nefrótica em 7,9%;
} glomerulonefrite rapidamente progressiva em 7,1%;
} hematúria isolada em apenas 1,9% dos casos.

Outras casuísticas podem apresentar perfis diferentes quanto a sintomas


clínicos indicativos de biópsia, acarretando inclusive diferentes distribui-
ções de glomerulopatias. É conhecido que, no Japão, onde a indicação de
biópsia renal por hematúria isolada é mais ampla, a frequência de glome-
rulopatia por IgA é maior.
22 Silvia Titan

Ainda em relação à casuística do RPG, apenas em glomerulopatias


primárias (n = 1.131), o perfil de distribuição é o seguinte:

} GESF: 29,7%;
} glomerulonefrite membranosa: 20,7%;
} glomerulopatia por IgA: 17,8%;
} glomerulopatia de lesões mínimas: 9,1%;
} glomerulonefrite membranoproliferativa: 7%;
} glomerulonefrite crescêntica: 4,1%;
} glomerulonefrite proliferativa não IgA: 3,8%;
} GNDA: 2,5% e outras.

Os dados da Tabela 1.7 mostram uma comparação entre nossa casuística


e a de outros países.

TABELA 1.7. FREQUÊNCIA DAS VÁRIAS FORMAS DE DOENÇAS GLOMERULARES PRIMÁRIAS EM DIFERENTES PAÍSES

Brasil n: Itália n: Romênia China n:


1.131 (%) 6.990 (%) Tcheca n: 9.278 (%)
2.333 (%)
GESF 6

Nefropatia por IgA

- 7
ferativa
Glomerulonefrite crescêntica
Glomerulonefrite mesangial não IgA -- --

Outras 7

} Tópicos importantes
} O acometimento glomerular pode ocorrer tanto em doenças sistêmicas
(glomerulopatia secundária), como em doenças originárias do rim (glo-
merulopatia primária).
} Geralmente o achado de doença glomerular implica a realização de bi-
ópsia renal. Há situações, entretanto, nas quais a biópsia renal não é ne-
Princípios Básicos de Nefrologia 23

cessária, por exemplo crianças com síndrome nefrótica isolada (doença


de lesões mínimas é o diagnóstico mais provável, permitindo o início de
tratamento com corticoide mesmo sem a realização da biópsia), em pa-
cientes com síndrome nefrítica aguda com características muito sugestivas
de glomerulonefrite pós-estreptocócica e que estejam evoluindo favoravel-
mente e em pacientes com quadro clínico muito sugestivo de nefropatia
diabética. A biópsia também não está rotineiramente indicada em pacien-
tes assintomáticos com doença glomerular leve (p. ex., hematúria isolada.

} Leituras sugeridas
Barros RT, Sens YAS. Propedêutica das glomerulopatias. In: Barros RT, Alves MAR,
Dantas M, Kirsztajn GM, Sens YAS. Glomerulopatias: patogenia, clínica e trata-
mento. 2. ed. São Paulo: Sarvier; 2005.
Carvalho MFC, Franco MF, Soares VA. Glomerulonefrites primarias. In: Riella MC.
Princípios de nefrologia e distúrbios hidroeletrolíticos. 4. ed. Rio de Janeiro: Gua-
nabara Koogan; 2003.
Covic A, Schiller A, Volovat C, Gluhovschi G, Gusbeth-Tatomir P, Petrica L, et al.
Epidemiology of renal disease in Romania: a 10 year review of two regional renal
biopsy databases. Nephrol Dial Transplant. 2006;21(2):419-24.
Feehally J, Johnson RJ. Introduction to glomerular disease: clinical presentations.
In: Feehally J, Floege J, Johnson RJ. Comprehensive clinical nephrology. 3rd ed.
Philadelphia: Mosby; 2007.
Gesualdo L, Di Palma AM, Morrone LF, Strippoli GF, Schena FP. The Italian ex-
perience of the national registry of renal biopsies. Kidney Int. 2004;66(3):890-4.
Haas M, Meehan SM, Karrison TG, Spargo BH. Changing etiologies of unexplained
adult nephrotic syndrome: a comparison of renal biopsy findings from 1976-1979
and 1995-1997. Am J Kidney Dis. 1997;30(5):621-31.
Jennette JC, Falk RJ. Glomerular clinicopathologic syndromes. In: Greenberg A. Primer
on kidney diseases. 3rd ed. San Diego: Academic; 2001. p. 129-43.
Li LS, Liu ZH. Epidemiologic data of renal diseases from a single unit in China: analysis
based on 13,519 renal biopsies. Kidney Int. 2004;66(3):920-3.
Malafronte P, Mastroianni-Kirsztajn G, Betônico GN, Romão Jr JE, Alves MA, Carvalho
MF, et al. Paulista registry of glomerulonephritis: 5-year data report. Nephrol Dial
Transplant. 2006;21(11):3098-105.
Meyer TW. Tubular injury in glomerular disease. Kidney Int. 2003;63:774-87.
Morales JV. Glomerulopatias. In: Barros E, Manfro RC, Thomé FS, Gonçalves LFS.
Nefrologia: rotinas, diagnóstico e tratamento. 3. ed. Porto Alegre: Artmed; 2006.
v. 1, p. 189-212.
24 Silvia Titan

Nachman PH, Jennette JC, Falk RJ. Primary glomerular disease. In: Brenner BM. Bren-
ner and Rector’s the kidney. 8th ed. Philadelphia: Saunders Elsevier; 2008. cap. 30.
Oliveira MB, Saldanha LB, Mota EL, Penna DO, Barros RT, Romão Junior JE. Primary
glomerular diseases in Brazil (1978-1999): is the frequency of focal and segmental
glomeruloesclerosis increasing? Clin Nephrol. 2004;61:90-7.
Rychlík I, Jancová E, Tesar V, Kolsky A, Lácha J, Stejskal J, et al. The Czech registry
of renal biopsies. Occurrence of renal diseases in the years 1994-2000. Nephrol
Dial Transplant. 2004;19(12):3040-9.
Wilmer WA, Rovin BH, Hebert CJ, Rao SV, Kumor K, Hebert LA. Management of
glomerular proteinuria: a commentary. J Am Soc Nephrol. 2003;14:3217-32.
ARTIGO 4

ACESSO VASCULAR
Saugel et al. Critical Care (2017) 21:225
DOI 10.1186/s13054-017-1814-y

REVIEW Open Access

Ultrasound-guided central venous catheter


placement: a structured review and
recommendations for clinical practice
Bernd Saugel1* , Thomas W. L. Scheeren2 and Jean-Louis Teboul3

Abstract
The use of ultrasound (US) has been proposed to reduce the number of complications and to increase the safety
and quality of central venous catheter (CVC) placement. In this review, we describe the rationale for the use of US
during CVC placement, the basic principles of this technique, and the current evidence and existing guidelines for
its use. In addition, we recommend a structured approach for US-guided central venous access for clinical practice.
Static and real-time US can be used to visualize the anatomy and patency of the target vein in a short-axis and a
long-axis view. US-guided needle advancement can be performed in an "out-of-plane" and an "in-plane" technique.
There is clear evidence that US offers gains in safety and quality during CVC placement in the internal jugular vein.
For the subclavian and femoral veins, US offers small gains in safety and quality. Based on the available evidence
from clinical studies, several guidelines from medical societies strongly recommend the use of US for CVC
placement in the internal jugular vein. Data from survey studies show that there is still a gap between the existing
evidence and guidelines and the use of US in clinical practice. For clinical practice, we recommend a six-step
systematic approach for US-guided central venous access that includes assessing the target vein (anatomy
and vessel localization, vessel patency), using real-time US guidance for puncture of the vein, and confirming the
correct needle, wire, and catheter position in the vein. To achieve the best skill level for CVC placement the
knowledge from anatomic landmark techniques and the knowledge from US-guided CVC placement need to be
combined and integrated.
Keywords: Central venous access, Ultrasound, Internal jugular vein, Subclavian vein, Femoral vein, Short axis,
Long axis, Out of plane, In plane

Background technique, and the current evidence and existing guide-


Although placement of a central venous catheter (CVC) lines for its use. In addition, we recommend a structured
is a routine procedure in intensive care medicine and approach for US-guided central venous access for clinical
anesthesiology, acute severe complications (such as practice.
arterial puncture or cannulation, hematoma, hemotho-
rax, or pneumothorax) occur in a relevant proportion
Rationale for ultrasound-guided central venous
of patients [1, 2]. The use of ultrasound (US) has been
catheter placement
proposed to reduce the number of CVC complications
Traditionally, CVC placement is performed using land-
and to increase the safety and quality of CVC placement.
mark techniques based on the knowledge of anatomic
In this review, we describe the rationale for the use of
structures and palpation of arteries next to the veins.
US during CVC placement, the basic principles of this
These landmark techniques cannot account for anatomic
* Correspondence: bcs.muc@gmx.de; bernd.saugel@gmx.de variations at the CVC insertion site. Anatomic variations
1
Department of Anesthesiology, Center of Anesthesiology and Intensive Care to the "normal anatomy", however, have been described
Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, in a relevant proportion of patients for the internal jugular
20246 Hamburg, Germany
Full list of author information is available at the end of the article vein (IJV), the subclavian vein (SV), and the femoral vein
(FV) [3–11]. In addition to anatomic variations, venous
© The Author(s). 2017 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Saugel et al. Critical Care (2017) 21:225 Page 2 of 11

thrombosis that is especially common in oncologic and the US screen (i.e., a cross-sectional image of the vessel).
critically ill patients can make CVC placement impossible A "long-axis" view (i.e., a longitudinal image of the ves-
or dangerous for the patient [9]. sel) is obtained by placing the US probe in a parallel
The described anatomic variations and the presence of position relative to the course of the vessel. Short-axis
venous thrombosis can hardly be identified using a land- and long-axis views can be used for both US assistance
mark technique. In contrast, US can be used to easily and guidance of CVC placement. Of note, the terms
visualize anatomic structures and confirm patency of the "out-of-plane" and "in-plane" describe the direction of the
vein and thus help to avoid unintended arterial puncture needle relative to the US plane, refer to US-guided needle
or unsuccessful cannulation. In addition, US can facili- advancement, and should not be mixed up with the terms
tate CVC placement in special clinical situations in "short-axis" and "long-axis".
which landmark techniques based on palpation of the For real-time US guidance, different US approaches
arterial pulse are challenging or impossible (e.g., femoral can be used. US guidance during needle advancement
CVC placement during cardiopulmonary resuscitation can be performed using: a short-axis probe orientation
[12] or in patients with a nonpulsatile ventricular assist and an out-of-plane view of the needle (Fig. 1a); a long-
device). axis probe orientation and an in-plane view of the needle
(Fig. 1b); or a so-called oblique orientation [15]. It is im-
Ultrasound for central venous catheter placement: portant to understand that the user needs to align the
basic principles and techniques US plane and the needle plane containing the needle
Ultrasound probe that appears on the screen as a point (short-axis/out-
US probes best suited for CVC placement are small linear of-plane) or an echogenic line (long-axis/in-plane)
array probes with high-frequency transducers (5–15 MHz) with ring-down artifacts [14].
[13]. These probes usually have a scanning surface of Whether or not one approach is superior to the other
about 20–50 mm and allow high-resolution imaging of cannot be answered rigorously based on the existing
superficial anatomic structures [13]. 2D imaging (comple- data. The advantage of the short-axis/out-of-plane view is
mented by Doppler US functions) is currently the stand- that it allows better visualization of the vein in relation to
ard technique used for US-guided central venous access the artery and other anatomic structures, and thus might
[13]. All US probes have an index mark (a small physical more sufficiently help to avoid accidental arterial puncture
notch on one side of the probe) that corresponds with an [15]. The short-axis/out-of-plane approach is easier to
orientation marker on one side of the US scan sector learn for physicians not familiar with US [16]. Among ex-
shown on the US device screen and thus helps to obtain perienced US users, the short-axis/out-of-plane approach
the correct probe orientation during US examination. seems to result in a higher success rate with the first at-
Preferably, US machines should have the ability to record tempt for CVC placement in the IJV and SV [17, 18].
and save US images and loops for clinical documentation However, in the short-axis view, the needle is only visual-
(and teaching purposes) [13]. ized as an echogenic point (that must not necessarily be
the tip of the needle). In contrast, when using the long-
Ultrasound techniques for central venous catheter axis/in-plane view, the entire needle in its complete course
placement and the depth of the needle tip can be visualized on the
US can be used in different ways to facilitate CVC place- US image, thus reducing the risk of penetration of the
ment. "Static" US (also called indirect US) describes a posterior vessel wall [15, 19].
technique using US only before CVC placement to iden- Combining advantages of both techniques, the oblique
tify the anatomy of the target vein and adjacent anatomic axis view (a view that is halfway between the short-axis
structures (including the patency of the vein and its and the long-axis view with the US probe placed at
dimensions and depth from the skin) [14]. This ap- approximately 45° with respect to the target vessel) can
proach of preprocedural US evaluation is also referred be used by experienced US users [20, 21].
to as "US-assisted" CVC placement.
In contrast, "real-time" US (also called direct US) Can ultrasound make central venous catheter placement
describes a technique of needle advancement and vessel safer? What is the evidence?
puncture under permanent US control (i.e., the needle is The use of US to reduce the number of complications
permanently visualized on the US screen). This is also related to vascular access for CVC placement has been
referred to as "US guidance" [14]. evaluated in numerous previous studies in a variety of
clinical settings. Recent Cochrane systematic reviews and
Short-axis/long-axis and out-of-plane/in-plane views meta-analyses summarize the current evidence for US
The US probe can be placed in a transverse position guidance versus anatomic landmark techniques for CVC
relative to the vessel, resulting in a "short-axis" view on placement in the IJV [22], SV [23], and FV [23] with
Saugel et al. Critical Care (2017) 21:225 Page 3 of 11

Fig. 1 Ultrasound probe orientation and view of the needle. Ultrasound guidance during needle advancement can be performed using a short-
axis probe orientation and an out-of-plane view of the needle (a) or a long-axis probe orientation and an in-plane view of the needle (b)

regard to complications of CVC placement. These meta- low for most outcome measures and the heterogeneity
analyses included adult and pediatric patients treated in among the studies was high.
the intensive care unit or the operating room and com- For the SV, a meta-analysis including nine studies with
pared conventional landmark techniques with techniques 2030 patients showed that the use of US resulted in a re-
using static or real-time US or Doppler US. The primary duced rate of accidental arterial puncture (US, 2/242
outcome measure was the total rate of peri-interventional (0.8%) vs landmark, 15/256 (5.9%); risk ratio (95% CI)
complications and adverse events. 0.21 (0.06–0.82)) and hematoma formation (US, 3/242
For the IJV, 35 trials enrolling a total of 5108 patients (1.2%) vs landmark, 17/256 (6.6%); risk ratio (95% CI) 0.26
were included in the meta-analysis [22]. The analysis (0.09–0.76)) [23]. However, no statistically significant
demonstrated that the use of US for CVC placement in difference was found between the use of US and the
the IJV reduces the total rate of complications compared conventional landmark technique with regard to the total
with conventional landmark techniques (US, 48 compli- complication rate, the overall success rate, the number of
cations in 1212 patients (4.0%) vs landmark, 161/1194 attempts until success, the time to successful cannulation,
(13.5%); risk ratio (95% confidence interval (CI)) 0.29 and the success rate with the first attempt [23].
(0.17–0.52)). The overall success rate was higher when For CVC placement in the FV, the use of US com-
US was used (US, 2120/2172 (97.6%) vs landmark, 1900/ pared with the landmark technique increased the overall
2168 (87.6%); risk ratio (95% CI) 1.12 (1.08–1.17)) [22]. success rate (US, 134/150 (89.0%) vs landmark, 127/161
In addition, the use of US resulted in a decrease in the (78.9%); risk ratio (95% CI) 1.11 (1.00–1.23)) and the
rate of arterial puncture, hematoma formation, and success rate with the first attempt (US, 91/107 (85.0%)
number of attempts and time until successful cannula- vs landmark, 57/117 (48.7%); risk ratio (95% CI) 1.73
tion, and in an increase in the success rate with the first (1.34–2.22)) [23].
attempt of puncture [22]. The benefits of US-guided or Although the use of US offers small gains in safety and
US-assisted CVC placement with regard to the total quality, the authors conclude that the meta-analysis does
complication rate, overall success rate, and number of not generally support the use of US for CVC placement
attempts until success were consistent across experi- in the SV and FV [23].
enced and inexperienced operators. Thus, this meta- On behalf of the Canadian Perioperative Anesthesia
analysis clearly provides evidence that US offers gains Clinical Trials Group, Lalu et al. [24] performed a
in safety and quality during CVC placement in the systematic review and meta-analysis of US-guided SV
IJV. The quality of the evidence, however, was very catheterization. Based on data from 10 studies (including
Saugel et al. Critical Care (2017) 21:225 Page 4 of 11

2168 patients; six real-time US studies, one static US recommendation (based on Level A evidence) that "US-
study, three Doppler US studies), the authors revealed guided vascular access has to be used because it results
that US reduced the overall complication rate compared in clinical benefits and reduced overall costs of care
with the landmark technique (odds ratio (95% CI) 0.53 makes it cost-effective” [13].
(0.41–0.69)). Real-time US particularly reduced acci- The guidelines for the appropriate use of bedside gen-
dental arterial puncture, pneumothorax, and hematoma eral and cardiac US from the American College of Critical
formation. Care Medicine [26] give a strong (1-A) recommendation
A CVC via the SV can be placed using either an infra- for the general use of US for central venous access in real-
clavicular (most commonly used) or a supraclavicular time technique (1-B) using a short-axis approach (1-B).
approach. To the best of our knowledge there are no Regarding the site for CVC placement, the guidelines give
randomized controlled trials on US-guided CVC place- a strong (1-A) recommendation for the IJV and the FV,
ment via the SV comparing the supraclavicular and the but a conditional recommendation (2-C) for the SV [26].
infraclavicular approach. The supraclavicular approach A guideline from the European Federation of Societies
(using different US probes) needs to be evaluated in fu- for Ultrasound in Medicine and Biology (EFSUMB) [9]
ture studies. also recommends pre-interventional US vessel screening
When discussing the evidence for US during CVC of target vessels to determine the most appropriate ana-
placement at the different anatomic sites based on the tomical site and the optimal patient position (5-D) and
available studies and meta-analyses, one needs to keep routine real-time US guidance during CVC placement
in mind that—compared to the IJV—it might be more (1-A) [9].
challenging to prove the advantages of US for CVC In 2016, the Association of Anaesthetists of Great
placement in the SV, because the ultrasound approach is Britain and Ireland [27] also recommended the routine
technically more challenging, and in the FV, because se- use of US for CVC placement in the IJV. In addition,
vere complications other than arterial puncture occur the expert group recommends US use "for all other
infrequently. central venous access sites, but recognizes evidence is,
at present, limited" [27]. Nevertheless, the recommen-
Guidelines for ultrasound-guided central venous dation also underlines that the understanding of the
catheter placement landmark technique is necessary for situations when
Various recommendations and guidelines with different US is not available.
clinical scopes and for different target audiences have
been published during the last years.
In 2012, a joint guideline from the American Society Use of ultrasound for central venous catheter
of Echocardiography and the Society of Cardiovascular placement in clinical practice
Anesthesiologists [15] strongly recommended the use of Several survey studies evaluated the attitudes and beliefs
real-time US for CVC placement in the IJV (category A, of intensivists and anesthesiologists on the use of US for
level 1 evidence), while it was not recommended for CVC placement and the frequency of its use in clinical
the SV (category A, level 3 evidence). For the FV, no practice.
recommendation for routine use of US was made be- In 2008, McGrattan et al. [28] performed a survey
cause of insufficient scientific evidence (category C, among 2000 senior anesthesiologists in the United King-
level 2 evidence). dom and revealed that only 27% of these stated using
A practice guideline from the American Society of US as the first-choice approach for CVC placement in
Anesthesiologists task force, also in 2012 [25], recom- the IJV (50% used the surface landmark technique and
mended the use of static US imaging in elective situa- 30% palpation of the carotid artery as first-choice
tions for prepuncture identification of the anatomy and approaches).
to evaluate the vessel localization and patency and real- Among emergency physicians in the United States,
time US for venipuncture for the IJV. Further, it is recom- 44% stated in 2014 that they never use US to guide CVC
mended that both static and real-time US "may" be used placement [10]. On the other hand, 20% and 9% of
for CVC placement in the SV or FV [25]. respondents stated using US in at least 90% and 100% of
For CVC placement in critically ill patients treated in cases, respectively.
the intensive care unit, an international expert panel rec- A survey among 784 intensivists in the United States
ommended in 2012 the routine use of US for short-term performed in 2016 [29] revealed a moderate to very
and long-term central venous access in adults [13]. More frequent use of US depending on the site for CVC
specifically, the panel recommended the utilization of placement ranging from 31% for the SV to 80% for the
2D US imaging with a long-axis/in-plane technique for IJV (45% for the FV). Barriers to the use of US re-
vascular access [13] and agreed on the very strong ported by these respondents were limited availability
Saugel et al. Critical Care (2017) 21:225 Page 5 of 11

of US equipment (28%), perception of increased time


for US-guided CVC insertion (22%), and concerns about a
losing skills for the landmark technique (13%) [29].
Among 190 French intensivists, a practice survey [30]
reported high rates of US use for CVC placement in
2016, with 18% and 50% of physicians always or almost
always, respectively, using an US-guided CVC technique
(6% never, 10% almost never, 17% half of the time).
Interestingly, a higher proportion of residents compared
with senior doctors stated always or at least almost al-
ways using US.

How to perform ultrasound-guided central venous b


catheter placement? Recommendations for clinical
practice—a systematic approach
For clinical practice, we recommend a systematic ap-
proach including the following steps:

I. Identify anatomy of the insertion site and


localization of the vein.
II. Confirm patency of the vein.
III.Use real-time US guidance for puncture of the vein.
IV.Confirm needle position in the vein.
V. Confirm wire position in the vein.
VI.Confirm catheter position in the vein. Fig. 2 Ultrasound views to identify the anatomy of the target vein.
Short-axis (transverse) view (a) and long-axis (longitudinal) view (b)
of the right internal jugular vein (*) and its anatomic relation to the
Identify anatomy of the insertion site and localization of carotid artery (#)
the vein
As a first step, one should use US to identify the anatomy Confirm patency of the vein
of the insertion site (vein and artery, adjacent anatomic By applying pressure to the vein and thus testing its
structures) and the localization of the target vein. This compressibility with the US probe, one can confirm the
includes checking for anatomic variations of the vessels patency of the vein and thus exclude venous thrombosis.
(both vein and artery) and the localization of the vein in Of note, in patients with very low arterial blood pressure
relation to the artery. This step requires combining a pro- (systolic arterial pressure < 60 mmHg), the artery might
found knowledge about anatomic structures and landmarks also be compressible [14].
with the competencies required for US-guided CVC place-
ment (such as knowledge about probe orientation and
image display, converting the 2D US image into 3D real-
ity, and hand–eye coordination) [31]. Given the variability
in anatomic structures, this first step of US assessment is
best performed before prepping and draping of the punc-
ture site and the US probe.
The location of the vein and its anatomic relation to
the artery is best identified when using both a short-axis
(transverse) and a long-axis (longitudinal) view of the
vessels (Fig. 2a, b). This also allows identifying hypoplas-
tic veins or underfilling of the veins due to intravascular
hypovolemia (Fig. 3). To exactly differentiate between
venous and arterial vessels one can additionally perform Fig. 3 Ultrasound view of a small internal jugular vein. Short-axis
color Doppler imaging and apply Doppler flow measure- (transverse) view of a small right internal jugular vein (*) and
its anatomic relation to the carotid artery (#) (e.g., in a patient
ments to derive venous and arterial Doppler flow pro-
with intravascular hypovolemia)
files (Fig. 4a, b).
Saugel et al. Critical Care (2017) 21:225 Page 6 of 11

body gown; covering the US probe and cable with a sterile


cover/shield; and using a sterile conductive medium
(US gel) [13, 32].
The position of the operator performing US-guided
CVC placement should be such that he/she has the in-
sertion site, the needle, and the US screen in their line
of sight during needle insertion [13]. Usually, the oper-
ator should hold the US probe with the nondominant
hand while advancing the needle with the dominant
hand. This approach is referred to as the "single-oper-
ator technique" and allows the operator to optimally
align the US plane and the direction of the needle.
These practical aspects of US-guided CVC placement
are illustrated in Fig. 5.
While advancing the needle, its tip should be con-
stantly identified with US during the needle approach to
the vein and puncture of the vein. This can be done
using a short-axis/out-of-plane view or a long-axis/in-
plane view.

Confirm needle position in the vein


The use of real-time US then allows confirmation that
the needle tip is placed centrally in the vein before ap-
proaching the guide wire (Fig. 6a, b).

Confirm wire position in the vein


As a next step after wire advancement, the correct pos-
ition of the guide wire should be confirmed in both a
short-axis and a long-axis US view (Fig. 6c, d).

Fig. 4 Color Doppler imaging and Doppler flow measurements.


Short-axis (transverse) view of the right internal jugular vein (blue)
and the carotid artery (red) using color Doppler imaging and
Doppler flow measurements of the venous (a) and arterial (b)
blood flow profile (Color figure online)

To further confirm the patency of the vein and to


quantify venous and arterial blood flow, color Doppler
imaging and Doppler flow measurements should be Fig. 5 Practical aspects of ultrasound-guided central venous catheter
performed (Fig. 4a, b). placement in the internal jugular vein using the "single-operator
technique”. An aseptic approach including covering the puncture
site with a large sterile drape, using sterile barriers (hat, mask, sterile
Use real-time ultrasound guidance for puncture of the vein gloves, sterile body gown), and covering the ultrasound probe and
CVC placement should be performed using US guidance. cable with a sterile cover is shown. The position of the operator
This requires an aseptic approach to avoid catheter-related (who holds the ultrasound probe with the nondominant hand while
bloodstream infections. An aseptic technique includes: advancing the needle with the dominant hand) allows aligning the
insertion site, the needle, and the ultrasound screen in the line of
prepping and covering the puncture site with a large sterile
sight during needle insertion (red lines) (Color figure online)
drape; wearing a hat, a mask, sterile gloves, and a sterile
Saugel et al. Critical Care (2017) 21:225 Page 7 of 11

Fig. 6 Ultrasound to confirm needle, wire, and catheter position in the vein. Ultrasound images during real-time ultrasound-guided central venous
catheter placement in the right internal jugular vein. Ultrasound guidance should include confirmation of the needle position in the vein before
approaching the guide wire (short-axis/out-of-plane view (a) and long-axis/in-plane view (b)). In addition, the correct position of the guide wire in the
vein (short-axis (c) and long-axis (d)) and the correct position of the catheter in the vein (short-axis (e) and long-axis (f)) should be confirmed

Confirm catheter position in the vein information on the effect of different interventions on
Finally, after placement of the CVC over the guide wire, the venous puncture sites.
the correct position of the CVC in the vein can be It has been demonstrated repeatedly that positioning of
visualized with US, again in a short-axis and a long-axis the patient in a head-down (Trendelenburg) position in-
view (Fig. 6e, f ). creases the filling and thus the cross-sectional lumen of the
Figure 7 summarizes the six-step approach to US-guided IJV [33]. On the contrary, to increase the lumen of the FV,
CVC insertion. patients can be positioned in a head-up (reverse Trende-
lenburg) position [34]. Positioning of the leg in an
abducted and externally rotated position also can help to
How to integrate knowledge from landmark and maximize the cross-sectional diameter of the FV [35].
ultrasound techniques? For the IJV, imaging studies showed that the position
To achieve the personal best skill level for CVC place- of the head plays an important role in optimizing the
ment, it is crucial that one combines and integrates the conditions during CVC placement. Several studies dem-
anatomic knowledge from landmark techniques and the onstrated that rotation of the head to the opposite side
knowledge gained from US-guided vascular access (know- increases the overlap of the IJV and the carotid artery
ledge about image display and converting the 2D image [36–38]. In a US study, Miki et al. [37] investigated in 30
into 3D reality, and hand–eye coordination) [31]. In volunteers the anatomical relationship between the IJV
this context, previous US studies provided important and the carotid artery during head rotation. The overlap
Saugel et al. Critical Care (2017) 21:225 Page 8 of 11

I. Identify anatomy of insertion site and localization of the vein

• Identify vein, artery, anatomic structures


• Check for anatomic variations
• Use short axis (transverse; A) and long
axis (longitudinal; B) view
• Perform this step before prepping and
draping of the puncture site

II. Confirm patency of the vein

• Use compression ultrasound to exclude Compression


venous thrombosis
• Use color Doppler imaging and Doppler
flow measurements to confirm the patency of
the vein and to quantify blood flow

III. Use real-time US guidance for puncture of the vein

• Use an aseptic approach


• Use a short axis/out-of-plane (A) or a long
axis/in-plane (B) approach
• Try to constantly identify the tip of the
needle during the needle approach to the
vein and puncture of the vein

IV. Confirm needle position in vein

• Confirm that the needle tip is placed


centrally in the vein before approaching the
guide wire

V. Confirm wire position in vein

• Confirm the correct position of the guide


wire in a short axis (A) and a long axis (B)
view

VI. Confirm catheter position in vein

• Confirm the correct position of the central


venous catheter in the vein in a short axis
(A) and a long axis (B) view

Fig. 7 Six-step approach to ultrasound-guided central venous catheter placement

of the IJV and the carotid artery gradually increased with aligning the angle between the US and the needle
increasing rotation of the head to the left. In parallel, plane so that the two planes intersect at the depth of
however, the flattening of the IJV decreased with head the vessel selected for cannulation [40]. Especially for
rotation to the left. DeAngelis et al. [39] described that inexperienced users [41], the needle guides help to
the IJV becomes more vertically separated from the ca- guide the needle along the path of the US beam at the
rotid artery at more extreme angles of contralateral head correct angle and distance depending on the depth of
rotation. These findings underline that US should be the targeted structure. Needle guides facilitate faster
used in each individual patient to assess the optimal cannulation for IJV CVCs (only for inexperienced op-
angle of head rotation and best approach to the IJV. erators) [42] and SV CVCs [40]. Nevertheless, in a
simulation model study, a needle guide used in a long-
Technical developments in the field of axis vessel approach improved needle visualization but
ultrasound-guided vascular access did not improve puncture of the target vessel compared
Needle guides are devices placed on the US transducer with a free-hand technique [43]. Altogether, based on the
that might improve the cannulation success by facilitating contradicting evidence [43, 44], no rigorous conclusion
Saugel et al. Critical Care (2017) 21:225 Page 9 of 11

about the clinical value of needle guide devices can comparison to the IJV and FV, the anatomic location
currently be drawn. and course of the SV under the clavicle bone can be
Different real-time 3D techniques (sometimes referred more difficult to visualize using US. Smaller US probes
to as 4D US with time being the fourth dimension) for can facilitate US-guided access to the SV [48, 49]. Of
US-guided CVC placement have been described [45, 46]. note, the use of US to puncture the SV results in a
Lower image resolution, larger US probe dimensions, and puncture site that is usually more lateral compared to
artifacts making needle visualization difficult, however, are the landmark puncture technique. The close proximity
still major limitations of this innovative concept [46]. of the vessels and the pleura must be kept in mind also
during US-guided puncture of the SV. Because the
Limitations of ultrasound-guided central venous angle of cannulation is usually steeper when using US, it is
catheter placement especially important to align and constantly visualize the
Although US is noninvasive and thus does not bear a needle to avoid pleural injury.
risk to directly harm the patient, some limitations and
disadvantages of US during central venous access are Conclusion
worth considering. US guidance can improve patient safety and procedural
One might argue that the risk of catheter-related quality during CVC placement in the IJV, FV, and SV.
bloodstream infections might be higher if US is used for Based on evidence from clinical studies, several guide-
CVC placement without applying a strict aseptic approach lines of medical societies strongly recommend the use of
as already described [47]. In addition, an insufficient num- US for CVC placement in the IJV. Data from survey
ber of US machines in a certain unit (intensive care unit studies show that there is still a gap between the existing
or anesthesia induction area) might cause procedural evidence and guidelines and the use of US in clinical
delays [47]. Moreover, it is expensive to purchase and practice. We recommend a six-step systematic approach
maintain US machines and to provide adequate training for US-guided central venous access. To achieve the best
for all operators involved in CVC placement [47]. skill level for CVC placement the knowledge from ana-
US might give the inexperienced user a false sense of tomic landmark techniques and the knowledge from
security and mislead him/her to neglect traditionally US-guided CVC placement need to be combined and
taught principles with regard to needle direction. It is key integrated.
to visualize the needle (or needle tip) constantly during
needle advancement to avoid accidental arterial puncture, Abbreviations
CVC: Central venous catheter; FV: Femoral vein; IJV: Internal jugular vein;
posterior wall penetration, or pneumothorax. In addition, SV: Subclavian vein; US: Ultrasound
rapid movements with the needle during "searching the
needle on the US screen" must be avoided rigorously. To Acknowledgements
The authors thank Oliver Diener, Maximilian Leistenschneider, and Elisabeth
overcome these problems related to insufficient US skills von Heckel for their support in obtaining the US images and the photographs
and to ensure high-quality care, formal education and illustrating the procedure for US-guided CVC placement.
training (including simulation) with a structured certifica-
Funding
tion of US skills for vascular access and the development None.
of a consensus standard for these training programs has
been suggested [13]. Availability of data and materials
Moreover, concerns have been expressed that routine Not applicable.
US use will result in a "de-skilling" with regard to the Authors’ contributions
landmark techniques because these techniques will not BS, TWLS, and J-LT conceived the article, performed the literature search,
be taught and practiced anymore, thus resulting in higher drafted the manuscript, and read and approved the final manuscript.
complication rates when CVCs need to be placed when
Ethics approval and consent to participate
US is not available (e.g., in emergencies) [47]. Not applicable.
Besides these general limitations, different problems
specific for the different anatomical sites for CVC place- Consent for publication
Written informed consent for publication of their ultrasound images was
ment might occur during US-guided CVC placement. In obtained from the patients and volunteers. A copy of the consent form is
patients with a shorter neck anatomy, the long-axis US available for review by the Editor of this journal.
view of the IJV might be difficult to obtain. Although
Competing interests
the FV can usually be visualized easily using US in adults, The authors declare that they have no competing interests.
in severely obese patients a second operator might be
necessary to provide access to the inguinal region. In
Publisher’s Note
addition, a curved-array abdominal US probe can be Springer Nature remains neutral with regard to jurisdictional claims in
necessary for visualizing deeper anatomic structures. In published maps and institutional affiliations.
Saugel et al. Critical Care (2017) 21:225 Page 10 of 11

Author details 18. Vezzani A, Manca T, Brusasco C, Santori G, Cantadori L, Ramelli A, Gonzi G,
1
Department of Anesthesiology, Center of Anesthesiology and Intensive Care Nicolini F, Gherli T, Corradi F. A randomized clinical trial of ultrasound-
Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, guided infra-clavicular cannulation of the subclavian vein in cardiac surgical
20246 Hamburg, Germany. 2Department of Anesthesiology, University of patients: short-axis versus long-axis approach. Intensive Care Med. 2017.
Groningen, University Medical Centre Groningen, Groningen, The doi: 10.1007/s00134-017-4756-6. [Epub ahead of print]
Netherlands. 3Service de Réanimation Médicale Hôpital de Bicêtre, Hôpitaux 19. Stone MB, Moon C, Sutijono D, Blaivas M. Needle tip visualization during
Universitaires Paris-Sud, AP-HP, Le Kremlin-Bicêtre, France. ultrasound-guided vascular access: short-axis vs long-axis approach.
Am J Emerg Med. 2010;28:343–7.
20. Phelan M, Hagerty D. The oblique view: an alternative approach for
ultrasound-guided central line placement. J Emerg Med. 2009;37:403–8.
21. Wilson JG, Berona KM, Stein JC, Wang R. Oblique-axis vs. short-axis view in
References ultrasound-guided central venous catheterization. J Emerg Med. 2014;47:45–50.
1. McGee DC, Gould MK. Preventing complications of central venous 22. Brass P, Hellmich M, Kolodziej L, Schick G, Smith AF. Ultrasound guidance
catheterization. N Engl J Med. 2003;348:1123–33. versus anatomical landmarks for internal jugular vein catheterization.
2. Merrer J, De Jonghe B, Golliot F, Lefrant JY, Raffy B, Barre E, Rigaud JP, Cochrane Database Syst Rev. 2015;1:Cd006962.
Casciani D, Misset B, Bosquet C, Outin H, Brun-Buisson C, Nitenberg G. 23. Brass P, Hellmich M, Kolodziej L, Schick G, Smith AF. Ultrasound guidance
Complications of femoral and subclavian venous catheterization in critically versus anatomical landmarks for subclavian or femoral vein catheterization.
ill patients: a randomized controlled trial. JAMA. 2001;286:700–7. Cochrane Database Syst Rev. 2015;1:Cd011447.
3. Hoffman T, Du Plessis M, Prekupec MP, Gielecki J, Zurada A, Shane Tubbs R, 24. Lalu MM, Fayad A, Ahmed O, Bryson GL, Fergusson DA, Barron CC, Sullivan P,
Loukas M. Ultrasound-guided central venous catheterization: a review of the Thompson C. Ultrasound-guided subclavian vein catheterization: a systematic
relevant anatomy, technique, complications, and anatomical variations. review and meta-analysis. Crit Care Med. 2015;43:1498–507.
Clin Anat. 2017;30:237–50. 25. Rupp SM, Apfelbaum JL, Blitt C, Caplan RA, Connis RT, Domino KB, Fleisher LA,
4. Gordon AC, Saliken JC, Johns D, Owen R, Gray RR. US-guided puncture of Grant S, Mark JB, Morray JP, Nickinovich DG, Tung A. Practice guidelines for
the internal jugular vein: complications and anatomic considerations. central venous access: a report by the American Society of Anesthesiologists
J Vasc Interv Radiol. 1998;9:333–8. Task Force on Central Venous Access. Anesthesiology. 2012;116:539–73.
5. Turba UC, Uflacker R, Hannegan C, Selby JB. Anatomic relationship of 26. Frankel HL, Kirkpatrick AW, Elbarbary M, Blaivas M, Desai H, Evans D,
the internal jugular vein and the common carotid artery applied to Summerfield DT, Slonim A, Breitkreutz R, Price S, Marik PE, Talmor D, Levitov A.
percutaneous transjugular procedures. Cardiovasc Intervent Radiol. Guidelines for the Appropriate Use of Bedside General and Cardiac
2005;28:303–6. Ultrasonography in the Evaluation of Critically Ill Patients—Part I: General
6. Denys BG, Uretsky BF. Anatomical variations of internal jugular vein location: Ultrasonography. Crit Care Med. 2015;43:2479–502.
impact on central venous access. Crit Care Med. 1991;19:1516–9. 27. Bodenham Chair A, Babu S, Bennett J, Binks R, Fee P, Fox B, Johnston AJ,
7. Benter T, Teichgraber UK, Kluhs L, Papadopoulos S, Kohne CH, Felix R, Klein AA, Langton JA, McLure H, Tighe SQ. Association of Anaesthetists of
Dorken B. Anatomical variations in the internal jugular veins of cancer Great Britain and Ireland: safe vascular access 2016. Anaesthesia. 2016;71:573–85.
patients affecting central venous access. Anatomical variation of the internal 28. McGrattan T, Duffty J, Green JS, O'Donnell N. A survey of the use of ultrasound
jugular vein. Ultraschall Med. 2001;22:23–6. guidance in internal jugular venous cannulation. Anaesthesia. 2008;63:1222–5.
8. Docktor B, So CB, Saliken JC, Gray RR. Ultrasound monitoring in cannulation 29. Soni NJ, Reyes LF, Keyt H, Arango A, Gelfond JA, Peters JI, Levine SM,
of the internal jugular vein: anatomic and technical considerations. Adams SG, Restrepo MI. Use of ultrasound guidance for central venous
Can Assoc Radiol J. 1996;47:195–201. catheterization: a national survey of intensivists and hospitalists.
9. Jenssen C, Brkljacic B, Hocke M, Ignee A, Piscaglia F, Radzina M, Sidhu PS, J Crit Care. 2016;36:277–83.
Dietrich CF. EFSUMB Guidelines on Interventional Ultrasound (INVUS). Part 30. Maizel J, Bastide MA, Richecoeur J, Frenoy E, Lemaire C, Sauneuf B, Dupont H,
VI—Ultrasound-Guided Vascular Interventions. Ultraschall Med. 2016;37:473–6. Tamion F, Nseir S, Du Cheyron D. Practice of ultrasound-guided central venous
10. Buchanan MS, Backlund B, Liao MM, Sun J, Cydulka RK, Smith-Coggins R, catheter technique by the French intensivists: a survey from the BoReal study
Kendall J. Use of ultrasound guidance for central venous catheter placement: group. Ann Intensive Care. 2016;6:76.
survey from the American Board of Emergency Medicine Longitudinal Study of 31. Weiner MM, Geldard P, Mittnacht AJ. Ultrasound-guided vascular access:
Emergency Physicians. Acad Emerg Med. 2014;21:416–21. a comprehensive review. J Cardiothorac Vasc Anesth. 2013;27:345–60.
11. Beaudoin FL, Merchant RC, Lincoln J, Gardiner F, Liebmann O, Cohn J. 32. Marhofer P, Fritsch G. Sterile working in ultrasonography: the use of
Bedside ultrasonography detects significant femoral vessel overlap: dedicated ultrasound covers and sterile ultrasound gel. Expert Rev Med
implications for central venous cannulation. CJEM. 2011;13:245–50. Devices. 2015;12:667–73.
12. Hilty WM, Hudson PA, Levitt MA, Hall JB. Real-time ultrasound-guided 33. Mallory DL, Shawker T, Evans RG, McGee WT, Brenner M, Parker M, Morrison G,
femoral vein catheterization during cardiopulmonary resuscitation. Mohler P, Veremakis C, Parrillo JE. Effects of clinical maneuvers on
Ann Emerg Med. 1997;29:331. sonographically determined internal jugular vein size during venous
13. Lamperti M, Bodenham AR, Pittiruti M, Blaivas M, Augoustides JG, Elbarbary M, cannulation. Crit Care Med. 1990;18:1269–73.
Pirotte T, Karakitsos D, Ledonne J, Doniger S, Scoppettuolo G, Feller-Kopman D, 34. Stone MB, Price DD, Anderson BS. Ultrasonographic investigation of the
Schummer W, Biffi R, Desruennes E, Melniker LA, Verghese ST. International effect of reverse Trendelenburg on the cross-sectional area of the femoral
evidence-based recommendations on ultrasound-guided vascular access. vein. J Emerg Med. 2006;30:211–3.
Intensive Care Med. 2012;38:1105–17. 35. Randall C, Schmeiser E, Fiers E, Little A, Dogbey G, Richardson G.
14. Dietrich CF, Horn R, Morf S, Chiorean L, Dong Y, Cui XW, Atkinson NS, Ultrasound investigation of leg position to enhance femoral vein
Jenssen C. Ultrasound-guided central vascular interventions, comments on exposure for cannulation. J Emerg Med. 2014;47:176–81.
the European Federation of Societies for Ultrasound in Medicine and Biology 36. Sulek CA, Gravenstein N, Blackshear RH, Weiss L. Head rotation during
guidelines on interventional ultrasound. J Thorac Dis. 2016;8:E851–68. internal jugular vein cannulation and the risk of carotid artery puncture.
15. Troianos CA, Hartman GS, Glas KE, Skubas NJ, Eberhardt RT, Walker JD, Anesth Analg. 1996;82:125–8.
Reeves ST. Special articles: guidelines for performing ultrasound guided 37. Miki I, Murata S, Nakazawa K, Onozawa S, Mine T, Ueda T, Yamaguchi H,
vascular cannulation: recommendations of the American Society of Yasui D, Takeda M, Kumita S. Anatomical relationship between the common
Echocardiography and the Society Of Cardiovascular Anesthesiologists. carotid artery and the internal jugular vein during head rotation.
Anesth Analg. 2012;114:46–72. Ultrasound. 2014;22:99–103.
16. Blaivas M, Brannam L, Fernandez E. Short-axis versus long-axis approaches 38. Troianos CA, Kuwik RJ, Pasqual JR, Lim AJ, Odasso DP. Internal jugular vein
for teaching ultrasound-guided vascular access on a new inanimate model. and carotid artery anatomic relation as determined by ultrasonography.
Acad Emerg Med. 2003;10:1307–11. Anesthesiology. 1996;85:43–8.
17. Chittoodan S, Breen D, O'Donnell BD, Iohom G. Long versus short axis 39. DeAngelis V, Denny J, Chyu D, Jan T, Lemaire A, Chiricolo A, Solina A.
ultrasound guided approach for internal jugular vein cannulation: The optimal angle of head rotation for internal jugular cannulation as determined
a prospective randomised controlled trial. Med Ultrason. 2011;13:21–5. by ultrasound evaluation. J Cardiothorac Vasc Anesth. 2015;29:1257–60.
Saugel et al. Critical Care (2017) 21:225 Page 11 of 11

40. Maecken T, Heite L, Wolf B, Zahn PK, Litz RJ. Ultrasound-guided


catheterisation of the subclavian vein: freehand vs needle-guided
technique. Anaesthesia. 2015;70:1242–9.
41. Jaffer U, Normahani P, Singh P, Aslam M, Standfield NJ. Randomized study
of teaching ultrasound-guided vascular cannulation using a phantom and
the freehand versus needle guide-assisted puncture techniques.
J Clin Ultrasound. 2015;43:469–77.
42. Augoustides JG, Horak J, Ochroch AE, Vernick WJ, Gambone AJ, Weiner J,
Pinchasik D, Kowalchuk D, Savino JS, Jobes DR. A randomized controlled
clinical trial of real-time needle-guided ultrasound for internal jugular
venous cannulation in a large university anesthesia department.
J Cardiothorac Vasc Anesth. 2005;19:310–5.
43. Ball RD, Scouras NE, Orebaugh S, Wilde J, Sakai T. Randomized, prospective,
observational simulation study comparing residents' needle-guided vs
free-hand ultrasound techniques for central venous catheter access.
Br J Anaesth. 2012;108:72–9.
44. Luyet C, Hartwich V, Urwyler N, Schumacher PM, Eichenberger U, Vogt A.
Evaluation of a novel needle guide for ultrasound-guided phantom vessel
cannulation. Anaesthesia. 2011;66:715–20.
45. Dowling M, Jlala HA, Hardman JG, Bedforth NM. Real-time three-dimensional
ultrasound-guided central venous catheter placement. Anesth Analg.
2011;112:378–81.
46. French JL, Raine-Fenning NJ, Hardman JG, Bedforth NM. Pitfalls of
ultrasound guided vascular access: the use of three/four-dimensional
ultrasound. Anaesthesia. 2008;63:806–13.
47. Hessel 2nd EA. Con: we should not enforce the use of ultrasound as a
standard of care for obtaining central venous access. J Cardiothorac Vasc Anesth.
2009;23:725–8.
48. Kim SC, Graff I, Sommer A, Hoeft A, Weber S. Ultrasound-guided
supraclavicular central venous catheter tip positioning via the right
subclavian vein using a microconvex probe. J Vasc Access. 2016;17:435–9.
49. Lanspa MJ, Fair J, Hirshberg EL, Grissom CK, Brown SM. Ultrasound-guided
subclavian vein cannulation using a micro-convex ultrasound probe.
Ann Am Thorac Soc. 2014;11:583–6.
ARTIGO 5

HIPERCALEMIAS
review www.kidney-international.org

Treatment of hyperkalemia: something old,


something new
Richard H. Sterns1,2, Marvin Grieff1,2 and Paul L. Bernstein1,2
1
Division of Nephrology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA;
and 2Department of Medicine, Rochester General Hospital, Rochester, New York, USA

T
Treatment options for hyperkalemia have not changed reatment options for hyperkalemia have not changed
much since the introduction of the cation exchange resin, much since the introduction of the cation exchange
sodium polystyrene sulfonate (Kayexalate, Covis resin, sodium polystyrene sulfonate ([SPS]; Kayexalate,
Pharmaceuticals, Cary, NC), over 50 years ago. Although Covis Pharmaceuticals, Cary, NC), over 50 years ago.1–4
clinicians of that era did not have ready access to Although clinicians of that era did not have ready access to
hemodialysis or loop diuretics, the other tools that we use hemodialysis or loop diuretics, the other tools that we use
today—calcium, insulin, and bicarbonate—were well today—calcium, insulin, and bicarbonate—were well known
known to them. Currently recommended insulin regimens to them.5,6 In recent years, our comfort with traditional
provide too little insulin to achieve blood levels with a therapies has been shaken by warnings that Kayexalate mixed
maximal kalemic effect and too little glucose to avoid with sorbitol may be harmful, and by a growing realization
hypoglycemia. Short-acting insulins have theoretical that many of our standard treatments for hyperkalemia have
advantages over regular insulin in patients with severe little evidence to support them.4,7–9 The coming year is likely
kidney disease. Although bicarbonate is no longer to see the release of 2 new pharmaceutical products, providing
recommended for acute management, it may be useful in clinicians with new therapeutic weapons for their arsenal.10
patients with metabolic acidosis or intact kidney function. This review is intended to weigh the available evidence on
Kayexalate is not effective as acute therapy, but a new both new and old treatments for hyperkalemia.
randomized controlled trial suggests that it is effective
when given more chronically. Gastrointestinal side effects Confirming the diagnosis
and safety concerns about Kayexalate remain. New When any degree of hyperkalemia is discovered, the accuracy
investigational potassium binders are likely to be approved of the measurement must be verified. A repeat serum po-
in the coming year. Although there are some concerns tassium concentration is often normal, without therapy,
about hypomagnesemia and positive calcium balance from because of distribution or excretion of recently ingested po-
patiromer, and sodium overload from ZS-9 (ZS Pharma, tassium, diurnal variation, or laboratory error.11–14 Pseudo-
Coppell, TX), both agents have been shown to be effective hyperkalemia (a falsely high potassium), caused by poor
and well tolerated when taken chronically. ZS-9 shows phlebotomy technique, hemolysis, laboratory processing,
promise in the acute treatment of hyperkalemia and may thrombocytosis, and leukocytosis, can lead to inappropriate
make it possible to avoid or postpone the most effective intervention.15 The serum potassium rises with exercise and
therapy, emergency hemodialysis. falls after.16 Because contractions of forearm muscles release
Kidney International (2016) 89, 546–554; http://dx.doi.org/10.1016/ intracellular potassium, fist clenching during phlebotomy
j.kint.2015.11.018 raises both serum and plasma potassium by as much as
KEYWORDS: cation exchange resins; hyperkalemia; insulin; potassium; renal 1 mmol/l.17–20 Potassium is released from platelets during
dialysis; sodium bicarbonate clotting, raising the serum but not plasma potassium in pa-
ª 2016 International Society of Nephrology tients with thrombocytosis. To exclude pseudohyperkalemia,
plasma potassium (obtained from a heparinized sample) or
whole blood potassium should be measured, if platelet counts
exceed 500,000.15,21 Leukemic lymphocytes are fragile and
release potassium during centrifugation, when exposed to
high concentrations of heparin in the test tube, or when
shaken by pneumatic tube transport. In patients with lym-
phocytic leukemia, the potassium concentration can be higher
in plasma than in serum; this observation led to the term
Correspondence: Richard H. Sterns, Rochester General Hospital, 1425 Port- “reverse pseudohyperkalemia” to contrast it with the previ-
land Avenue, Rochester, New York 14621, USA. E-mail: richard.sterns@ ously reported pseudohyperkalemia caused by thrombocy-
rochesterregional.org tosis (in which the potassium concentration in serum is
Received 17 September 2015; revised 23 October 2015; accepted 11 higher than in plasma).15 When the potassium concentration
November 2015; published online 2 February 2016 is falsely elevated because of mechanical fragmentation of

546 Kidney International (2016) 89, 546–554


RH Sterns et al.: Treatment of hyperkalemia review

lymphocytes, both serum and plasma potassium are rhythm should be balanced against the potential adverse
affected.22–24 To avoid confusion, we suggest the terms effect of intravenous calcium in the presence of digoxin
“platelet-induced serum pseudohyperkalemia,” “lymphocyte- toxicity.
induced plasma pseudohyperkalemia,” and “shaken-lympho-
cyte pseudohyperkalemia.” To exclude lymphocyte-induced Promoting uptake of potassium by cells
plasma pseudohyperkalemia, serum potassium or whole Skeletal muscle is the reservoir for more than 70% of body
blood potassium from a sample drawn in a blood gas syringe potassium. Transport of extracellular potassium into muscle
(which contains lower concentrations of heparin) should be cells in exchange for intracellular sodium, by the membrane-
measured; if shaken-lymphocyte pseudohyperkalemia is sus- bound sodium pump, sodium-potassium adenosine triphos-
pected, samples should be hand-carried to the laboratory.22–24 phatase, serves as the primary extrarenal mechanism for
achieving potassium homeostasis, with a calculated maximal
The electrocardiogram in hyperkalemia transport rate of 134 mmol/min—enough to transfer one-half
Hyperkalemia decreases the transmembrane potassium of the potassium normally residing in the extracellular space
gradient leading to increased potassium conductance, and this (or the potassium absorbed in a large meal) within 15 sec-
shortens the duration of the action potential.25 As potassium onds. Insulin, beta-2 agonists, and bicarbonate accelerate the
rises to 5.5 to 6.5 mmol/l, peaked T-waves and a prolonged movement of potassium into muscle cells, and these agents
PR segment may be seen, advancing with higher levels of are widely used to treat “severe” hyperkalemia.
potassium to progressive widening of the QRS complex, Insulin. When insulin binds to its receptor on skeletal
fascicular and bundle branch blocks, a “sine-wave” appear- muscle, the abundance and activity of sodium-potassium
ance, and asystole.26–29 adenosine triphosphatase and the abundance of the glucose
The electrocardiogram is insensitive in assessing the transporter, GLUT4, on the cell membrane increase through
severity of hyperkalemia.30 Profound hyperkalemia can occur independent signaling pathways (reviewed in Ho39). Thus,
without electrocardiographic manifestations.31–34 Cardiac while the glycemic response is maximal at insulin levels of
conduction defects, most commonly severe bradycardia, can approximately 100 mU/ml, the kalemic effect of the hormone
be the presenting manifestation of hyperkalemia and hyper- continues to increase as insulin levels rise. Studies utilizing the
kalemia can cause malfunction of pacemakers and implant- euglycemic insulin clamp technique show that infusion of
able cardioverter-defibrillators.35,36 Abnormalities include regular insulin at 20 U/h after a 6.6-U priming dose in a 70-kg
widening of the QRS complex, increased pacing thresholds, healthy subject will rapidly raise insulin levels to approxi-
which can lead to failure to capture, as well as oversensing of mately 500 mU/ml, with a near maximal kalemic effect; to
the paced or spontaneous T-wave by the implantable maintain euglycemia at these insulin levels, infusion of
cardioverter-defibrillator and potentially inappropriate glucose at 40 g/h is required.40,41 Although uremia and type-2
shocks.36 diabetes cause resistance to the glycemic effect, insulin’s
ability to enhance potassium uptake by skeletal muscle and
Intravenous calcium liver are unimpaired.42,43
Calcium antagonizes the effects of hyperkalemia at the The most commonly recommended regimen for emer-
cellular level through effects on the threshold potential and gency treatment of severe hyperkalemia is a bolus intravenous
the speed of impulse propagation.25 In 1964, Chamberlain37 injection of 10 U of regular insulin, which, if blood glucose
reported 5 patients with serum potassium concentrations is <250 mg/dl, is given with a bolus injection of 25 g of
ranging from 8.6 to 10 mmol/l, illustrating “immediate” glucose (50 ml of a 50% solution).7,44,45 This regimen and
(within 5 minutes) resolution of the most advanced electro- others have been studied under standardized conditions in
cardiographic findings after intravenous calcium. Our several small trials of stable, mildly hyperkalemic patients
knowledge of when to use this intervention, or what dose and with dialysis-dependent kidney disease.43,46–57 Although in-
formulation (calcium gluconate or calcium chloride) to use sulin given as a 10-U bolus or as a 1-hour 20-U infusion
has not advanced since these early observations. The most without a loading dose lowers the serum potassium by about
common dose of calcium recommended today is 10 to 20 ml 1 mmol/l within an hour, Figure 1 illustrates why both of
of 10% calcium gluconate given intravenously as a bolus and these regimens are suboptimal.58 Neither regimen provides
repeated as needed. maximal kalemic insulin levels for very long, and both lead to
Because digoxin, an inhibitor of sodium-potassium persistently elevated insulin levels that can cause hypoglyce-
adenosine triphosphatase, increases intracellular calcium, mia. If glucose is given as a bolus, hyperglycemia occurs in the
there are theoretical concerns about calcium treatment for first few minutes, which may blunt the kalemic effect of in-
hyperkalemia caused by or associated with digitalis toxicity, sulin; hyperglycemia leads to water movement from the
and there have been case reports of adverse effects.38 A small intracellular to extracellular compartment, favoring potas-
case-controlled study found no mortality differences sium efflux from cells through solvent drag.59,60 Hypoglyce-
between 23 patients with hyperkalemia and digitalis toxicity mia often develops an hour or more after the start of therapy
who were treated with calcium and 136 patients who were for 2 reasons: (i) the amount of glucose is insufficient to
not.38 Nonetheless, the risk of hyperkalemia on the cardiac replace the glucose utilized in response to exogenous insulin;

Kidney International (2016) 89, 546–554 547


review RH Sterns et al.: Treatment of hyperkalemia

10-U insulin bolus 20-U insulin infusion Beta-2 agonists. The beta-2 agonist albuterol (also called
Maximum
salbutamol) administered by inhalation, nebulization, or
600 kalemic intravenously has been studied in stable hyperkalemic patients
effect with end-stage renal disease.47,68 The serum potassium falls by
Plasma insulin (μU/ml)

0.3 to 0.6 mmol/l within 30 minutes, regardless of mode of


400
administration, but some patients fail to respond. The doses
used when albuterol is given by inhalation (the only formu-
Maximum
200 glycemic lation available in the United States) are 4 to 8 times those
effect prescribed for the treatment of acute asthma, and although no
severe adverse events have been reported in studies of stable
0
0 20 40 60 80 100 120 patients, some studies excluded patients with heart disease. At
Minutes high doses, albuterol may stimulate both beta-1 receptors,
Figure 1 | Idealized plasma insulin levels after commonly used which can precipitate arrhythmias, and alpha-receptors, which
regimens in a patient with ESRD. After a 10-U bolus, insulin levels cause potassium release from the liver and can transiently in-
are transiently very high but quickly become suboptimal. After a crease serum potassium by >0.4 mmol/l.69 Subcutaneous
1-hour infusion of 20 U without a loading dose, insulin levels are
initially suboptimal. After both regimens, insulin levels persist at levels terbutaline, known to cause hypokalemia when used to treat
high enough to cause hypoglycemia unless glucose administration is premature labor, also lowers the serum potassium in mildly
continued for more than an hour. ESRD, end-stage renal disease. hyperkalemic patients with end-stage renal disease.
Several studies have documented a substantially greater fall
(ii) insulin’s prolonged half-life in end-stage renal disease in potassium when beta-2 agonists are combined with insulin
leads to insulin levels high enough to promote glucose utili- than when either agent is given alone and hypoglycemia is less
zation for more than an hour.40,61–63 Retrospective studies likely with combination therapy than with insulin
confirm that hypoglycemia commonly occurs within 1 to 3 alone.47,68,69 However, these studies employed submaximal
hours when hyperkalemia is treated with insulin and indicate doses of insulin and glucose, and it is unclear whether
that its incidence depends more on the dose of glucose than coadministration would offer any advantage over an adequate
on the dose of insulin; for example, in one study, 2 of 5 pa- dose of insulin given with sufficient glucose.8
tients given only 5 units of insulin with 25 g of glucose Sodium bicarbonate. Administration of bicarbonate pro-
developed a blood glucose <2.2 mmol/l.64 motes uptake of potassium by skeletal muscle by favoring
Short-acting insulins (lispro and aspart) have shorter half- sodium-bicarbonate cotransport and sodium-hydrogen ex-
lives than regular insulin and, in contrast to regular insulin, change, which, by increasing intracellular sodium, increases
their half-lives are not prolonged by kidney failure.58,61,65 A the activity of sodium-potassium adenosine triphosphatase.60
randomized, prospective study of infusion therapy to main- In 1959, Schwarz showed that infusion of between 144 and
tain normoglycemia in critical care patients showed the 408 mmol of sodium bicarbonate over 2 to 4 hours lowered
postinfusional drop in blood glucose to be less profound and the serum potassium by 2 to 3 mmol/l in 4 patients with
of briefer duration after lispro than after regular insulin.62 A severe acidosis.70 For many years, bicarbonate was often
retrospective study of outcomes after a 10-U insulin aspart chosen as the first-line treatment for acute hyperkalemia.71
bolus for hyperkalemia found rates of hypoglycemia compa- Bicarbonate ceased to be a recommended intervention for
rable to previously reported rates after regular insulin; how- acute hyperkalemia after the publication of studies showing
ever, only 25 g of glucose were given, an insufficient dose to that bicarbonate has little effect on the serum potassium
prevent hypoglycemia.66 concentration in stable hemodialysis patients.46,56,57,72,73
Based on what is known of physiology and drug However, bicarbonate therapy may be beneficial for patients
kinetics,40,58,61,67 the most logical regimen for a 70-kg subject with metabolic acidosis.74,75 A 4-hour infusion of sodium
(with weight adjustment of dosages for others) would be an bicarbonate in 5% dextrose in patients with chronic kidney
infusion of short-acting insulin at 20 U/h after a 6-U loading disease (CKD) resulted in a substantial fall in serum potas-
dose, given with 60 g of glucose per hour. Particularly if in- sium concentration that was proportional to the increase in
sulin is continued for more than an hour, administration of serum bicarbonate, with a mean decrease of approximately 2
an adequate amount of glucose intravenously is difficult. mmol/l after a 10-mmol/l increase in serum bicarbonate
Infusion of 10% glucose would require 600 ml/h, causing concentration.76 Bicarbonate is also rational therapy to
hyponatremia. High concentrations of glucose to reduce the enhance potassium excretion in patients with intact kidney
volume infused would require use of a central vein. It should function.
be possible to give glucose orally to avoid the need for central Combination “cocktails.”. In 1954, Meroney and Hern-
venous access. Because short-acting insulin is more rapidly don77 reported on a standard battlefield intravenous solution
absorbed than regular insulin, subcutaneous administration containing 400 ml of 25% dextrose, 50 U of regular insulin,
would be expected to be effective, but the dose needed to 50 mmol of sodium bicarbonate, which, along with boluses of
achieve maximally kalemic insulin levels is not known. intravenous calcium as needed, was effective in controlling
Clinical trials to assess these theoretical regimens are needed. hyperkalemia for several days in soldiers with traumatic acute

548 Kidney International (2016) 89, 546–554


RH Sterns et al.: Treatment of hyperkalemia review

kidney injury. More recently, Janjua et al.78 reported that a a End dialysis 2-h postdialysis
similar standard “cocktail” (containing sodium lactate instead 4
of sodium bicarbonate to permit the inclusion of calcium in 3.5

ΔPlasma potassium (mmol/l)


the solution) controlled hyperkalemia for up to 32 hours in 3
children and adolescents.
2.5

Elimination of potassium 2

Strategies to shift potassium into cells are temporizing ma- 1.5


neuvers that should be followed by efforts to eliminate excess 1
potassium. Reducing total body potassium involves decreased
0.5
potassium intake, enhanced urinary and fecal potassium
excretion, and dialysis. It is important for ambulatory patients 0
3 3.5 4 4.5 5 5.5 6 6.5 7
with hyperkalemia to see a dietician to guide food choices. Predialysis plasma potassium (mmol/l)
Patients should be instructed to limit intake of citrus fruit,
potatoes, tomato products, and salt substitutes, which are b 3.5
made of potassium salts.79
3
Patients with hyperkalemia are commonly taking po-
ΔPlasma K per 100 mmol
tassium supplements or medications that limit potassium 2.5
excretion; discontinuation or temporary dose reduction K+ removed
may be all that is required to restore normokalemia.80 2

Drug-induced hyperkalemia occurs most often in patients 1.5


with impaired kidney function and associated hypo-
reninemic hypoaldosteronism, and it is particularly com- 1

mon in the elderly.81–86 Common medications that cause 0.5


hyperkalemia include potassium supplements, angiotensin-
converting enzyme inhibitors, angiotensin-II blockers, 0
3 4 5 6 7
trimethoprim, calcineurin inhibitors, heparin, potassium- Predialysis plasma potassium (mmol/l)
sparing diuretics, digoxin, beta-blockers, and nonsteroidal Figure 2 | Response of plasma potassium to potassium removal
anti-inflammatory agents.79 Combinations of these agents by dialysis. (a) Change in plasma potassium levels at the end of a
are most likely to increase the risk of hyperkalemia and 3-hour dialysis against a zero potassium dialysate (blue symbols) and
sudden death.82,85,87–89 2 hours after dialysis (red symbols). There is a significantly greater fall
in plasma potassium when the predialysis potassium concentration is
Patients with severe hyperkalemia require active mea- higher. (b) The change in plasma potassium per 100 mmol of po-
sures to remove potassium. Studies of potassium removal by tassium removed (mmol/kg data normalized to 70-kg subject) at the
hemodialysis show that for a given negative potassium end of dialysis (blue symbols) and 2 hours after dialysis (red symbols).
balance, patients with higher serum potassium concentra- For a given amount of potassium removal, there is a significantly
greater fall in plasma potassium when the predialysis potassium
tions experience a larger fall in serum potassium (Figure 2a concentration is higher. Based on data in Feig et al.,90 used with
and b).74,75,90 This phenomenon makes it difficult to permission.
interpret studies purporting to show a “dose-response”
relationship to potassium binders when patients with higher
serum potassium concentrations are given higher doses of perfusion of peripheral tissues is minimal.95–97 In more
the agent.91 stable patients, over 100 mmol of potassium can be removed
Dialysis. Hemodialysis is the most effective way to during a 4-hour dialysis session; the amount removed de-
eliminate excess potassium. Although potassium is directly pends on the plasma-to-dialysate concentration gradient,
removed from plasma, distribution of potassium between the blood and dialysate flow rates, and total body potassium
plasma and interstitial fluid is nearly instantaneous, so po- stores (a function of muscle mass).90,92–94 Because replen-
tassium is effectively removed from extracellular fluid. The ishment from cellular stores continue when potassium
effect of dialysis on the plasma potassium concentration removal stops, there is a substantial postdialysis rebound of
depends on the rate of potassium removal from the extra- plasma potassium (Figure 3).90,93
cellular fluid and the rate that potassium is replenished from Urinary excretion. In patients with only moderately
intracellular stores.90,92–94 In a 70-kg subject, unreplenished compromised kidney function, loop diuretics, flu-
removal of only 14 mmol of potassium from the extracellular drocortisone, and sodium bicarbonate can substantially in-
fluid will decrease plasma potassium by 1 mmol/l. At a blood crease urinary potassium losses and have been used to
pump speed of 0.3 l/min and a plasma-to-dialysate concen- manage extremely severe hyperkalemia without dial-
tration gradient >5 mmol/l, such a decrease can be achieved ysis.60,81,98–100 Although sodium retention and the potential
within minutes. This explains why hemodialysis can be for adverse mineralocorticoid effects on the myocardium
successful during cardiopulmonary resuscitation, when make chronic fludrocortisone unattractive for chronic

Kidney International (2016) 89, 546–554 549


review RH Sterns et al.: Treatment of hyperkalemia

et al.105 found that 30 g/day of Kayexalate without sorbitol for


Dialysis Postdialysis 7 days was superior to placebo in the reduction of serum
7 potassium (mean difference between groups 1.07 mmol/l;
95% confidence interval: -1.37 to -0.71). There were trends
Plasma potassium (mmol/l)

for patients treated with Kayexalate to have more hypomag-


nesemia (P ¼ 0.08), hypocalcemia (P ¼ 0.10), nausea, and
6
constipation than those in the control group. Neither urinary
nor stool potassium balance was measured.
Chernin et al.106 retrospectively analyzed the course of all
patients taking target doses of renin-angiotensin-aldosterone
5
system inhibitors for heart disease who were maintained on
long-term therapy with a low dose of sorbitol-free Kayexalate
(15 g/day in water) after a serum potassium $6.0 mmol/l
4 had been identified. During 289 months of Kayexalate
therapy with continued use of renin-angiotensin-aldosterone
0 1 2 3 1 2 3 4 system inhibitors, the mean serum potassium fell from
Hours
6.4 " 0.3 mmol/l (range 6.0–7.1) to 4.6 " 0.6 (range
Figure 3 | Plasma potassium concentration during and after 3.0–5.8 mmol/l), P < 0.01. The study had no control group,
dialysis. Modified, with permission, from Figure 3 in Blumberg A,
Roser HW, Zehnder C, Muller-Brand J. Plasma potassium in patients
and there was no routine monitoring of serum magnesium.
with terminal renal failure during and after haemodialysis; Other than this study, data on long-term use of Kayexalate or
relationship with dialytic potassium removal and total body SPS in sorbitol are lacking.107
potassium. Nephrol Dial Transplant. 1997;12:1629–1634.93 Serious gastrointestinal complications from SPS have been
reported, given with and without sorbitol, including fatal
colonic perforation (reviewed in Harel et al.108). Because a
therapy, it can be useful acutely as it begins to work within small study in rats suggested that sorbitol was the cause of
3 hours.98,101 Large doses of fludrocortisone (up to 0.4 mg colonic perforation,109 the U.S. Food and Drug Administra-
daily) may be required, as patients with kidney disease may tion issued a warning to avoid administration of Kayexalate
have aldosterone resistance as well as aldosterone deficiency.98 with sorbitol.4 This advice was problematic, because most
Unfortunately, although many clinicians use such measures hospital pharmacies in the United States stock a premixed
routinely, they have not been well studied. preparation of SPS suspended in 33% sorbitol rather than
Sodium polystyrene sulfonate (Kayexalate). Kayexalate is a powdered Kayexalate. A retrospective cohort study of 123,391
cation exchange resin, first introduced in the 1950s, before the adult inpatients identified 2194 patients who had been treated
U.S. Food and Drug Administration was required to establish with SPS in sorbitol, of whom, 0.14% developed colonic
that drugs are safe and effective before approving them.4 necrosis, a rate that did not differ significantly from the rate
Kayexalate was the name given to the powdered form of in patients not receiving the drug (0.07%).110 Assuming sta-
SPS, which exchanges sodium for calcium, ammonium, and tistical significance, the number needed to harm was esti-
magnesium in addition to potassium.102 At an acid pH, its mated at 1395. A much larger population would be required
sulfonate groups are occupied by hydrogen ions and are un- for a rigorous multivariate analysis of the risk of SPS-
able to bind potassium. For this reason, and because of higher associated colonic necrosis after acute administration.
potassium concentrations in the distal colon, Kayexalate is Patiromer. Patiromer is an investigational nonabsorbable
most effective in binding potassium when it reaches the synthetic polymer consisting of smooth spherical beads
rectum, either by retention enema or by oral administration approximately 100 mm in diameter; unlike Kayexalate,
with cathartics.103 Because the resin swells when it contacts patiromer does not swell appreciably when exposed to water
water, large doses of Kayexalate can cause bowel obstruction. and it does not require a laxative to reach the distal colon.
To avoid this complication and to speed its delivery to the Patiromer’s active groups are comprised of alpha-fluo-
distal colon, Kayexalate has been given together with sorbitol, rocarboxylic acids that are paired with calcium ions rather
an osmotic cathartic.1 A placebo-controlled trial failed to than sodium. As the resin travels through the gastrointestinal
show any difference between the amount of fecal potassium tract, some of the calcium is replaced with hydrogen ions. The
within 12 hours of oral ingestion when sorbitol is given with pKa of Patiromer’s cation binding groups is such that at the
or without Kayexalate to patients with end-stage renal disease, prevailing pH in the colon, the acid groups are dissociated,
and there was no effect on the serum potassium concentra- allowing them to bind potassium, which is present in high
tion.104 No controlled trials in humans or animals have concentrations in this bowel segment, as well as ammonium
demonstrated that Kayexalate increases fecal potassium losses. and magnesium.86 Controlled experiments in both experi-
However, in a double-blind, randomized, placebo- mental animals and normal volunteers have demonstrated
controlled trial, conducted in 33 ambulatory patients with that when taken orally, the polymer increases fecal potassium
CKD and mild hyperkalemia (5.0–5.9 mmol/l), LePage in a dose-related fashion; doses of 15 to 30 g/day increased

550 Kidney International (2016) 89, 546–554


RH Sterns et al.: Treatment of hyperkalemia review

daily fecal potassium by approximately 15 to 20 mmol.111 In has been studied in an open-label 28-day trial,119 and it is
subjects studied while on a potassium-restricted and sodium- currently being studied in a larger 52-week trial. In addition
restricted diet, patiromer decreased serum potassium by 0.23 to minor gastrointestinal side effects, edema developed in
mmol/l within 7 hours.112 The drug’s ability to achieve nor- 6% of patients taking 10 g/day and 14% of patients taking
mokalemia has been proven in randomized multicenter 15 g/day (as compared to 2% of control subjects).119 ZS-9
placebo-controlled trials involving a total of 603 hyperkalemic exchanges sodium and hydrogen ions for potassium and
patients on active treatment;113–115 its safety was demon- maintenance doses of the drug (5, 10, or 15 g/day) provide
strated in an open-label 52-week trial.115 In addition to minor approximately 17, 34, and 50 mmol of sodium per day.
and infrequent gastrointestinal side effects, the most impor- Because ZS-9 does not contain acid groups that dissociate,
tant recognized adverse drug effect is hypomagnesemia (0.58 it binds potassium throughout the gastrointestinal tract.116
mmol/l), which developed in the first month of therapy in This suggests that it will be effective in the management
8.6% of patients; hypomagnesemia responded readily to of acute hyperkalemia. In a subgroup of 45 patients with
magnesium supplementation and did not progress. Because serum potassium concentrations of at least 6 mmol/l (6.1 to
patiromer exchanges calcium for potassium it has the po- 7.2 mmol/l), who participated in 2 controlled trials, admin-
tential of causing positive calcium balance and ectopic calci- istration of 10 g of ZS-9 significantly reduced the serum
fications; this theoretical concern would be difficult to potassium concentration below baseline by 0.4 mmol/l at
disprove without very long-term studies. Patiromer was 1 hour, by 0.6 mmol/l at 2 hours, and by 0.7 mmol/l at
approved by the United States Food and Drug Administration 4 hours (P < 0.001).120
in October 2015 and should be available in early 2016. Based
on in vitro data showing that Patiromer binds to many orally Who to treat
administered medications, which could decrease their ab- Our knowledge of how to decrease the serum potassium
sorption and reduce their effectiveness, a black-box warning concentration, while imperfect, has improved substantially
was issued indicating that the drug should be separated by 6 in the past 2 years. Our understanding of when to treat
hours from other orally administered medications. hyperkalemia has lagged behind. Opinions vary widely as
Sodium zirconium cyclosilicate (ZS-9). Unlike patiromer to what level of serum potassium should be defined as
and SPS, the investigational drug, ZS-9 (ZS Pharma, Coppell, “severe” or what level constitutes a hyperkalemic emergency
TX) is not a polymer. It is a crystal that is highly selective for (Table 1).5,44,45,121 Hospital admission is often recommended
potassium and ammonium ions through mechanisms that are for patients with a serum potassium >6 mmol/l and elec-
very similar to those of naturally occurring ion channels.116 trocardiographic monitoring and acute interventions for any
Potassium and ammonium ions, which are nearly identical patient with a serum potassium >6.5 mmol/l.73 Although that
in size, must first shed their hydration shells before they enter is our practice, we recognize that it has not been established
the crystal structure, a process that requires energy; unhy- that this is necessary; 1 small study showed favorable out-
drated ions are of the right size to form thermodynamically comes among patients with serum potassium concentrations
stable and energetically favorable hydrogen bonds to sur- >6 mmol/l (6.7 " 0.5 mmol/l) who were managed as out-
rounding oxygen atoms in the crystal structure. After shed- patients,122 and no deaths were recorded in another study of
ding their hydration shells, sodium, calcium, and magnesium 242 consecutively admitted patients with a serum potassium
ions are too small to form such stable bonds, making it >6 mmol/l despite substantial delays in treatment.80
thermodynamically unfavorable for them to be bound by the Recent studies have reported increased risk of mortality
crystal. Controlled trials have proven that ZS-9 increases fecal among patients with hyperkalemia, findings that are likely to
potassium losses in rats, in a dose-dependent manner.117 ZS-9 be emphasized when the new potassium binders reach the
has been shown to be effective in maintaining normokalemia market.28,123–126 For example, a 1-year retrospective analysis
in randomized placebo controlled trials involving 1101 of a national cohort of 245,808 veterans with at least 1 hos-
hyperkalemic patients taking the active drug.117–119 Its safety pital admission and 1 inpatient or outpatient potassium value

Table 1 | Definitions of the severity of hyperkalemia


Minimal or mild
Author Year hyperkalemia Moderate hyperkalemia Severe hyperkalemia
5
Levinsky 1966 <6.5 mmol/l 6.5–8 mmol/l with ECG >8 mmol/l or any level associated with prolongation
limited to peaked T-waves of the QRS complex, ventricular arrhythmias, or
heart block
Vanden Hoek et al., for the 2005 5.1–5.9 mmol/l 6.0–6.9 mmol/l >7 mmol/l
American Heart
Association45
Soar et al., for the European 2010 5.5–5.9 mmol/l 6.0–6.4 mmol/l $6.5 mmol/l
Resuscitation Council44
El-Sherif and Turitto121 2011 5.5–7.5 mmol/l 7.5–10 mmol/l >10 mmol/l
ECG, electrocardiogram.

Kidney International (2016) 89, 546–554 551


review RH Sterns et al.: Treatment of hyperkalemia

found that among patients with CKD the risk of dying within 7. Elliott MJ, Ronksley PE, Clase CM, et al. Management of patients with
acute hyperkalemia. CMAJ. 2010;182:1631–1635.
a day of documenting potassium between 5.5 and <6 mmol/l 8. Kamel KS, Wei C. Controversial issues in the treatment of
as an outpatient, values usually labeled as “mild or minimal hyperkalaemia. Nephrol Dial Transplant. 2003;18:2215–2218.
hyperkalemia” (Table 1), was 2.73 times higher than with 9. Mahoney BA, Smith WA, Lo DS, et al. Emergency interventions for
hyperkalaemia. Cochrane Database Syst Rev. 2005;(2):CD003235.
potassium <5.5 mmol/l; the risk was 13 times higher for 10. Ingelfinger JR. A new era for the treatment of hyperkalemia? N Engl J
potassium >6 mmol/l.123 These alarming statistics should be Med. 2015;372:275–277.
placed in perspective. The absolute mortality rates were 0.2% 11. Gumz ML, Rabinowitz L, Wingo CS. An integrated view of potassium
homeostasis. N Engl J Med. 2015;373:60–72.
for potassium between 5.5 and <6 mmol/l and 0.9% for
12. Yang A, Singh B, Lavin PT, et al. Placebo effect in clinical studies in
potassium >6 mmol/l (although much higher rates were hyperkalemia: a double-blind, randomized, placebo-controlled phase 3
recorded among hospitalized patients). Curiously, both rela- study of sodium zirconium cyclosilicate (ZS-9) [Abstract]. Am J Kidney
Dis. 2015;65:A91.
tive risks (6.17 and 27.4) and absolute risks (0.4% and 1.7%)
13. Gumz ML, Rabinowitz L. Role of circadian rhythms in potassium
of dying within 24 hours for the same potassium values were homeostasis. Semin Nephrol. 2013;33:229–236.
much higher among patients without CKD. One may specu- 14. Schmidt ST, Ditting T, Deutsch B, et al. Circadian rhythm and day to day
late with the authors that this finding reflects an adaptation to variability of serum potassium concentration: a pilot study. J Nephrol.
2015;28:165–172.
more chronic hyperkalemia, but the paradoxical finding that 15. Meng QH, Wagar EA. Pseudohyperkalemia: a new twist on an old
CKD (a powerful predictor of mortality) was protective may phenomenon. Crit Rev Clin Lab Sci. 2015;52:45–55.
also mean that deaths in patients without CKD were not 16. Bia MJ, DeFronzo RA. Extrarenal potassium homeostasis. Am J Physiol.
1981;240:F257–268.
caused by hyperkalemia per se, but rather from underlying 17. Graber M, Subramani K, Corish D, Schwab A. Thrombocytosis elevates
cardiovascular disease. serum potassium. Am J Kidney Dis. 1988;12:116–120.
Retrospectively determined associations between hyper- 18. Don BR, Sebastian A, Cheitlin M, et al. Pseudohyperkalemia caused by
fist clenching during phlebotomy. N Engl J Med. 1990;322:1290–1292.
kalemia and mortality or between resolution of hyper- 19. Seimiya M, Yoshida T, Sawabe Y, et al. Reducing the incidence of
kalemia and improved mortality do not prove cause and pseudohyperkalemia by avoiding making a fist during phlebotomy: a
effect, and they are a poor basis for therapeutic strategies quality improvement report. Am J Kidney Dis. 2010;56:686–692.
20. Bailey IR, Thurlow VR. Is suboptimal phlebotomy technique impacting on
that are likely to be expensive127 and potentially risky. potassium results for primary care? Ann Clin Biochem. 2008;45:266–269.
Currently, because of hyperkalemia, many patients cannot be 21. Thurlow V, Ozevlat H, Jones SA, Bailey IR. Establishing a practical blood
treated with inhibitors of the renin-angiotensin-aldosterone platelet threshold to avoid reporting spurious potassium results due to
thrombocytosis. Ann Clin Biochem. 2005;42:196–199.
system that have been shown to prolong life. This population 22. Chawla NR, Shapiro J, Sham RL. Pneumatic tube “pseudo tumor lysis
is likely to be the major beneficiary of new agents to treat syndrome” in chronic lymphocytic leukemia. Am J Hematol. 2009;84:
hyperkalemia. However, as pointed out in a recent edito- 613–614.
23. Kellerman PS, Thornbery JM. Pseudohyperkalemia due to pneumatic
rial,128 it is quite possible that development of hyperkalemia tube transport in a leukemic patient. Am J Kidney Dis. 2005;46:746–748.
under renin-angiotensin-aldosterone system blockade is a 24. Sindhu SK, Hix JK, Fricke W. Pseudohyperkalemia in chronic
marker for underlying pathology that may lead to mortality lymphocytic leukemia: phlebotomy sites and pneumatic tubes. Am J
Kidney Dis. 2011;57:354–355.
regardless of treatment; control of the plasma potassium 25. Parham WA, Mehdirad AA, Biermann KM, Fredman CS. Hyperkalemia
concentration is ultimately a surrogate marker for the hard revisited. Tex Heart Inst J. 2006;33:40–47.
outcomes that should be studied: mortality rates, reduced 26. Bashour T, Hsu I, Gorfinkel HJ, et al. Atrioventricular and intraventricular
conduction in hyperkalemia. Am J Cardiol. 1975;35:199–203.
progression of CKD, deferral of dialysis, improved heart 27. Ettinger PO, Regan TJ, Oldewurtel HA. Hyperkalemia, cardiac conduction,
failure outcomes. It is highly unlikely that we will see such and the electrocardiogram: a review. Am Heart J. 1974;88:360–371.
outcome trials in the foreseeable future. Without them, the 28. McCullough PA, Beaver TM, Bennett-Guerrero E, et al. Acute and chronic
cardiovascular effects of hyperkalemia: new insights into prevention and
next 50 years may be as frustrating as the last for those who clinical management. Rev Cardiovasc Med. 2014;15:11–23.
must treat hyperkalemia. 29. Mattu A, Brady WJ, Robinson DA. Electrocardiographic manifestations
of hyperkalemia. Am J Emerg Med. 2000;18:721–729.
DISCLOSURE 30. Wrenn KD, Slovis CM, Slovis BS. The ability of physicians to predict
All the authors declared no competing interests. hyperkalemia from the ECG. Ann Emerg Med. 1991;20:1229–1232.
31. Aslam S, Friedman EA, Ifudu O. Electrocardiography is unreliable in
detecting potentially lethal hyperkalaemia in haemodialysis patients.
REFERENCES Nephrol Dial Transplant. 2002;17:1639–1642.
1. Flinn RB, Merrill JP, Welzant WR. Treatment of the oliguric patient with a 32. Khattak HK, Khalid S, Manzoor K, Stein PK. Recurrent life-threatening
new sodium-exchange resin and sorbitol: a preliminary report. N Engl J hyperkalemia without typical electrocardiographic changes.
Med. 1961;264:111–115. J Electrocardiol. 2014;47:95–97.
2. Anonymous. Treatment of potassium retention. N Engl J Med. 1961;264: 33. Martinez-Vea A, Bardaji A, Garcia C, Oliver JA. Severe hyperkalemia with
149–150. minimal electrocardiographic manifestations: a report of seven cases.
3. Scherr L, Ogden DA, Mead AW, et al. Management of hyperkalemia J Electrocardiol. 1999;32:45–49.
with a cation-exchange resin. N Engl J Med. 1961;264:115–119. 34. Szerlip HM, Weiss J, Singer I. Profound hyperkalemia without
4. Sterns RH, Rojas M, Bernstein P, Chennupati S. Ion-exchange resins for electrocardiographic manifestations. Am J Kidney Dis. 1986;7:461–465.
the treatment of hyperkalemia: are they safe and effective? J Am Soc 35. Chon SB, Kwak YH, Hwang SS, et al. Severe hyperkalemia can be
Nephrol. 2010;21:733–735. detected immediately by quantitative electrocardiography and
5. Levinsky NG. Management of emergencies. VI. Hyperkalemia. N Engl J clinical history in patients with symptomatic or extreme bradycardia:
Med. 1966;274:1076–1077. a retrospective cross-sectional study. J Crit Care. 2013;28:
6. Merrill JP, Levine HD, Somerville W, Smith S. Clinical recognition and 1112.e7–1112.e13.
treatment of acute potassium intoxication. Ann Intern Med. 1950;33: 36. Barold SS, Herweg B. The effect of hyperkalaemia on cardiac rhythm
797–830. devices. Europace. 2014;16:467–476.

552 Kidney International (2016) 89, 546–554


RH Sterns et al.: Treatment of hyperkalemia review

37. Chamberlain M. Emergency treatment of hyperkalaemia. Lancet. 1964;1: 62. Bilotta F, Badenes R, Lolli S, et al. Insulin infusion therapy in critical care
464–467. patients: regular insulin vs short-acting insulin. A prospective,
38. Levine M, Nikkanen H, Pallin DJ. The effects of intravenous calcium in crossover, randomized, multicenter blind study. J Crit Care. 2015;30:
patients with digoxin toxicity. J Emerg Med. 2011;40:41–46. 437.e1–437.e6.
39. Ho K. A critically swift response: insulin-stimulated potassium and 63. DeFronzo RA, Alvestrand A, Smith D, et al. Insulin resistance in uremia.
glucose transport in skeletal muscle. Clin J Am Soc Nephrol. 2011;6: J Clin Invest. 1981;67:563–568.
1513–1516. 64. Schafers S, Naunheim R, Vijayan A, Tobin G. Incidence of hypoglycemia
40. DeFronzo RA, Felig P, Ferrannini E, Wahren J. Effect of graded doses of following insulin-based acute stabilization of hyperkalemia treatment.
insulin on splanchnic and peripheral potassium metabolism in man. Am J Hosp Med. 2012;7:239–242.
J Physiol. 1980;238:E421–427. 65. Czock D, Aisenpreis U, Rasche FM, Jehle PM. Pharmacokinetics and
41. DeFronzo RA, Tobin JD, Andres R. Glucose clamp technique: a method pharmacodynamics of lispro-insulin in hemodialysis patients with
for quantifying insulin secretion and resistance. Am J Physiol. 1979;237: diabetes mellitus. Int J Clin Pharmacol Ther. 2003;41:492–497.
G214–G223. 66. Estep P, Efird L. Evaluation of hypoglycemia incidence and risk factors
42. Alvestrand A, Wahren J, Smith D, DeFronzo RA. Insulin-mediated in patients treated with IV insulin aspart for hyperkalemia. Endocrinol
potassium uptake is normal in uremic and healthy subjects. Am J Diabetes Res. 2015;3:1–4.
Physiol. 1984;246:E174–E180. 67. Silvers A, Swenson RS, Farquhar JW, Reaven GM. Derivation of a three
43. Nguyen TQ, Maalouf NM, Sakhaee K, Moe OW. Comparison of insulin compartment model describing disappearance of plasma insulin-131-I
action on glucose versus potassium uptake in humans. Clin J Am Soc in man. J Clin Invest. 1969;48:1461–1469.
Nephrol. 2011;6:1533–1539. 68. Allon M, Dunlay R, Copkney C. Nebulized albuterol for acute
44. Soar J, Perkins GD, Abbas G, et al. European Resuscitation Council hyperkalemia in patients on hemodialysis. Ann Intern Med. 1989;110:
Guidelines for Resuscitation 2010 Section 8. Cardiac arrest in special 426–429.
circumstances: electrolyte abnormalities, poisoning, drowning, 69. Mandelberg A, Krupnik Z, Houri S, et al. Salbutamol metered-dose
accidental hypothermia, hyperthermia, asthma, anaphylaxis, cardiac inhaler with spacer for hyperkalemia: how fast? How safe? Chest.
surgery, trauma, pregnancy, electrocution. Resuscitation. 2010;81: 1999;115:617–622.
1400–1433. 70. Schwarz KC, Cohen BD, Lubash GD, Rubin AL. Severe acidosis and
45. Vanden Hoek TL, Morrison LJ, Shuster M, et al. Part 12: cardiac arrest in hyperpotassemia treated with sodium bicarbonate infusion. Circulation.
special situations: 2010 American Heart Association Guidelines for 1959;19:215–220.
Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. 71. Iqbal Z, Friedman EA. Preferred therapy of hyperkalemia in renal
Circulation. 2010;122(suppl 3):S829–S861. insufficiency: survey of nephrology training-program directors. N Engl J
46. Allon M, Shanklin N. Effect of bicarbonate administration on plasma Med. 1989;320:60–61.
potassium in dialysis patients: interactions with insulin and albuterol. 72. Gutierrez R, Schlessinger F, Oster JR, et al. Effect of hypertonic versus
Am J Kidney Dis. 1996;28:508–514. isotonic sodium bicarbonate on plasma potassium concentration in
47. Allon M, Copkney C. Albuterol and insulin for treatment of patients with end-stage renal disease. Miner Electrolyte Metab. 1991;17:
hyperkalemia in hemodialysis patients. Kidney Int. 1990;38:869–872. 297–302.
48. Chothia MY, Halperin ML, Rensburg MA, et al. Bolus administration of 73. Greenberg A. Hyperkalemia: treatment options. Semin Nephrol. 1998;18:
intravenous glucose in the treatment of hyperkalemia: a randomized 46–57.
controlled trial. Nephron Physiol. 2014;126:1–8. 74. Sterns RH, Spital A. Disorders of internal potassium balance. Semin
49. Lens XM, Montoliu J, Cases A, et al. Treatment of hyperkalaemia in renal Nephrol. 1987;7:399–415.
failure: salbutamol v. insulin. Nephrol Dial Transplant. 1989;4:228–232. 75. Sterns RH, Cox M, Feig PU, Singer I. Internal potassium balance and
50. Ljutic D, Rumboldt Z. Should glucose be administered before, with, the control of the plasma potassium concentration. Medicine. 1981;60:
or after insulin, in the management of hyperkalemia? Ren Fail. 1993;15: 339–354.
73–76. 76. Fraley DS, Adler S. Correction of hyperkalemia by bicarbonate despite
51. Mushtaq MA, Masood M. Treatment of hyperkalemia with salbutamol constant blood pH. Kidney Int. 1977;12:354–360.
and insulin. Pakistan J Med Sci. 2006;22:176. 77. Meroney W, Herndon R. The management of acute renal insufficiency.
52. Ngugi NN, McLigeyo SO, Kayima JK. Treatment of hyperkalaemia by J Am Med Assoc. 1954;155:877–883.
altering the transcellular gradient in patients with renal failure: effect of 78. Janjua HS, Mahan JD, Patel HP, et al. Continuous infusion of a standard
various therapeutic approaches. East Afr Med J. 1997;74:503–509. combination solution in the management of hyperkalemia. Nephrol Dial
53. Duranay M, Ates K, Erturk S, et al. Comparison of aminophylline and Transplant. 2011;26:2503–2508.
insulin infusions in treatment of hyperkalemia in patients with end- 79. Perazella MA. Drug-induced hyperkalemia: old culprits and new
stage renal disease. Nephron. 1996;73:105. offenders. Am J Med. 2000;109:307–314.
54. Mahajan SK, Mangla M, Kishore K. Comparison of aminophylline 80. Acker CG, Johnson JP, Palevsky PM, Greenberg A. Hyperkalemia in
and insulin-dextrose infusions in acute therapy of hyperkalemia in hospitalized patients: causes, adequacy of treatment, and results of an
end-stage renal disease patients. J Assoc Physicians India. 2001;49: attempt to improve physician compliance with published therapy
1082–1085. guidelines. Arch Intern Med. 1998;158:917–924.
55. Kim HJ. Combined effect of bicarbonate and insulin with glucose in 81. Palmer BF. Managing hyperkalemia caused by inhibitors of the renin–
acute therapy of hyperkalemia in end-stage renal disease patients. angiotensin–aldosterone system. N Engl J Med. 2004;351:585–592.
Nephron. 1996;72:476–482. 82. Antoniou T, Gomes T, Juurlink DN, et al. Trimethoprim-
56. Blumberg A, Weidmann P, Ferrari P. Effect of prolonged bicarbonate sulfamethoxazole-induced hyperkalemia in patients receiving inhibitors
administration on plasma potassium in terminal renal failure. Kidney Int. of the renin-angiotensin system: a population-based study. Arch Intern
1992;41:369–374. Med. 2010;170:1045–1049.
57. Blumberg A, Weidmann P, Shaw S, Gnädinger M. Effect of various 83. Gentry CA, Nguyen AT. An evaluation of hyperkalemia and serum
therapeutic approaches on plasma potassium and major regulating creatinine elevation associated with different dosage levels of
factors in terminal renal failure. Am J Med. 1988;85:507–512. outpatient trimethoprim-sulfamethoxazole with and without
58. Galloway JA, Spradlin CT, Nelson RL, et al. Factors influencing the concomitant medications. Ann Pharmacother. 2013;47:1618–1626.
absorption, serum insulin concentration, and blood glucose responses 84. Lam N, Weir MA, Juurlink DN, et al. Hospital admissions for
after injections of regular insulin and various insulin mixtures. Diabetes hyperkalemia with trimethoprim-sulfamethoxazole: a cohort study
Care. 1981;4:366–376. using health care database codes for 393,039 older women with urinary
59. Goldfarb S, Cox M, Singer I, Goldberg M. Acute hyperkalemia induced tract infections. Am J Kidney Dis. 2011;57:521–523.
by hyperglycemia: hormonal mechanisms. Ann Intern Med. 1976;84: 85. Schepkens H, Vanholder R, Billiouw J, Lameire N. Life-threatening
426–432. hyperkalemia during combined therapy with angiotensin-converting
60. Palmer BF. Regulation of potassium homeostasis. Clin J Am Soc Nephrol. enzyme inhibitors and spironolactone: an analysis of 25 cases. Am J
2015;10:1050–1060. Med. 2001;110:438–441.
61. Iglesias P, Díez JJ. Insulin therapy in renal disease. Diabetes Obes Metab. 86. Weir MA, Juurlink DN, Gomes T, et al. Beta-blockers, trimethoprim-
2008;10:811–823. sulfamethoxazole, and the risk of hyperkalemia requiring

Kidney International (2016) 89, 546–554 553


review RH Sterns et al.: Treatment of hyperkalemia

hospitalization in the elderly: a nested case-control study. Clin J Am Soc chronic hyperkalemia. Clinical Nephrology, 2014;81:259–268. Clin
Nephrol. 2010;5:1544–1551. Nephrol. 2015;83:380–381.
87. Tamirisa KP, Aaronson KD, Koelling TM. Spironolactone-induced renal 108. Harel Z, Harel S, Shah PS, et al. Gastrointestinal adverse events with
insufficiency and hyperkalemia in patients with heart failure. Am Heart J. sodium polystyrene sulfonate (Kayexalate) use: a systematic review. Am
2004;148:971–978. J Med. 2013;126:264.e9–264.e24.
88. Susantitaphong P, Sewaralthahab K, Balk EM, et al. Efficacy and safety 109. Lillemoe KD, Romolo JL, Hamilton SR, et al. Intestinal necrosis due
of combined vs. single renin-angiotensin-aldosterone system blockade to sodium polystyrene (Kayexalate) in sorbitol enemas: clinical
in chronic kidney disease: a meta-analysis. Am J Hypertens. 2013;26: and experimental support for the hypothesis. Surgery. 1987;101:267–272.
424–441. 110. Watson MA, Baker TP, Nguyen A, et al. Association of prescription of
89. Fralick M, Macdonald EM, Gomes T, et al, for the Canadian Drug Safety oral sodium polystyrene sulfonate with sorbitol in an inpatient setting
and Effectiveness Research Network. Co-trimoxazole and sudden death with colonic necrosis: a retrospective cohort study. Am J Kidney Dis.
in patients receiving inhibitors of renin-angiotensin system: population 2012;60:409–416.
based study. BMJ. 2014;349:g6196. 111. Huang I. RLY5016: a novel, non-absorbed, therapeutic polymer for
90. Feig P, Shook A, Sterns R. Effect of potassium removal during hemodialysis serum potassium control. J Am Soc Nephrol. 2010;21:482A–483A.
on the plasma potassium concentration. Nephron. 1981;27:25–30. 112. Bushinsky DA. Patiromer induces a rapid and sustain potassium lowering
91. Kessler C, Ng J, Valdez K, et al. The use of sodium polystyrene sulfonate in in CKD patients with hyperkalemia. Kidney Int. 2015;88:1427–1433.
the inpatient management of hyperkalemia. J Hosp Med. 2011;6:136–140. 113. Pitt B, Anker SD, Bushinsky DA, et al. PEARL-HF Investigators. Evaluation
92. Agar BU, Culleton BF, Fluck R, Leypoldt JK. Potassium kinetics during of the efficacy and safety of RLY5016, a polymeric potassium binder, in
hemodialysis. Hemodial Int. 2015;19:23–32. a double-blind, placebo-controlled study in patients with chronic heart
93. Blumberg A, Roser HW, Zehnder C, Muller-Brand J. Plasma potassium in failure (the PEARL-HF) trial. Eur Heart J. 2011;32:820–828.
patients with terminal renal failure during and after haemodialysis; 114. Weir MR, Bakris GL, Bushinsky DA, et al, for the OPAL-HK Investigators.
relationship with dialytic potassium removal and total body potassium. Patiromer in patients with kidney disease and hyperkalemia receiving
Nephrol Dial Transplant. 1997;12:1629–1634. RAAS inhibitors. N Engl J Med. 2015;372:211–221.
94. Bolasco P, Concas G, Steckiph D, et al. Simple model of intra- 115. Bakris GL, Pitt B, Weir MR, et al, for the AMETHYST-DN Investigators.
extracellular potassium kinetics and removal applied to constant and Effect of patiromer on serum potassium level in patients with
potassium-profiled dialysis. J Nephrol. 2008;21:384–393. hyperkalemia and diabetic kidney disease: the AMETHYST-DN
95. Kao KC, Huang CC, Tsai YH, et al. Hyperkalemic cardiac arrest randomized clinical trial. JAMA. 2015;314:151–161.
successfully reversed by hemodialysis during cardiopulmonary 116. Stavros F, Yang A, Leon A, et al. Characterization of structure and
resuscitation: case report. Chang Gung Med J. 2000;23:555–559. function of ZS-9, a Kþ selective ion trap. PLoS One. 2014;9:e114686.
96. Lin JL, Lim PS, Leu ML, Huang CC. Outcomes of severe hyperkalemia in 117. Ash SR, Singh B, Lavin PT, et al. A phase 2 study on the treatment of
cardiopulmonary resuscitation with concomitant hemodialysis. hyperkalemia in patients with chronic kidney disease suggests that the
Intensive Care Med. 1994;20:287–290. selective potassium trap, ZS-9, is safe and efficient. Kidney Int. 2015;88:
97. Lin JL, Huang CC. Successful initiation of hemodialysis during 404–411.
cardiopulmonary resuscitation due to lethal hyperkalemia. Crit Care 118. Packham DK, Rasmussen HS, Lavin PT, et al. Sodium zirconium
Med. 1990;18:342–343. cyclosilicate in hyperkalemia. N Engl J Med. 2015;372:222–231.
98. DeFronzo RA. Hyperkalemia and hyporeninemic hypoaldosteronism. 119. Kosiborod M, Rasmussen HS, Lavin P, et al. Effect of sodium zirconium
Kidney Int. 1980;17:118–134. cyclosilicate on potassium lowering for 28 days among outpatients
99. Carvalhana V, Burry L, Lapinsky SE. Management of severe with hyperkalemia: the HARMONIZE randomized clinical trial. JAMA.
hyperkalemia without hemodialysis: case report and literature review. 2014;312:2223–2233.
J Crit Care. 2006;21:316–321. 120. Kosiborod M, Peacock WF, Packham DK. Sodium zirconium cyclosilicate
100. Dick T, Raines A, Stinson J, et al. Fludrocortisone is effective in the for urgent therapy of severe hyperkalemia. N Engl J Med. 2015;372:
management of tacrolimus-induced hyperkalemia in liver transplant 1577–1578.
recipients. Transplant Proc. 2011;43:2664–2668. 121. El-Sherif N, Turitto G. Electrolyte disorders and arrhythmogenesis.
101. Kamel KS, Ethier JH, Quaggin S, et al. Studies to determine the basis for Cardiol J. 2011;18:233–245.
hyperkalemia in recipients of a renal transplant who are treated with 122. Charytan D, Goldfarb DS. Indications for hospitalization of patients with
cyclosporine. J Am Soc Nephrol. 1992;2:1279–1284. hyperkalemia. Arch Intern Med. 2000;160:1605–1611.
102. Evans B, Milne M, Jones NH, Yellowlees H. Ion-exchange resins in the 123. Einhorn LM, Zhan M, Hsu VD, et al. The frequency of hyperkalemia and
treatment of anuria. Lancet. 1953;265:791–795. its significance in chronic kidney disease. Arch Intern Med. 2009;169:
103. Agarwal R, Afzalpurkar R, Fordtran JS. Pathophysiology of potassium 1156–1162.
absorption and secretion by the human intestine. Gastroenterology. 124. Khanagavi J, Gupta T, Aronow WS, et al. Hyperkalemia among
1994;107:548–571. hospitalized patients and association between duration of
104. Gruy-Kapral C, Emmett M, Santa Ana CA, et al. Effect of single dose hyperkalemia and outcomes. Arch Med Sci. 2014;10:251–257.
resin-cathartic therapy on serum potassium concentration in patients 125. McMahon GM, Mendu ML, Gibbons FK, Christopher KB. Association
with end-stage renal disease. J Am Soc Nephrol. 1998;9:1924–1930. between hyperkalemia at critical care initiation and mortality. Intensive
105. LePage L. Sodium polystyrene sulfonate for the treatment of mild Care Med. 2012;38:1834–1842.
hyperkalemia in chronic kidney disease: a randomized clinical trial. Clin 126. Conway R, Creagh D, Byrne DG, et al. Serum potassium levels as an
J Am Soc Nephrol. 2015;10:2136–2142. outcome determinant in acute medical admissions. Clin Med (Lond).
106. Chernin G, Gal-Oz A, Ben-Assa E, et al. Secondary prevention of 2015;15:239–243.
hyperkalemia with sodium polystyrene sulfonate in cardiac and kidney 127. Little DJ, Nee R, Abbott KC, et al. Cost-utility analysis of sodium
patients on renin-angiotensin-aldosterone system inhibition therapy. polystyrene sulfonate vs. potential alternatives for chronic
Clin Cardiol. 2012;35:32–36. hyperkalemia. Clin Nephrol. 2014;81:259–268.
107. Straube B, Reaven N, Funk S, Little D, Nee R, Abbott K, et al. Cost utility 128. Winkelmayer WC. Treatment of hyperkalemia: from “Hyper Kþ”
analysis of sodium polystyrene sulfate vs. potential alternatives for strikeout to home run? JAMA. 2015;314:129–130.

554 Kidney International (2016) 89, 546–554


ARTIGO 6

HIPERCALCEMIAS
BMJ 2015;350:h2723 doi: 10.1136/bmj.h2723 (Published 2 June 2015) Page 1 of 9

Clinical Review

CLINICAL REVIEW

The diagnosis and management of hypercalcaemia


Salvatore Minisola professor, Jessica Pepe consultant, Sara Piemonte consultant, Cristiana Cipriani
consultant
Department of Internal Medicine and Medical Disciplines, “Sapienza” Rome University, 00161 Rome, Italy

Hypercalcaemia is a common finding in the setting of primary


care,1 as well as in emergency departments2 and patients What is the prevalence of
admitted to hospital.3 Primary hyperparathyroidism and hypercalcaemia?
malignancy are the two most common causes of increased serum
calcium levels, together accounting for about 90% of all cases.4 Primary hyperparathyroidism is a relatively common endocrine
The remaining 10% represent an important figure, and thus the disorder, with an estimated prevalence of 1-7 cases per 1000
need to consider other disorders in the evaluation of patients adults.4 5 It is considered the most common cause of
with hypercalcaemia. This review aims to give an overview of hypercalcaemia, predominantly affecting the older population
the diagnosis and clinical management of hypercalcaemia for (≥65 years) and women two or three times more frequently than
non-specialist clinicians and health professionals. men.4 5 The incidence of primary hyperparathyroidism is poorly
defined; evidence from the United Kingdom5 and the United
What is the definition of hypercalcaemia? States6 shows a wide range, from 0.41 to 21.6 cases per 100 000
Hypercalcaemia is diagnosed when the concentration of serum population annually. This variety arises from heterogeneity in
calcium is 2 standard deviations above the mean of values found screening methods, case definition, and population studied, as
in people with normal calcium levels, in at least two samples well as unexplained annual fluctuations in incidence within the
at least one week apart over a period of three months. The serum same population.7
concentration of total calcium in adults usually ranges between Data on the prevalence and incidence of hypercalcaemia from
2.15 and 2.60 mmol/L (8.6-10.4 mg/dL; 4.3-5.2 mEq/L). About other causes are poor. Malignancy associated hypercalcaemia
45% of calcium in blood is bound to plasma proteins, is estimated to affect 2.7% of people with cancer in the USA.8
particularly albumin, and approximately 10% is bound to anions Data in children with cancer indicates a lower frequency of
such as phosphate and citrate; free or ionised calcium (normal hypercalcaemia (about 0.5-1%).9
values 1.17-1.33 mmol/L) represents about 45% of total calcium.
Although the ionised fraction of calcium is the one readily
available for activating cellular processes, measurement of total
What causes hypercalcaemia?
serum calcium is mostly requested in clinical practice. However, The box summarises the most common causes of
when the serum protein concentration fluctuates, the total serum hypercalcaemia. Mechanisms associated with hypercalcaemia
calcium level may vary accordingly, while ionised calcium are classically divided into parathyroid hormone and
remains stable. Total serum calcium can then be derived from non-parathyroid hormone mediated.
the formula: total calcium
concentration(mmol/L)+0.02(40−serum albumin Parathyroid hormone mediated
concentration(g/L). hypercalcaemia
Changes in blood pH can alter the equilibrium constant of the Parathyroid related causes of hypercalcaemia comprise primary
albumin-ionised calcium complexes, with acidosis reducing the (including the various genetic forms) and tertiary
binding and alkalosis enhancing it. When major changes in hyperparathyroidism. Parathyroid hormone is the main regulator
serum proteins or pH are suspected, measurement of ionised of calcium homeostasis and its primary increased secretion alters
calcium is recommended to determine the physiological serum the regulation of serum calcium by acting on different target
calcium level. organs (bone, kidney, gut).
A particular genetic form is represented by familial benign
hypocalciuric hypercalcaemia. This disorder results from altered
calcium sensing receptor function and a decreased sensitivity
to increases in extracellular calcium; the latter determines an
impaired suppression of parathyroid hormone secretion by the

Correspondence to: S Minisola salvatore.minisola@uniroma1.it

For personal use only: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe
BMJ 2015;350:h2723 doi: 10.1136/bmj.h2723 (Published 2 June 2015) Page 2 of 9

CLINICAL REVIEW

The bottom line


• Primary hyperparathyroidism and malignancy are the two most common causes of increased serum calcium levels
• The diagnosis of hypercalcaemia is made when the corrected serum calcium concentration is 2 standard deviations above the mean
of values found in people with normal calcium levels, in at least two samples at least one week apart over a period of three months
• The presence of high or not adequately suppressed serum parathyroid hormone levels should point the diagnosis towards
hypercalcaemia of parathyroid origins
• Mild hypercalcaemia is usually caused by primary hyperparathyroidism, the treatment for which is typically surgery; those aged 50 or
more with serum calcium levels <0.25 mmol/L above the upper limit of normal and without end organ damage may be followed up
conservatively. Treatment with a calcimimetic agent, cinacalcet, is an option in selected cases
• Severe hypercalcaemia requires admission to hospital and treatment with aggressive intravenous hydration and bisphosphonates
along with treatment of the underlying disease

Sources and selection criteria


We carried out a search through Medline and PubMed of articles published from 1990 to 2015 using the terms “mild hypercalcaemia” and
“severe “hypercalcemia”, “primary hyperparathyroidism”, “hypercalcemia of malignancy”, “parathyroid hormone measurement”,
“parathyroidectomy”, and “cinacalcet” and through the National Cancer Institute using the term “hypercalcaemia”. We also retrieved personal
archived references to identify peer reviewed articles. We gave priority to randomised controlled trials, systematic reviews, meta-analyses,
and prospective epidemiological studies. As appropriate we also included observational, retrospective, and non-randomised studies and
case reports.

Box Common causes of hypercalcaemia. Adapted from Minisola et al26


Parathyroid hormone mediated
• Sporadic (adenoma, hyperplasia, or carcinoma)
• Familial (multiple endocrine neoplasia 1, 2a, or 4, hyperparathyroidism jaw tumour syndrome, familial isolated hyperparathyroidism,
familial hypocalciuria hypercalcaemia)
• Ectopic parathyroid hormone in malignancy (rare)
• “Tertiary” hyperparathyroidism

Malignancy
• Humoral hypercalcaemia of malignancy (parathyroid hormone related protein)
• Local osteolysis (cytokines, chemokines, parathyroid hormone related protein)
• Ectopic parathyroid hormone in malignancy (rare)
• Calcitriol related hypercalcaemia

Vitamin D related
• Granulomatous disease (for example, sarcoidosis, tuberculosis, berylliosis, coccidiodomycosis, histoplasmosis, leprosy, inflammatory
bowel disease, foreign body granuloma)
• Vitamin D intoxication (vitamin D supplements, metabolites, or analogues)

Endocrine disorders
• Thyrotoxicosis
• Adrenal insufficiency
• Pheochromocytoma
• VIPoma (Verner-Morrison) syndrome

Drugs
• Thiazide diuretics
• Lithium
• Milk-alkali syndrome (calcium and antacids)
• Vitamin A
• Parathyroid hormone

Other
• Coexisting malignancy and primary hyperparathyroidism
• Immobilisation
• Acute renal failure
• Chronic renal failure treated with calcium and calcitriol or vitamin D analogues
• Renal transplant

parathyroid cells and continuous reabsorption of calcium by the Lithium induced hypercalcaemia could be considered as a
kidney tubules. As a consequence, such people develop reversible form of parathyroid hormone mediated
hypocalciuria, with tubular calcium reabsorption being increased hypercalcaemia. Lithium can directly stimulate parathyroid
by parathyroid hormone as well. hormone secretion and increase renal calcium reabsorption;
these effects may be reversed by withdrawal of the drug.

For personal use only: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe
BMJ 2015;350:h2723 doi: 10.1136/bmj.h2723 (Published 2 June 2015) Page 3 of 9

CLINICAL REVIEW

Non-parathyroid hormone mediated Malignancy related hypercalcaemia


hypercalcaemia The occurrence of hypercalcaemia together with systemic
Hypercalcaemia of non-parathyroid origin is mostly related to symptoms (for example, fever, weight loss, decreased appetite,
production of parathyroid hormone related protein, calcitriol, worsening malaise) or rapid onset hypercalcaemia, typically
or cytokines as mediators (box). with very high serum calcium levels, should raise suspicion of
Malignancy related hypercalcaemia—humoral hypercalcaemia malignancy. In particular, suppressed or undetectable serum
of malignancy is a paraneoplastic syndrome resulting from the parathyroid hormone levels are found in the setting of
secretion of parathyroid hormone related protein by the tumour.10 hypercalcaemia of malignancy. Hypercalcaemia is usually a
Although any kind of neoplasia may cause the syndrome of late finding in malignancy and therefore the underlying disease
humoral hypercalcaemia of malignancy, squamous carcinomas is often known when it occurs. If the primary malignancy is
are most commonly implicated. Hypercalcaemia may be due to unknown, the need for a rapid differential diagnosis is critical,
local osteolysis, most usually observed in haematological as hypercalcaemia represents a negative prognostic factor in
cancers. Overproduction of calcitriol represents the key people with cancer. Physical examination (including lymph
mechanism in the development of hypercalcaemia associated nodes, rectal, breasts, gynaecological, mouth, and ear, nose, and
with some forms of malignancy and with granulomatous diseases throat) should be performed, as well as laboratory assessment
(box). Malignant cells and granulomas can over-express including blood count, biochemistry, serum tumour markers,
1-α-hydroxylase and increase the conversion of calcidiol to the chest, abdomen, and pelvis imaging. Serum and urinary
active form of vitamin D, calcitriol, leading to increased immunoelectrophoresis are recommended, whereas in selected
intestinal absorption of calcium, hypercalciuria, and cases, measurement of serum parathyroid hormone related
hypercalcaemia. Finally, authentic ectopic hyperparathyroidism protein (where available)19 and calcitriol might be of value. The
is a rare cause of hypercalcaemia, with few cases described in finding of an increased parathyroid hormone related protein
the literature.10 level implies the need to search for solid tumours (lung,
oesophagus, skin, cervix, breast, and kidney are the most
Thiazide diuretics—thiazides are commonly prescribed and can
commons sites)19 (see box). High serum calcitriol levels are
increase renal reabsorption of calcium, resulting in hypocalciuria
typically associated with lymphoproliferative and granulomatous
and eventually high serum calcium levels. As many as 8% of
disorders (table). In this context, diagnoses should be considered
people develop hypercalcaemia while taking thiazides.11 12
even in the setting of normal serum calcitriol levels when
Endocrine disorders—hypercalcaemia is relatively common in parathyroid hormone and parathyroid hormone related protein
people with hyperthyroidism and is probably related to increases levels are suppressed. The production of the active form of
in RANK-ligand mediated bone resorption, stimulated by an vitamin D is no longer subject to regulation by parathyroid
excess of thyroid hormones.10 Hypercalcaemia can be sustained hormone or parathyroid hormone related protein, but rather
by primary hyperparathyroidism in patients with primarily driven in these conditions by the underlying disease.
pheochromocytoma in the setting of multiple endocrine
neoplasia type 2. Some pheochromocytomas have been found Parathyrotoxic crisis
to secrete parathyroid hormone related protein or to directly
stimulate bone resorption.10 Hypercalcaemia is not a common Although acute and severe hypercalcaemia is mostly associated
finding in Addisonian crisis, but it could occur in association with malignancy, the measurement of parathyroid hormone
with the underlying disorder, such as tuberculosis; reduction in levels has a key role in excluding parathyrotoxic crisis, the
the extracellular fluid volume associated with the relative association of which with parathyroid carcinoma has been
hyperalbuminaemia may lead to factitious hypercalcaemia.10 described in case reports.20 21 The clinical presentation is severe
and patients require admission to hospital. Parathyrotoxic crisis
Acute renal failure and rhabdomyolysis—patients with comprises profound volume depletion, coma, heart failure, and
rhabdomyolysis associated acute renal failure may develop abdominal pain possibly mimicking acute abdomen.
hypercalcaemia. The hypercalcaemia is probably related to
severe secondary hyperparathyroidism in the acute, oliguric Hyperthyroid activity can be associated with hypercalcaemia
phase—the effect of parathyroid hormone on bone along with and suppressed parathyroid hormone serum levels. Evaluation
the release of calcium phosphate into soft tissues during the of thyroid function is therefore needed.
early hypocalcaemic and hyperphosphataemic phase. The use Mediators of hypercalcaemia are typically suppressed in
of calcium, calcitriol, or vitamin D analogues supplementation immobilisation hypercalcaemia; serum phosphorus levels are
in patients who have end stage renal disease or receive dialysis often high, thus helping to differentiate those cases from primary
may cause hypercalcaemia; in this setting, hypercalcaemia may hyperparathyroidism, where phosphorus levels are typically
also occur in association with tertiary hyperparathyroidism. low.
Finally, increased serum calcium levels may be observed after Hypercalcaemia associated with suppressed parathyroid
kidney transplantation.10 hormone, normal parathyroid hormone related protein, and high
Immobilisation hypercalcaemia—this arises from suppression or normal serum calcitriol levels strongly suggest the diagnosis
of bone formation and increased bone resorption, with of calcitriol mediated diseases (box); however, other possible
consequent loss of calcium from the skeleton and diagnoses should be taken into account. Levels of calcidiol
hypercalcaemia. Immobilisation hypercalcaemia is generally should also be checked, particularly in those whose clinical
observed when there is a concomitant cause of high bone presentation does not suggest the presence of malignancy.
turnover, such as in younger people. However, data from Clinical case reports suggest that the occurrence of vitamin D
observational and retrospective studies suggest that toxicity, although unusual, should be excluded, particularly
hypercalcaemia may develop in cases of prolonged when the consumption of high doses of exogenous vitamin D
immobilisation from different causes, such Parkinson’s disease is unrecognised.22 The misuse of high dose vitamin D
or prolonged intensive care for major burns.13 14 preparations (daily dosing rather than weekly or monthly) should
be investigated.

For personal use only: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe
BMJ 2015;350:h2723 doi: 10.1136/bmj.h2723 (Published 2 June 2015) Page 4 of 9

CLINICAL REVIEW

Thiazide diuretics hour urine collection for calcium and creatinine determination
In patients who were hypercalcaemic while taking thiazide should therefore be performed to calculate the calcium to
diuretics, serum calcium and parathyroid hormone levels should creatinine clearance ratio. As calcium excretion could possibly
be re-evaluated at least three weeks after withdrawal of the be decreased in association with vitamin D deficiency, the
drugs. In a population based study, about two third of patients accuracy of this evaluation implies the need for replenishment
who discontinued thiazides had persistence of hypercalcaemia, in deficient patients. Calcium to creatinine clearance values less
suggesting that primary hyperparathyroidism is common in than 0.01 are strongly indicative of familial hypocalciuric
those who develop hypercalcaemia while taking thiazides.12 hypercalcaemia and require an evaluation of family history of
hypercalcaemia and eventually screening of serum calcium in
family members. Serum magnesium could be helpful in pointing
How should hypercalcaemia be towards the differential diagnosis of familial hypocalciuric
investigated in primary care? hypercalcaemia, as it is typically in the high range of normal or
modestly increased in this condition. Genetic testing is useful
The primary goal in the differential diagnosis of hypercalcaemia
for confirmation of the diagnosis.16
is to determine the underlying mechanism.
Medical history should focus on the use of supplements and
How is hypercalcaemia treated?
drugs possibly causing hypercalcaemia (box) and include an
evaluation of family history aimed at identifying underlying Understanding the mechanism of hypercalcaemia is crucial for
genetic forms of primary hyperparathyroidism. the most efficient management. Regardless of the diagnosis, all
Clinicians need to evaluate carefully the severity of clinical patients with hypercalcaemia require hydration. The timing and
presentation, degree of hypercalcaemia, and timing of regimens of hydration strongly depend on the severity of the
development of the condition. It is clinically relevant to hypercalcaemia.
distinguish those people with mild hypercalcaemia from those
with a more severe form, as this could help in diagnosis and Mild hypercalcaemia
guiding further investigation. The table⇓ describes the clinical Mild hypercalcaemia (values not exceeding 0.25 mmol/L above
presentation of people with hypercalcaemia. Importantly, normal range or <3 mmol/L) is usually caused by primary
symptoms associated with chronic hypercalcaemia are related hyperparathyroidism. Adults aged 50 or more with primary
to severe forms—those with chronic mild hypercalcaemia are hyperparathyroidism, a serum calcium level less than 0.25
typically asymptomatic. mmol/L above the upper limit of normal, and without end organ
Contrary to what is observed among inpatients, hypercalcaemia damage may be followed up conservatively—that is, without
is most commonly attributable to primary hyperparathyroidism intervention and specific drugs. People with serum calcium
in the outpatient setting. In this context the finding of levels greater than 0.25 mmol/L above the normal range, even
pre-existing mild hypercalcaemia may suggest the diagnosis of if asymptomatic, should be referred for surgery. In addition,
primary hyperparathyroidism. However, the detection of mildly regardless of calcium levels, the most recent guidelines for
increased serum calcium levels on a routine biochemical panel asymptomatic people with primary hyperparathyroidism suggest
in asymptomatic people is also a common finding. a more complete evaluation of skeletal and renal complications,
The evaluation of “outpatients” with hypercalcaemia usually including imaging studies.23 24 Skeletal (osteoporosis, as
follows a stepwise diagnostic approach (figure⇓). Laboratory evaluated by bone mineral density measurement, fragility
evaluation should first include the confirmation of fractures) or renal involvement (nephrolithiasis or
hypercalcaemia by remeasuring serum calcium levels and nephrocalcinosis, creatinine clearance <60 mL/min, or
correcting for albumin or by measuring serum ionised calcium hypercalciuria >10 mmol/d associated with an increased risk of
wherever available. Renal function should also be evaluated. stone disease) and age less than 50 years are considered criteria
Second or third generation immunoradiometric parathyroid for surgery in people with primary hyperparathyroidism, even
hormone assays should be used, as they have been proved to when calcium levels are not greater than 0.25 mmol/L above
perform similarly and better than first generation assays15 16; the normal range.23 In those who decline surgery or are not
with a sensitivity in diagnosis of primary hyperparathyroidism suitable candidates for surgery, serum calcium and creatinine
ranging from 88% to 97%. Hence, confirmation of levels should be measured every year and bone density measured
hypercalcaemia in association with an increased or every one or two years, together with monitoring by renal
non-suppressed or normal parathyroid hormone concentration imaging. During follow-up, if the increase in serum calcium
suggests primary hyperparathyroidism as the most likely levels is greater than 0.25 mmol/L or there is renal or skeletal
diagnosis. involvement the patient should be referred for surgery.23
Assessment of vitamin D status is indicated, as low serum If surgery is not performed, or not indicated, patients should be
calcidiol (25(OH)D) levels are highly prevalent in people with encouraged to have an above average intake of fluids and avoid
primary hyperparathyroidism and have been associated with drugs, such as thiazide diuretics, that can increase plasma
many negative outcomes in cross sectional studies.16 Most recent calcium levels.24
guidelines suggest a cautious replenishment with supplemental Recently, cinacalcet, a calcimimetic agent, has been proved in
doses of vitamin D in case of hypovitaminosis17; however, since a prospective observational study to be effective in lowering
no data from large randomised controlled trials are currently serum calcium levels in people with sporadic and familial
available, there is no specific recommendation on the dose primary hyperparathyroidism, but it has no effects on other
regimen of vitamin D. Serum levels between 50 and 75 nmol/L features of primary hyperparathyroidism—that is, bone mineral
are considered the goal of treatment in these patients.17 18 density and hypercalciuria.25 Cinacalcet given orally in a dosing
The diagnosis of primary hyperparathyroidism should be regimen of 30-120 mg/d is generally well tolerated, with only
confirmed by ruling out familial hypocalciuric hypercalcaemia, nausea described as a common adverse event. The European
another possible cause of high serum calcium associated with Medicines Agency in 2008 and the US Food and Drug
high or unsuppressed serum parathyroid hormone (box). A 24 Administration in 2011 approved the use of cinacalcet in people
For personal use only: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe
BMJ 2015;350:h2723 doi: 10.1136/bmj.h2723 (Published 2 June 2015) Page 5 of 9

CLINICAL REVIEW

with primary hyperparathyroidism with specific indications. clodronate has been reported to be effective, with no or minimal
The EMA panel stated that cinacalcet can be an option in those toxicity. This form of treatment may avoid hospital stay and
where parathyroidectomy is indicated based on serum calcium overcome any possible problems related to difficult intravenous
levels but for whom surgery is otherwise “not clinically access.31
appropriate or contraindicated.” The FDA approves the use of Although bisphosphonates are proved to be effective in the
cinacalcet in primary hyperparathyroidism for people with treatment of hypercalcaemia, a drug with a rapid hypocalcaemic
severe hypercalcaemia who are unable to undergo effect, such as calcitonin, could be used when a prompt
parathyroidectomy. resolution is needed. Calcitonin inhibits bone resorption and
also decreases renal tubular reabsorption of calcium. Its onset
Severe hypercalcaemia of action is within two hours of being administered, but the
If serum calcium levels are moderately increased (3.0-3.5 effect is short, and drug tolerance commonly develops within
mmol/L), the type of treatment and timing for administering two days. Thus, calcitonin is used as an early treatment for
drugs should be guided by clinical manifestations. Admission severe hypercalcaemia until the onset of the hypocalcaemic
to hospital is required for people with severe hypercalcaemia effects of other drugs.32
(>3.5 mmol/L); emergency treatment includes aggressive A recent single harm intervention study in people with persistent
intravenous hydration with 3-4 litres of 0.9% saline daily, or hypercalcaemia of malignancy despite intravenous
1-2 litre bolus of 0.9% saline, followed by 200-250 mL saline bisphosphonate treatment showed that denosumab (120 mg
hourly.25 The rationale underlying hydration is that people with subcutaneously on days 1, 8, 15, and 29 and then every four
hypercalcaemia are often severely dehydrated, mainly because weeks) lowered serum calcium levels in 64% of patients within
of nephrogenic diabetes insipidus due to hypercalcaemia and 10 days.33 Denosumab is a fully human monoclonal antibody
reduced water intake. The latter results from anorexia, nausea, that binds to RANK-ligand and inhibits the maturation,
and vomiting induced by the hypercalcaemia itself as well as activation, and function of osteoclasts; it could be given even
the causative disease, such as neoplasia. Hydration alone may to those with a creatinine clearance of less than 30 mL/min.
be effective in slowly reducing serum calcium levels; however, Further trials with denosumab are needed before it can be
most commonly it is not the only treatment and may lead to broadly recommended for use in people with hypercalcaemia.
fluid overload. Caution is therefore needed to avoid excessive Haemodialysis (as well as peritoneal dialysis) against a low or
fluid loading in patients with cardiac and renal disease. In such zero calcium dialysate is a treatment option in cases of treatment
patients, it is important to assess serum electrolytes and to carry failure or when calcium levels are so high as to be life
out electrocardiography during treatment. threatening; patients already receiving haemodialysis or with
Loop diuretics, such as furosemide (frusemide), which could severe renal insufficiency may take advantage of this
theoretically enhance calcium excretion, may worsen electrolyte procedure.34
derangements and volume depletion when administered at high It should be borne in mind that in hypercalcaemia of malignancy
doses. Thus, even in patients with volume overload, loop treatment of the underlying malignancy will also reduce serum
diuretics should be used with caution. A recent review of calcium levels. Surgical removal of the lesion is currently the
randomised controlled trials, prospective single group trials, only cure for severe hypercalcaemic crisis associated with
systematic reviews, and meta-analyses shows limited or no parathyroid carcinoma, an extremely rare presentation requiring
evidence to support the use of loop diuretics in people with urgent admission to hospital.
hypercalcaemia.27
Different treatments need to be considered in people with
Since the major mechanism responsible for severe hypercalcaemia from other causes, such as vitamin D
hypercalcaemia is the increased bone resorption from activation intoxication or granulomatous disorders. Since in these cases,
of osteoclasts, bisphosphonates are the treatment of choice as the underlying cause is an increased production of calcidiol,
they inhibit the osteoclast’s’ activity. Pamidronate and drugs that enhance vitamin D metabolism, such as
zoledronic acid are approved by EMA and FDA for the treatment glucocorticoids, are indicated. Prednisone inhibits
of hypercalcaemia of malignancy, both having shown 1-α-hydroxylase and activates 24-hydroxylase, thus reducing
effectiveness in clinical trials.28 29 Head to head comparison of hypercalcaemia; it is usually administered orally at a dose of
pamidronate and zoledronic acid in two randomised controlled 20-40 mg daily. The effects are observed within 24-72 hours of
trials showed that zoledronic acid was superior to pamidronate starting treatment.
in both efficacy and duration of response.30 A single 15 minute
intravenous infusion of 4 mg of zoledronic acid in 100 mL of Contributors: SM conceived the review, helped to write the article,
isotonic saline, with adequate hydration, resulted in complete coordinated with other authors’ work, dealt with the editors, reviewed
normalisation of serum calcium levels in less than three days the final version of the article, and helped check the proofs. JP and SP
in 80-100% of patients. Zoledronic acid can be readministered helped to write the article and reviewed the final version. CC helped to
as necessary to control hypercalcaemia. The most common write the article, designed the figure and table, helped check the proofs,
reported side effects have been transient fever, myalgias, and and reviewed the final article. All authors are the guarantors.
infusion site reaction. Zoledronic acid is contraindicated in
Competing interests: We have read and understood the BMJ policy on
patients with creatinine clearance values lower than 30 mL/min;
declaration of interests and declare the following: none.
in this situation, dose reduction according to creatinine clearance
values could be an option. Ibandronate, a bisphosphonate with Provenance and peer review: Not commissioned; externally peer
lower renal toxicity, is approved by EMA for the treatment of reviewed.
malignancy related hypercalcaemia.
1 Dalemo S, Eggertsen R, Hjerpe P, et al. Long-term follow-up of patients with elevated
As the administering of intravenous bisphosphonates out of serum calcium concentrations in Swedish primary care. Scand J Prim Health Care
hospital can be troublesome, patients with hypercalcaemia and 2012;30:48-54.
2 Lindner G, Felber R, Schwarz C, et al. Hypercalcemia in the ED: prevalence, etiology,
end stage malignancy could benefit from subcutaneous and outcome. Am J Emerg Med 2013;31:657-60.
clodronate when discharged from hospital. Subcutaneous 3 National Cancer Institute. PDQ® Hypercalcemia. 2013. www.meb.uni-bonn.de/Cancernet/
CDR0000062737.html.

For personal use only: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe
BMJ 2015;350:h2723 doi: 10.1136/bmj.h2723 (Published 2 June 2015) Page 6 of 9

CLINICAL REVIEW

Questions for future research


• What is the epidemiology of primary hyperparathyroidism and hypercalcaemia from non-parathyroid causes in different geographical
areas of the world?
• What is a safe vitamin D serum level to target supplementation in patients with primary hyperparathyroidism who are deficient in
vitamin D?
• Randomised controlled trials to define the role and safety of denosumab use in the treatment of severe hypercalcemia: could denosumab
be a second line after bisphosphonates use or possible first line therapy?

Tips for non-specialists


• The diagnosis of hypercalcaemia is made when the corrected concentration of serum calcium is 2 standard deviations above the
mean of values found in people with normal calcium levels, in at least two samples at least one week apart over a period of three
months
• Measurement of serum parathyroid hormone and patient’s clinical presentation should be rapidly assessed, particularly in severe
hypercalcaemic states
• People presenting with a history of mild asymptomatic hypercalcaemia typically have a diagnosis of primary hyperparathyroidism and
could be managed in the outpatient setting
• People with severe, new onset hypercalcaemia with symptoms require admission to hospital for intravenous treatment and diagnosis

Additional educational resources


Resources for healthcare professionals
Rosen CJ. Primer on the metabolic bone diseases and disorders of mineral metabolism, 8th ed. Wiley-Blackwell, 2013—discusses the
pathophysiology and clinical concepts of hypercalcaemia
Bilezikian JP, Marcus R, Levine M, et al. Parathyroids, basic and clinical concepts. 3rd ed. Academic Press, 2014—discusses the
pathophysiology, differential diagnosis, and clinical algorithm for the management of hypercalcaemia
National Cancer Institute, med News. Hypercalemia (www.meb.uni-bonn.de/Cancernet/CDR0000062737.html) (no registration required)

Resources for patients


Mayoclinic.org (www.mayoclinic.org/diseases-conditions/hypercalcemia/basics/definition/con-20031513) (no registration
required)—discusses symptoms and causes of hypercalcaemia
BMJ Best Practice (http://bestpractice.bmj.com/best-practice/monograph/159/diagnosis.html)—(no registration required) provides a step
by step diagnostic approach of hypercalcaemia; can be a useful resource for patients and clinicians to look at together for shared decision
making

4 Potts JT Jr, Jüppner H. Disorders of the parathyroid gland and calcium homeostasis. In: 21 Dionisi S, Minisola S, Pepe J, et al. Concurrent parathyroid adenomas and carcinoma in
Longo DL, Fauci AS, Kasper DL, Hauser S, eds. Harrison’s principles of internal medicine. the setting of multiple endocrine neoplasia type 1: presentation as hypercalcemic crisis.
18th ed. McGraw-Hill, 2012:3096-120. Mayo Clin Proc 2002;77:866-9.
5 Yu N, Donnan PT, Murphy MJ, et al. Epidemiology of primary hyperparathyroidism in 22 Lowe H, Cusano NE, Binkley N, et al. Vitamin D toxicity due to a commonly available
Tayside, Scotland, UK. Clin Endocrinol 2009;71:485-93. “over the counter” remedy from the Dominican Republic. J Clin Endocrinol Metab 2011
6 Wermers RA, Khosla S, Atkinson EJ, et al. Incidence of primary hyperparathyroidism in ;96:291-5.
Rochester, Minnesota, 1993-2001: an update on the changing epidemiology of the disease. 23 Bilezikian JP, Brandi ML, Eastell R, et al. Guidelines for the management of asymptomatic
J Bone Miner Res 2006;21:171-7. primary hyperparathyroidism: summary statement from the Fourth International Workshop.
7 Yeh MW, Ituarte PH, Zhou HC, et al. Incidence and prevalence of primary J Clin Endocrinol Metab 2014;99:3561-9.
hyperparathyroidism in a racially mixed population. J Clin Endocrinol Metab 24 Silverberg SJ, Clarke BL, Peacock M, et al. Current issues in the presentation of
2013;98:1122-9. asymptomatic primary hyperparathyroidism: proceedings of the Fourth International
8 Gastanaga V, Jain R, Pirolli M, et al. Prevalence of hypercalcemia of malignancy in the Workshop. J Clin Endocrinol Metab 2014;99:3580-94.
United States. Projection methods using oncology electronic health records. Eur J Cancer 25 Schwarz P, Body JJ, Cap J, et al. The PRIMARA study: a prospective, descriptive,
2013;49:S302-3. observational study to review cinacalcet use in patients with primary hyperparathyroidism
9 McKay C, Furman WL. Hypercalcemia complicating childhood malignancies. Cancer in clinical practice. Eur J Endocrinol 2014;171:727-35.
1993;72:256-60. 26 Minisola S, Romagnoli E, Carnevale V, et al. Acute management of hypercalcemia. In:
10 Horwitz MJ, Hodak SP, Stewart AF. Non-parathyroid hypercalcemia. In: Rosen CJ, ed. Bilezikian JP, Marcus R, Levine M, Marcocci, C, Silverberg SJ, Potts J, eds. Parathyroids,
Primer on the metabolic bone diseases and disorders of mineral metabolism. 8th ed. basic and clinical concepts. 3rd ed. Academic Press, 2014:617-29.
Wiley-Blackwell, 2013:562-71. 27 LeGrand SB, Leskuski D, Zama I. Narrative review: furosemide for hypercalcemia: an
11 Grieff M, Bushinsky DA. Diuretics and disorders of calcium homeostasis. Semin Nephrol unproven yet common practice. Ann Intern Med 2008;149:259-63.
2011;31:535-41. 28 Kawada K, Minami H, Okabe K, et al. A multicenter and open label clinical trial of zoledronic
12 Wermers RA, Kearns AE, Jenkins GD, et al. Incidence and clinical spectrum of acid 4 mg in patients with hypercalcemia of malignancy. Jpn J Clin Oncol 2005;35:28-33.
thiazide-associated hypercalcemia. Am J Med 2007;120:911.e9-15. 29 Purohit OP, Radstone CR, Anthony C, et al. A randomised double-blind comparison of
13 Kohut B, Rossat J, Raffoul W, et al. Hypercalcaemia and acute renal failure after major intravenous pamidronate and clodronate in the hypercalcaemia of malignancy. Br J Cancer
burns: An under-diagnosed condition. Burns 2010;36:360-6. 1995;72:1289-93.
14 Sato Y, Honda Y, Iwamoto J, et al. Abnormal bone and calcium metabolism in immobilized 30 Major P, Lortholary A, Hon J, et al. Zoledronic acid is superior to pamidronate in the
Parkinson’s disease patients. Mov Disord 2005;20:1598-603. treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled
15 Carnevale V, Dionisi S, Nofroni I, et al. Potential clinical utility of a new IRMA for parathyroid clinical trials. J Clin Oncol 2001;19:558-67.
hormone in postmenopausal patients with primary hyperparathyroidism. Clin Chem 31 Roemer-Bécuwe C, Vigano A, Romano F, et al. Safety of subcutaneous clodronate and
2004;50:626-31. efficacy in hypercalcemia of malignancy: a novel route of administration. J Pain Symptom
16 Eastell R, Brandi ML, Costa AG, et al. Diagnosis of asymptomatic primary Manage 2003;26:843-8.
hyperparathyroidism: proceedings of the fourth international workshop. J Clin Endocrinol 32 Reagan P, Pani A, Rosner MH. Approach to diagnosis and treatment of hypercalcemia
Metab 2014;99:3570-9. in a patient with malignancy. Am J Kidney Dis 2014;63:141-7.
17 Marcocci C, Bollerslev J, Khan AA, et al. Medical management of primary 33 Hu MI, Glezerman IG, Leboulleux S, et al. Denosumab for treatment of hypercalcemia of
hyperparathyroidism: proceedings of the fourth International Workshop on the Management malignancy. J Clin Endocrinol Metab 2014;99:3144-52.
of Asymptomatic Primary Hyperparathyroidism. J Clin Endocrinol Metab 2014;99:3607-18. 34 Kindgen-Milles D, Kram R, Kleinekofort W, et al. Treatment of severe hypercalcemia using
18 Minisola S, Romagnoli E, Scillitani A, et al. Hypovitaminosis D in primary continuous renal replacement therapy with regionalcitrate anticoagulation. ASAIO J
hyperparathyroidism: to treat or not to treat? That is the question. J Endocrinol Invest 2008;54:442-4.
2014;37:413-4.
19 Streeten EA, Jaimungal S. The differential diagnosis of hypercalcemia. The parathyroids.
3rd ed. Academic Press, 2014:607-16. Cite this as: BMJ 2015;350:h2723
20 Minisola S, Romagnoli E, Scarnecchia L, et al. Parathyroid storm: immediate recognition
© BMJ Publishing Group Ltd 2015
and pathophysiological considerations. Bone 1993;14:703-6.

For personal use only: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe
BMJ 2015;350:h2723 doi: 10.1136/bmj.h2723 (Published 2 June 2015) Page 7 of 9

CLINICAL REVIEW

For personal use only: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe
BMJ 2015;350:h2723 doi: 10.1136/bmj.h2723 (Published 2 June 2015) Page 8 of 9

CLINICAL REVIEW

Table

Table 1| Clinical presentation of hypercalcaemia

System Acute hypercalcaemia Chronic hypercalcaemia


General Flushing, fatigue, weight loss Fatigue
Cardiovascular Prolonged PR interval, widened QRS complex, shortened QT interval, Prolonged PR interval, widened QRS complex, shortened QT interval,
bundle branch block, bradycardia, arrhythmias, syncope, cardiac arrest bundle branch block, bradycardia, arrhythmias, hypertension, valvular
heart disease, vascular calcification
Renal Thirst, polydipsia; dehydration; polyuria; nocturia; frequent urination; Nephrocalcinosis, nephrolithiasis, chronic renal failure, renal
renal failure from obstructive uropathy, nephrolithiasis, osteodystrophy
nephrocalcinosis, or pre-renal causes
Neurological Tiredness, obtundation, lethargy, confusion, delirium, somnolence, Dementia, memory loss, sleep disturbance, decreased concentration
stupor, coma, hypotonia, hyporeflexia, paresis
Psychiatric Irritability, depression, anxiety, hallucination, psychosis Irritability, depression, anxiety
Gastrointestinal Anorexia, nausea, vomiting, abdominal pain, dyspepsia, constipation, Anorexia, dyspepsia, constipation, pancreatitis, peptic ulcer
pancreatitis, peptic ulcer
Skeletal and muscle Bone pain, muscle weakness Bone pain, muscle weakness, myalgias, osteoporosis, osteopenia,
fragility fractures, osteitis fibrosa cystica, bone cysts, brown tumours
of long bones, condrocalcinosis, joint calcification
Haematological Anaemia Anaemia
Ocular — Band keratopathy (cornea)

For personal use only: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe
BMJ 2015;350:h2723 doi: 10.1136/bmj.h2723 (Published 2 June 2015) Page 9 of 9

CLINICAL REVIEW

Figure

Suggested algorithm for diagnosis of hypercalcaemia; based on available evidence, mostly derived from retrospective or
observational, non-randomised, non-blinded studies. The algorithm also underlines the need for clinical evaluation as a
key guide for diagnosis and management in any given patient. Corrected calcium (mmol/L)=total calcium concentration
(mmol/L)+0.02(40−serum albumin concentration (g/L). Serum ionised calcium (Ca2+) should be directly measured, whenever
available, through the ion specific electrode and could increase accuracy of diagnosis. GFR=glomerular filtration rate

For personal use only: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe
ARTIGO 7

SÍNDROMES CARDIORRENAIS
Review
Journal of the Royal Society of Medicine; 2016, Vol. 109(1) 12–17
DOI: 10.1177/0141076815616091

Cardiorenal syndrome: review of our current


understanding

Savvas Hadjiphilippou and Sui Phin Kon


King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK
Corresponding author: Sui Phin Kon. Email: suiphin.kon@nhs.net

Summary poor prognosis. Concurrent renal disease and HF,


given their close bi-directional relationship, are as a
Renal and cardiac diseases are both prevalent and carry
significant morbidity and mortality. They share common consequence associated with a worse prognosis1 and
vascular risk factors and are physiologically interlinked. hence a greater burden on our healthcare system and
Dysfunction in one organ affects the other. Concurrent patient quality of life. The existence of this dual dys-
renal and cardiac disease is associated with a poor progno- function has been recently termed as ‘cardiorenal
sis. This close relationship is reflected through cardiorenal syndrome’.
syndrome. A classification system has been proposed; how- Cardiorenal syndrome has been difficult to define
ever, the underlying process is complex and multifactorial. because it encompasses complex multifactorial facets
Management of this syndrome focuses on improving heart including physiological, biochemical and hormonal.
function, reducing volume overload, and managing heart Following a consensus conference in 2008,2 the
failure and chronic kidney disease. This, however, is chal- report defines cardiorenal syndrome as ‘disorders of
lenging, limited by paucity of evidence and may lead to the heart and kidneys whereby acute or chronic dys-
suboptimal therapy. Increased recognition of this syndrome
function in one organ may induce acute or chronic
should raise awareness in providing early therapy and
avoiding adverse outcomes due to under-treatment. In
dysfunction of the other’.
this article, we provide an overview of our current under- The purpose of this article is to provide an over-
standing of cardiorenal syndrome, as well as its pathophysi- view of our current understanding of cardiorenal syn-
ology and treatment options. drome as well as its pathophysiology and treatment
options.
Keywords
Heart failure, acute kidney injury, chronic kidney disease,
cardiorenal syndrome, pathophysiology Methods
Within this review, we aimed to summarise the results
of a MEDLINE, PubMed, Cochrane Library,
Google and Google Scholar search (from January
1931 to September 2014) on the current understand-
Introduction ing of the epidemiology, pathophysiology and treat-
Within the human body, interactions between various ment of cardiorenal syndrome. Databases were
organs are tightly controlled. The heart is responsible searched using the term ‘cardiorenal syndrome’, and
for ensuring that blood is circulated within the body, articles were selected and analysed according to their
including to the kidneys, while the kidneys are relevance.
responsible for filtering the circulating blood and
managing electrolyte homeostasis. Both organs are
Epidemiology
undoubtedly crucial to our survival, tightly linked
and inevitably inter-reliant. The maintenance of car- Renal dysfunction frequently coexists with HF. The
diovascular haemostasis is therefore dependent upon Acute Decompensated Heart Failure National
fine interactions between the heart and kidneys. It is Registry demonstrated that 30% of patients admitted
thus not surprising that dysfunction in one organ will with acute decompensated HF had chronic kidney
affect the other. disease (CKD) and 21% had a creatinine of >2 mg/
Indeed, cardiac disease and renal disease share dL.3 In a study by Forman et al.,4 irrespective of HF,
common vascular risk factors (hypertension and dia- with or without preserved ejection fraction, raised
betes to name but a few). Both renal disease and heart serum creatinine on admission to hospital and wor-
failure (HF) are independently associated with a sening renal impairment during admission were both

! The Royal Society of Medicine 2016


Reprints and permissions: sagepub.co.uk/journalsPermissions.nav
Hadjiphilippou and Kon 13

Table 1. Classification of cardiorenal syndrome based on a system proposed by Ronco and McCullough.2

Cardiorenal syndrome type Characteristics

Type 1 (acute cardiorenal) Acute cardiac impairment leading to acute kidney injury

Type 2 (chronic cardiorenal) Chronic cardiac impairment leading to renal impairment

Type 3 (acute renocardiac) Acute kidney injury leading to cardiac impairment

Type 4 (chronic renocardiac) Chronic kidney disease leading to cardiac impairment

Type 5 (secondary cardiorenal) Systemic condition leading to both cardiac and renal impairment

associated with prolonged inpatient stay and and has been reported in 63% of patients admitted
increased mortality. with congestive HF.2,8 The mechanism underlying
Worse baseline renal function is a powerful inde- this process is likely to be due to chronic renal hypo-
pendent risk factor for adverse cardiovascular out- perfusion although there is limited evidence as of yet
comes.5 However, not all studies show an adverse to suggest how left ventricular function correlates to
association with declining glomerular filtration rate GFR levels.7
(GFR). The Evaluation Study of Congestive Heart
Failure and Pulmonary Artery Catheterisation
Acute renocardiac syndrome (type 3)
Effectiveness trail did not show statistical significance
or increased mortality with a 25% decline in GFR.6 Cardiorenal syndrome type 3 is characterised by
This reflects that a decline in kidney function due to acute cardiac dysfunction (uraemic cardiomyopathy,
adequate diuresis or blockade of the renin–angioten- arrhythmias due to hyperkalaemia) as a result of
sin–aldosterone system (RAAS) may be associated acute renal impairment. Defining the epidemiology
with improved outcomes. in this subtype has proven a challenge due to different
methods for defining AKI, different baseline risks for
developing acute cardiac dysfunction and limited
Classification
reporting by studies of AKI on the incidence of
Recognising the bi-directional relationship between acute cardiac dysfunction as an outcome measure.2
cardiac and renal dysfunction, the classification
system from the 2008 consensus conference2 is five-
part and recognises that patients may move between
Chronic renocardiac syndrome (type 4)
subtypes during the course of their disease (Table 1). Cardiorenal syndrome type 4 describes CKD leading
to cardiac dysfunction (left ventricular failure or dia-
stolic HF). Cardiac disease in patients with CKD is
Acute cardiorenal syndrome (type 1)
common, and adverse cardiac outcomes correlate
Cardiorenal syndrome type 1 is characterised by well with severity of CKD.2
acute worsening of cardiac function (pulmonary
oedema, cardiogenic shock, acute HF) leading to an
acute kidney injury (AKI). Approximately 27% of
Secondary cardiorenal syndromes (type 5)
patients admitted with acute decompensated HF Cardiorenal syndrome type 5 is characterised by sim-
develop AKI.4 The challenge in this subtype is the ultaneous cardiac and renal dysfunction as a part of a
early identification of an AKI as creatinine will systemic condition whether that may be acute or
increase once AKI is established and early bio- chronic. This most commonly includes systemic con-
markers such as neutrophil gelatinase-associated lipo- ditions such as sepsis and less so others such as amyl-
calin have been studied as early predictors of AKI.7 oid or vasculitis.

Chronic cardiorenal syndrome (type 2) Pathophysiology


Cardiorenal syndrome type 2 is characterised by A number of mechanisms have been identified
chronic cardiac dysfunction leading to renal dysfunc- as having a role in the development of cardiorenal
tion and can be used to describe chronic HF leading syndrome. The pathogenesis is complex involving
to renal failure. This syndrome is the most common multiple pathways which have not yet been fully
14 Journal of the Royal Society of Medicine 109(1)

elucidated; however, a number of mechanisms have cardiorenal syndrome individually. Instead, treat-
been proposed. Detailed descriptions of these are ment strategies focus on managing the predominant
beyond the scope of this article; however, chronic underlying condition, whether that is cardiac or
over-activation of the neurohormonal systems and renal. As our understanding of the pathophysiology
increased venous pressure are thought to be import- improves, we can look forward to more targeted
ant factors. therapies.
In this article, the management of cardiorenal syn-
drome has not been divided by subtype. This reflects
Neurohormonal system the close interrelations between the different subtypes
Initial triggering of the neurohormonal systems such and in particular how therapies targeted to one sub-
as the RAAS, the sympathetic nervous system (SNS), type can also have a beneficial effect for other sub-
the arginine-vasopressin system and the endothelin types, such as the use of RAAS blockade in type 2
system is thought to be protective and aims to main- and 4. In general, however, within subtypes 1 and 2,
tain homeostasis. The chronic activation, however, of therapy predominantly focuses on managing acute
all these mechanisms can have deleterious effects on and chronic HF. For subtype 3, management of the
both the cardiac and renal system. underlying cause of AKI is most appropriate, while in
Activation of the RAAS in the kidney leads to type 4, management is challenging due to the multi-
sodium and water retention, systemic vasoconstric- factorial nature of HF secondary to CKD. Therapies
tion and further reduced glomerular filtration. for type 5 focus primarily on treating underlying
The RAAS activation increases oxidative stress and pathology such as sepsis, amyloid or vasculitis.
further downstream profibrotic neurohormonal We have subdivided treatment options therefore
enhancement resulting in ventricular remodelling.9 into the three following areas:
Chronic SNS over-activation results in reduced car-
diac b-adrenoceptor density and sensitivity,10 cardio- 1. Improving cardiac function:
myocyte hypertrophy11 and, through the
enhancement of neuropeptide Y, vascular neointimal . Inotropes
formation.12 Impaired left ventricular function fur-
ther contributes to decreased forward flow. The use of inotropic drugs is relevant in the treat-
ment of cardiogenic shock. However, the hypothesis
that use of intravenous milrinone in patients with HF
Increased venous pressure in order to permit more effective diuresis has been
HF is marked by an increase in central venous pres- refuted.14 Similarly, no clinical benefit has been
sure which leads to a reduction in the perfusion gra- demonstrated with low-dose dopamine when used
dient across the glomerular capillary bed and decline for the treatment or prevention of AKI.15
in renal function. The result of increased back pres-
sure and its effects on urine formation was first stu- . Vasodilators
died around a century ago. In an experiment by
Winton,13 it was noted that urine formation was Vasodilators such as nitrates form part of the
reduced with rising pressure. Right ventricular dila- treatment for HF. The effects of nesiritide (a recom-
tation and dysfunction from elevated venous pressure binant human brain natriuretic peptide) on renal
further impairs left ventricular filling. function in the treatment of acute decompensated
HF are conflicting. The Acute Study of Clinical
Effectiveness of Nesiritide in Decompensated Heart
Management
Failure trial found no change in risk of worsening
The management of cardiorenal syndrome can be renal function with nesiritide therapy, but it was asso-
challenging. Treatment for HF may worsen renal ciated with higher rates of hypotension.16
function, and thus patients may receive suboptimal
therapy impacting on their prognosis. Primary pre- . Cardiac resynchronisation
vention cannot be overemphasised for both renal and
cardiac disease as they share common risk factors. In a study by Van Bommel et al.,17 patients with
This includes blood pressure, cholesterol and glucose HF who had a GFR of <60 ml/min/1.73 m2 demon-
management as well as physical activity and smoking strated a reduced response to cardiac resynchronisa-
cessation. Although established guidelines exist for tion therapy (CRT), defined as a decrease in left
the management of HF and CKD separately, there ventricular end-systolic volume >15% at six-month
is as of yet no consensus on the management of each follow-up, compared to those with normal renal
Hadjiphilippou and Kon 15

function. Worse renal function was also associated licensed for the treatment of hyponatraemia second-
with worse long-term prognosis after CRT. Despite ary to the syndrome of inappropriate antidiuretic
this, a systematic review by Garg et al.18 demon- hormone secretion.
strated that CRT in patients with CKD led to an
improvement in left ventricular ejection as well as . Ultrafiltration
GFR. More studies are required to evaluate this fur-
ther; however, this suggests a degree of reversibility in In recent years, ultrafiltration has had an increas-
cardiorenal syndromes 1 and 2. ing role in patients with decompensated HF and renal
dysfunction, particularly affecting cardiorenal syn-
. Beta-blockers drome subtype 2. Three randomised trials
(Ultrafiltration versus Intravenous Diuretics for
Beta-blockers are avoided in acute HF but used in Patients Hospitalised for Acute Decompensated
the management of chronic HF. In a systematic Congestive Heart Failure [UNLOAD], Relief for
review by Badve et al.,19 of patients with HF and Acutely Fluid-Overloaded Patients With
CKD, the use of b-blockers reduced the risk of all- Decompensated Congestive Heart Failure [RAPID-
cause and cardiovascular mortality. However, it was CHF], Cardiorenal Rescue Study in Acute
associated with an increase in bradycardia and hypo- Decompensated Heart Failure [CARESS-HF]) have
tension. Caution is to be exercised in their use in compared diuretic therapy in patients with acute
acute decompensated HF as they may further decompensated HF.23–25 In UNLOAD and RAPID-
reduce forward flow and exacerbate renal CHF, ultrafiltration was associated with greater fluid
dysfunction. loss than diuretic therapy but no difference in serum
creatinine, while in CARESS-HF weight loss was
. Adenosine-receptor antagonists similar, but there was an increase in serum creatinine
and greater adverse events. The 2013 American
These drugs increase GFR by causing vasodilata- College of Cardiology Foundation/American Heart
tion and increased renal perfusion. However, data Association guidelines for the management of HF
from the PROTECT trial20 suggests no difference in stated that ‘‘ultrafiltration may be considered for
cardiovascular outcomes compared to placebo in the patients with refractory congestion not responding
treatment of acute HF with renal dysfunction. to medical therapy.’’26 Peritoneal dialysis has been
trialled for the symptomatic management of refrac-
2. Decreasing volume overload: tory dyspnoea in end-stage HF with paucity of data
from large randomised controlled trials.27 Additional
. Diuretics trials are ongoing to see the benefit of ultrafiltration.

First, low sodium intake needs to be reinforced. 3. Drugs for reduced ejection fraction and CKD:
Diuretics are the mainstay of management in fluid
overload in both HF and renal failure. Aggressive . Renin–angiotensin–aldosterone system antagonism
diuresis improves survival despite worsening renal
function.21 Intravenous diuretics, high dose loop RAAS antagonism is an integral part of therapy
diuretics and adding thiazides to loop diuretics are for acute and chronic HF and renal failure. Subgroup
needed to try to overcome diuretic resistance where analyses of clinical trials of RAAS antagonism in HF
necessary. Diuretics are particularly useful in the have demonstrated that the beneficial effects on mor-
management of type 1, 2 and 4 cardiorenal syndrome. bidity are not reduced by concurrent CKD.28 The
risk of hyperkalaemia and worsening renal function
. Vasopressin receptor antagonists is higher in patients with CKD28 and therefore
requires regular monitoring.
Tolvaptan is a selective vasopressin 2 receptor Similarly, aldosterone antagonists showed a bene-
antagonist whose effect on cardiovascular actions fit in cardiovascular outcome despite a fall in GFR29
was described in the Efficacy of Vasopressin with the same problem of hyperkalaemia.
Antagonism in Heart Failure Outcome Study With Angiotensin-converting enzyme inhibition and
Tolvaptan-Outcomes trial.22 The study showed no angiotensin receptor blockers are known to worsen
effect on long-term mortality or HF-related morbid- renal function prompting reluctance to prescribe and
ity. However, there was an increase in urine output a low threshold for stopping. They should not be
resulting in improved dyspnoea as it increased free discontinued as such patients had decreased mortality
water clearance. Currently in the UK, tolvaptan is despite a decline in GFR.30 Caution or specialist
16 Journal of the Royal Society of Medicine 109(1)

Table 2. Key points.

Key points

1 Renal disease and heart failure frequently coexist due to their close bi-directional relationship.

2 Pathophysiology is poorly understood; however, it carries a poor prognosis.

3 Management is challenging often leading to under-treatment. Current strategies focus on the predominant failing
organ rather than on both organs.

4 Recognition of this syndrome is crucial in providing early and appropriate therapy.

advice is appropriate; however, this should not be References


interpreted as withholding treatment, while renal 1. Schrier RQ. Cardiorenal versus renocardiac syndrome: is
impairment should not be a contraindication to there a difference? Nat Clin Pract Nephrol 2007; 3: 637.
RAAS blockade but instead should reinforce fre- 2. Ronco C and McCullough SD. Cardio-renal syn-
dromes: reports from the consensus conference of the
quent monitoring and a multidisciplinary manage-
acute dialysis quality initiative. Eur Heart J 2010; 31:
ment approach with nephrology specialists.
703–711.
3. Adams KF Jr, Fonarow GC, Emerman CL, LeJemtel
Conclusion TH, Costanzo MR, Abraham WT, et al. Characteristics
and outcomes of patients hospitalized for heart failure
There is a high prevalence of cardiorenal syndrome in the United States: rationale, design, and preliminary
where patients with HF have concurrent renal dys- observations from the first 100,000 cases in the Acute
function. Reduced renal function is associated with Decompensated Heart Failure National Registry
increased morbidity and mortality in this population. (ADHERE). Am Heart J 2005; 149: 209–216.
It is a bi-directional relationship where dysfunction in 4. Forman DE, Butler J, Wang Y, Abraham WT,
either the heart or kidneys worsens the function of O’Connor CM, Gottlieb SS, et al. Incidence, predictors
the other. Cardiorenal syndrome is difficult to define at admission, and impact of worsening renal function
because it has multiple contributing factors; however, among patients hospitalized with heart failure. J Am
potential mechanisms include neurohormonal system Coll Cardiol 2004; 43: 61–67.
5. Tonelli M, Wiebe N, Culleton B, House A, Rabbat C, Fok
activation leading to reduced forward flow and renal
M, et al. Chronic kidney disease and mortality risk: a sys-
perfusion and increased venous pressure. Medical
tematic review. J Am Soc Nephro 2006; 17: 2034–2047.
treatment options are primarily focused on improv- 6. Nohria A, Hasselblad V, Stebbins A, Pauly DF,
ing cardiac function, reducing volume overload and Fonarow GC, Shah M, et al. Cardiorenal interactions:
managing HF and CKD. Although management of insights from the ESCAPE trial. J Am Coll Cardiol
cardiorenal syndrome remains a challenge, increased 2008; 51: 1268–1274.
recognition of this syndrome should raise our aware- 7. Ronco C, Haapio M, House AA, Anavekar N and
ness in providing early therapy to improve patient Bellomo R. Cardiorenal syndrome. J Am Coll Cardiol
care and avoid adverse outcomes due to under-treat- 2008; 52: 1527.
ment (Table 2). 8. Heywood JT, Fonarow GC, Costanzo MR, Mathur
VS, Wigneswaran JR, Wynne J, et al. High prevalence
of renal dysfunction and its impact on outcome in
Declarations 118,465 patients hospitalized with acute decompen-
Competing interests: None declared sated heart failure: a report from the ADHERE data-
base. J Card Fail 2007; 13: 422–430.
Funding: None declared
9. Bock JS and Gottlieb SS. Cardiorenal syndrome: new
Ethical approval: Not required perspectives. Circulation 2010; 121: 2592–2600.
10. Bristow MR, Ginsburg R, Minobe W, Cubicciotti RS,
Guarantor: SPK Sageman WS, Lurie K, et al. Decreased catecholamine
sensitivity and b-adrenergic-receptor density in failing
Contributorship: Both SH and SPK contributed equally to the human hearts. N Engl J Med 1982; 307: 205–211.
literature search, analysis, design and write-up of this manuscript. 11. Amin JK, Xiao L, Pimental DR, Pagano PJ, Singh K,
Sawyer DB, et al. Reactive oxygen species mediate
Acknowledgements: None
alpha-adrenergic receptor-stimulated hypertrophy in
Provenance: Not commissioned; peer-reviewed by Muhammad adult rat ventricular myocytes. J Mol Cell Cardiol
Siddiqui. 2001; 33: 131–139.
Hadjiphilippou and Kon 17

12. Li L, Lee EW, Ji H and Zukowska Z. Neuropeptide 23. Bart BA, Boyle A, Bank AJ, Anand I, Olivari MT and
Y-induced acceleration of postangioplasty occlusion of Kraemer M. Ultrafiltration versus usual care for hos-
rat carotid artery. Arterioscler Thromb Vasc Biol 2003; pitalized patients with heart failure: the Relief for
23: 1204–1210. Acutely Fluid-Overloaded Patients With
13. Winton FR. The influence of venous pressure on the Decompensated Congestive Heart Failure (RAPID-
isolated mammalian kidney. J Physiol 1931; 72: 49–61. CHF) trial. J Am Coll Cardiol 2005; 46: 2043.
14. Cuffe MS, Califf RM, Adams KF Jr, Benza R, Bourge 24. Costanzo MR, Guglin ME, Saltzberg MT, Jessup ML,
R, Colucci WS, et al. Short-term intravenous milrinone Bart BA, Teerlink JR, et al. Ultrafiltration versus intra-
for acute exacerbation of chronic heart failure: a ran- venous diuretics for patients hospitalized for acute
domized controlled trial. JAMA 2002; 287: 1541–1547. decompensated heart failure. J Am Coll Cardiol 2007;
15. Kellum JA and Decker J. Use of dopamine in acute 49: 675.
renal failure: a metaanalysis. Crit Care Med 2001; 29: 25. Bart BA, Goldsmith SR, Lee KL, Givertz MM,
1526–1531. O’Connor CM, Bull DA, et al. Ultrafiltration in
16. O’Connor CM, Starling RC, Hernandez AF, decompensated heart failure with cardiorenal syn-
Armstrong PW, Dickstein K, Hasselblad V, et al. drome. N Engl J Med 2012; 367: 2296.
Effect of nesiritide in patients with acute decompen- 26. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE
sated heart failure. N Engl J Med 2011; 365: 32. Jr, Drazner MH et al. ACCF/AHA guideline for the
17. Van Bommel RJ, Mollema SA, Borleffs CJ, Bertini M, management of heart failure: A report of the American
Ypenburg C, Marsan NA, et al. Impaired renal func- College of Cardiology Foundation/American Heart
tion is associated with echocardiographic nonresponse Association Task Force on practice guidelines.
and poor prognosis after cardiac resynchronization Circulation 2013; 128: e240–327.
therapy. J Am Coll Cardiol 2011; 57: 549–555. 27. Khalifeh N, Vychytil A and Hörl WH. The role of
18. Garg N, Thomas G, Jackson G, Rickard J, Nally JV Jr, peritoneal dialysis in the management of treatment-
Tang WH, et al. Cardiac resynchronization therapy in resistant congestive heart failure: a European perspec-
CKD: a systematic review. Clin J Am Soc Nephrol tive. Kidney Int Suppl 2006; 70: S72–S75.
2013; 8: 1293–1303. 28. Anand IS, Bishu K, Rector TS, Ishani A, Kuskowski
19. Badve SV, Roberts MA, Hawley CM, Cass A, Garg MA and Cohn JN. Proteinuria, chronic kidney disease,
AX, Krum H, et al. Effects of beta-adrenergic antag- and the effect of an angiotensin receptor blocker in
onists in patients with chronic kidney disease: a system- addition to an angiotensin-converting enzyme inhibitor
atic review and meta-analysis. J Am Coll Cardiol 2011; in patients with moderate to severe heart failure.
58: 1152–1161. Circulation 2009; 120: 1577.
20. Massie BM, O’Connor CM, Metra M, Ponikowski P, 29. Rossignol P, Cleland JG, Bhandari S, Tala S,
Teerlink JR, Cotter G, et al. Rolofylline, an adenosine Gustafsson F, Fay R, et al. Determinants and conse-
A1-receptor antagonist, in acute heart failure. N Engl J quences of renal function variations with aldosterone
Med 2010; 363: 1419. blocker therapy in heart failure patients after myocar-
21. Testani JM, Chen J, McCauley BD, Kimmel SE and dial infarction: insights from the Eplerenone Post-
Shannon RP. Potential effects of aggressive deconges- Acute Myocardial Infarction Heart Failure Efficacy
tion during the treatment of decompensated heart fail- and Survival Study. Circulation 2012; 125: 271–279.
ure on renal function and survival. Circulation 2010; 30. Testani JM, Kimmel SE, Dries DL and Coca SG.
122: 265. Prognostic importance of early worsening renal func-
22. Konstam MA, Gheorghiade M, Burnett JC Jr, tion after initiation of angiotensin-converting enzyme
Grinfeld L, Maggioni AP, Swedberg K, et al. Effects inhibitor therapy in patients with cardiac dysfunction.
of oral tolvaptan in patients hospitalized for worsening Circ Heart Fail 2011; 4: 685–691.
heart failure: the EVEREST outcome trial. JAMA
2007; 297: 1319.
ARTIGO 8

TESTE DE ESTRESSE COM


FUROSEMIDA
REVIEW

Renal Stress Testing in the Assessment


of Kidney Disease
Lakhmir S. Chawla1,2 and Claudio Ronco3,4
1
Department of Medicine, Veterans Affairs Medical Center, Washington, DC, USA; 2Department of Medicine, George Wash-
ington University, Washington, DC, USA; 3Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital,
Vicenza, Italy; and 4International Renal Research Institute of Vicenza, Vicenza, Italy

As part of human evolutionary development, many human organ systems have innate mechanisms to
adapt to increased “work demand” or stress. This reserve capacity can be informative and is used
commonly in cardiology to assess cardiac function (e.g., treadmill test). Similarly, the kidney possesses
reserve capacity, which can be demonstrated in at least 2 of the following renal domains: glomerular and
tubular. When appropriate stimulants are used, healthy patients with intact kidneys can significantly in-
crease their glomerular filtration rate and their tubular secretion. This approach has been used to develop
diagnostics for the assessment of renal function. This article reviews both glomerular and tubular kidney
stress tests and their respective diagnostic utility.
KI Reports (2016) 1, 57–63; http://dx.doi.org/10.1016/j.ekir.2016.04.005
KEYWORDS: acute kidney injury; chronic kidney disease; furosemide stress test; glomerular filtration; kidney stress
test; maximum GFR
ª 2016 International Society of Nephrology. Published by Elsevier Inc. This is an open access article under the
CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

A s part of human evolutionary development, many


human organ systems have innate mechanisms to
adapt to increased “work demand” or stress. At rest,
may diverge. An assessment of both glomerular and
tubular function may be more informative than just 1
of these domains. Glomerular reserve testing has been
organ systems operate at baseline capacity, and this well established but is used infrequently in routine
capacity can be increased to a certain maximum ca- clinical care. Tubular function diagnostic testing is
pacity. A familiar example of this concept is cardiac relatively new and in its clinical “infancy.” However,
function. In a healthy person at rest, cardiac output is tubular assessment appears to hold significant promise
approximately 5.0 liters/min. However, when a healthy for the assessment of both chronic and acute kidney
person exercises, the cardiac output can double or even disease.
triple. Similarly, the kidney has reserve capacity of its
multiple physiological functions (Figure 1). The ability Renal Functional Reserve–Glomerular
to test the reserve of an organ system is often an Because of the common use of estimated glomerular
excellent diagnostic tool to uncover subclinical disease filtration rate (GFR) equations, there is a tendency for
(e.g., treadmill test). Similarly, stress testing of the non-nephrologists to think that the GFR is a constant.
kidney appears to generate insights into the presence In fact, the actual GFR changes throughout the day,
or absence of kidney disease and parenchymal loss due particularly after meals, based on physiological needs.1
to injury and potentially fibrosis. The 2 main domains One of the kidney’s primary roles is to effectively
of kidney stress testing are glomerular and tubular. In a remove nitrogenous waste, and as a consequence, the
healthy kidney, these 2 components of the nephron consumption and metabolism of protein results in an
work in concert. However, when the kidney is diseased increase in GFR.2 GFR can also be increased through
or injured, the glomerular and tubular function may be other mechanisms that work along the protein meta-
affected equally, or their form and functional capacity bolic pathway. For instance, an i.v. infusion of amino
acids will result in an increase in GFR.3 This increase in
GFR over baseline GFR is known as renal functional
reserve–glomerular (RFR-G).4 Protein ingestion,
Correspondence: Lakhmir S. Chawla, Veterans Affairs Medical particularly red meat, is a potent stimulant for
Center, 50 Irving Street, Washington, DC 20310, USA. E-mail:
minkchawla@gmail.com increasing GFR, and the teleologic explanation is likely
Received 8 March 2016; revised 25 April 2016; accepted 26 April related to an adaptive response to increased protein in
2016; published online 5 May 2016 the diet.5
Kidney International Reports (2016) 1, 57–63 57
REVIEW LS Chawla and C Ronco: Renal Stress Testing in the Assessment of Kidney Disease

seen in patients who donate a kidney; despite a halving


of their nephron mass, their serum creatinine and
calculated eGFR are “normal.”10 Therefore, when renal
disease becomes apparent due to an elevated serum
creatinine, this occurs only after the residual nephrons
can no longer compensate for the functional loss
(Figure 3).8

Renal Functional Reserve (RFR-G)


Normal subjects display a significant increase in GFR 1
or 2 hours after an acute protein load (1–1.2 g/kg) over
their baseline GFR. The difference between peak or
“maximum” GFR (maxGFR) and baseline GFR describes
the renal functional reserve of glomerular function
Figure 1. Comparison of stressors in the heart and kidney. C-R, (RFR-G). Fliser and colleagues11 compared the baseline
cardiorenal.
and maxGFR in young and elderly healthy subjects and
found that RFR was significantly lower in elderly than
Bosch and colleagues first described glomerular in young healthy individuals while virtually all base-
functional reserve (RFR-G) in 1983.4 In this seminal line GFR values of elderly were within the reference
paper, Bosch and colleagues demonstrated that the range. The renal reserve as assessed by RFR-G is a
consumption of protein, not carbohydrates or fat, re- measure of the kidney’s capacity to increase GFR by a
sults in a substantial increase in GFR in patients with combination of nephron recruitment and increases in
healthy kidneys. Multiple subsequent studies have renal blood flow coupled with hyperfiltration.12–15
confirmed these findings. The clinical implications of The stimulus to tap into this reserve capacity can
RFR-G will be reviewed. arise from adaptive physiological needs like pregnancy
Baseline (Unstressed) GFR or the presence of a solitary kidney. Utilization of RFR
GFR is normally utilized as a surrogate of kidney in non-disease states is best illustrated by pregnancy.
function in healthy subjects as well as in patients with In pregnancy, GFR significantly increases during each
kidney disease. Studies in healthy subjects under the trimester, such that there is a significant rise in bGFR
age of 50 have identified the average baseline normal from first to last trimester. Studies done on normal
values of GFR to be between 100 and 130 ml/min per pregnant women in each trimester have shown a pro-
1.73 m2.6 Evaluation of population-wide “normal” gressive increase of baseline GFR with a parallel
values is useful, but the concept of “normal” GFR in reduction of RFR due to its progressive utilization.13
the single individual is more nuanced. It is important to MaxGFR in normal pregnant women, however, does
recognize that a person’s GFR at any given point in not change. However, pathological states can also
time will vary in relation to the physiological demands initiate processes that increase GFR above the normal
of dietary and hemodynamic conditions. Baseline value baseline. Primary hyperfiltration in kidney disease has
for GFR (bGFR) also depends on age, sex, and body been shown in patients with diabetes mellitus, poly-
size, with considerable variation among healthy in- cystic kidney disease, secondary focal segmental
dividuals. Overall, the average daily GFR is remarkably
stable over years, although there is an age-related
decline in GFR physiologically by 0.8 ml/min per
1.73 m2 per year, after the age of 30 years.6,7
In general, serum creatinine tends to remain rela-
tively normal even in the presence of kidney damage,
until approximately 50% of nephrons are lost or
simply when bGFR approaches 60 ml/min per 1.73 m2
(Figure 2).8 For this reason serum creatinine cannot be
considered an accurate marker of renal function when
GFR is above 60 ml/min per 1.73 m2. Similarly, GFR
estimation (eGFR) by creatinine-derived equations (e.g.,
MDRD9) cannot be considered a sensitive index for
early detection of renal disease during the early phases Figure 2. Relationship between glomerular filtration rate (GFR) and
of parenchymal damage. A good example of this can be serum creatinine changes. RFR-G, renal functional reserve–glomerular.

58 Kidney International Reports (2016) 1, 57–63


LS Chawla and C Ronco: Renal Stress Testing in the Assessment of Kidney Disease REVIEW

overall increase in blood flow is the main mechanism


rather than a temporary hemodynamic perturbation in
the afferent and efferent tone and equilibrium
(C. Ronco and colleagues, unpublished data).
Stress testing with a protein load is the definitive
way to assess for the loss of RFR-G, but the significance
of renal reserve is not just a diagnostic consideration. It
is important to recognize that the loss of renal reserve
may also manifest as a loss in autoregulation capacity in
the kidney. This loss of autoregulation may increase
the vulnerability of those patients with CKD to volume
depletion and certain nephrotoxins (e.g., nonsteroidal
anti-inflammatory drugs). Population studies suggest
that increased creatinine variability, which could be
Figure 3. Variation in baseline glomerular filtration rate (GFR). due to the loss of autoregulation, predicts progression
to end-stage renal disease.20
glomerulosclerosis, sickle cell anemia, high-altitude
renal syndrome, obesity, hypertension, nephrotic Kidney Stress Testing of GFR in Clinical
syndromes, and glomerulonephritis.16 In physiological Practice and Future Research
states of diminished RFR, the observed hyperfiltration Assuming RFR-G represents the difference between
is likely due to recruitment of more nephron units, maximal filtration capacity of the kidney (maxGFR) and
whereas in pathological states, hyperfiltration is prob- the baseline GFR (bGFR), a protein load is the basis of a
ably due to an increase in single nephron filtration kidney stress test forcing the kidneys to utilize the
fraction. This, in part, is the basis of angiotensin II entire filtration capacity. This technique can be used to
blockade in chronic kidney disease (CKD), and is often “reveal” subclinical kidney disease. MaxGFR and bGFR
demonstrable by a drop in GFR when angiotensin- assessment with protein loading has been extensively
converting enzyme inhibitors are given to patients studied and can be used in the clinic to assess RFR-G in
with CKD. patients with kidney disease.4,8,13,14 Because dietary
A current limitation on the use of RFR-G assessment protein raises GFR, establishing bGFR is important
is that these assessments have not been conducted in when attempting to assess RFR-G; developing stan-
large cross-sectional cohorts, thus the population dardized protocols to accomplish this is an important
variability of the RFR response is not known. Several research recommendation. There is another approach
investigators have estimated RFR by measuring the that would allow single GFR assessment instead of
difference between protein-stimulated GFR and base- having to conduct a baseline and a stimulated stress
line GFR after a protein load.4 In a separate study,17 test. In this approach, the maxGFR would be assessed
Bosch and colleagues demonstrated the estimation of among healthy patients across a wide age range, ethnic
RFR by a short-term oral protein loading method. De range, and in both genders. Once these data were
Nicola and colleagues18 demonstrated that the estima- known, then normative values could be determined for
tion of RFR can be assessed by amino acid infusion. maxGFR. These data would be used for diagnostic
Numerous mechanisms have been hypothesized for purposes for patients who underwent a kidney stress
the increase in GFR after protein load. In their study, test to achieve maxGFR. Those patients who could not
Woods and colleagues19 hypothesized that protein achieve the appropriate maxGFR adjusted for age,
loading increases GFR because digested protein raises gender, and race could be referred for further work-up.
plasma amino acid levels, which are then filtered at the As part of a future research plan, the safety of
glomerulus, thereby stimulating proximal tubular ab- repeated protein loading in patients with CKD should
sorption. In addition, filtered amino acids change the also be assessed. Since protein is a stimulant for GFR,
sensitivity of macula densa sensing mechanisms, the effects of repeated protein loading in patients with
causing release of nitric oxide and prostaglandins CKD is unknown. The exposure of repeated high levels
locally resulting in vasodilation, increasing renal blood of protein in patients with CKD might be deleterious,
flow and GFR. In our own laboratory we analyzed the but might also “condition” the kidney as well and
response to acute protein loading, and we detected an stimulate restorative or protective effects—this concept
increase in GFR proportional to an increase in renal should be studied further.
blood flow with a constant of filtration fraction. This The idea of assessing renal reserve has been present
observation seems to support the hypothesis that an for decades, but is infrequently used in clinical
Kidney International Reports (2016) 1, 57–63 59
REVIEW LS Chawla and C Ronco: Renal Stress Testing in the Assessment of Kidney Disease

practice, whereas the cardiac stress test is used tubule’s capacity to secrete acid or sodium can be
routinely. Why is this the case? In our view, the simple assessed via acid or salt loading. The tubule’s concen-
reason is that cardiologists perceive that they can trating capacity can be assessed via water deprivation
intervene on patients with diminished cardiac reserve or exogenous administration of desmopressin (DDAVP).
(e.g., heart failure treatment), whereas the nephrology Among these different techniques, thus far the primary
community may not feel that an intervention is avail- methodology to assess tubular functional capacity in
able, and therefore may be unwilling to perform an patients with kidney disease has been via tubular
extra test. We hypothesize that patients with loss of secretion of either creatinine or an exogenous drug
renal reserve are at risk for CKD. Future trials should (e.g., furosemide).
assess whether early identification of diminished renal
reserve can reliably predict the risk of progression to Tubular Function Assessment in CKD
CKD. If this can be shown, early screening of renal The first studies of tubular functional capacity in patients
reserve may prompt early intervention and forestall the with CKD utilized the difference between creatinine
development of CKD. clearance and inulin clearance as an assessment of tubular
function. Herrera and colleagues24 developed an elegant
Tubular Function Assessment in Kidney Disease study to demonstrate the potential use of tubular secre-
The renal tubule portion of the nephron is tasked with tion. In this study, the investigators took the following 3
an enormous portfolio of responsibilities. Chief among cohorts of patients: normal, renal allograft donors (uni-
those chores are the handling of electrolytes, water, nephrectomized), and CKD. In these subjects, baseline
and amino acids, catabolism of various proteins, and creatinine clearance and inulin-based GFR were
the active secretion of endogenous and exogenous measured and then reassessed after a protein meal. They
acids. Tubular function assessment may be more found that both healthy patients and patients with CKD
informative than glomerular reserve in patients who were able to increase their inulin-measured GFR in
already have advanced kidney disease. When patients response to a protein meal; as expected, healthy patients
do not have obvious kidney disease, the loss of could increase their GFR after stimulation much more
glomerular reserve (RFR-G) can be an indicator of loss than CKD patients. Similarly, healthy patients were able
of nephron mass.8 However, once a patient has kidney to increase their tubular secretion of creatinine (TScr),
injury or disease, glomerular reserve is already sub- but CKD patients were unable to increase their TScr.
stantially reduced and therefore is less informative. When all 3 groups of patients were compared, uni-
In patients with decreased GFR, tubular function nephrectomized patients were able to increase their
appears to be more variable. One reason for this TScr (but to a lesser degree than normal healthy subjects),
observation may be due to renal fibrosis. During the while CKD patients were unable to increase their TScr.
assessment of kidney disease by tissue biopsy, the These data are consistent with previous studies that show
level of interstitial fibrosis is one of the strongest pre- that patients with CKD maintain some glomerular renal
dictors of renal survival.21,22 Interstitial fibrosis can reserve at all levels of baseline GFR.8 In addition, this
represent scarred tubules that are fibrosed, or the study demonstrated that CKD patients likely operate at
secretion of matrix that fills in between the nephrons, near their maximum TScr, and thus are less able to in-
or both of these. However, because CKD is generally crease their TScr when challenged with a protein meal.
marked by a reduction of kidney size, this makes the In a second trial, this same group of investigators
possibility of “extra” matrix an unlikely sole expla- assessed TScr by infusing i.v. creatinine into normal
nation for fibrosis (an exception to this would be subjects and kidney transplant recipients.25 They found
multiple myeloma). In most forms of kidney disease, that creatinine infusion did not increase GFR, and that
the kidneys shrink and become more echogenic over an infusion of i.v. creatinine resulted in an increased
time. Based on this observation, we believe that it is TScr in healthy patients, but not in kidney transplant
more likely that diseased tubules are replaced by ma- recipients. Thus, a tubular functional assessment with a
trix and fibrosis. In order to test the notion that tubular challenge of i.v. creatinine had the capacity to reveal the
function may identify patients who are at increased subjects with decreased nephron mass who otherwise
risk for worse outcomes, various studies in patients had normal serum creatinine levels.
with both acute and chronic kidney disease have been In aggregate, preliminary studies suggest that
conducted to determine the utility of tubular secretion tubular stress tests that measure the secretory capacity
capacity to predict outcomes.23,24 of the renal tubule are informative and predictive of
Different aspects of tubular function can be inter- outcomes.23–26 However, it should be noted that
rogated in various ways depending on what feature of tubular stress tests remain research tools and have not
tubular function is being assessed. For instance, the yet been deployed into the clinic for CKD.
60 Kidney International Reports (2016) 1, 57–63
LS Chawla and C Ronco: Renal Stress Testing in the Assessment of Kidney Disease REVIEW

Tubular Assessment in Acute Kidney Injury test to be safe and valid, and any volume losses induced
The aforementioned studies in CKD used creatinine by the diuresis should be replaced.
secretion to assess tubular functional assessment. In A version of the FST has also been analyzed in patients
patients with acute kidney injury (AKI), many factors, with advanced-stage AKI requiring renal replacement
including lack of steady state, increased catabolism, therapy to determine whether a standardized furosemide
and concurrent medications that interfere with creati- challenge can predict renal recovery. van der Voort and
nine secretion, preclude the use of TScr as a reliable colleagues reported that a standardized 4-hour infusion
measure of tubular secretion. One approach uses i.v. of furosemide was also an excellent predictor of renal
furosemide to assess tubular function. Furosemide, a recovery.26,33 This analysis was a post hocassessment of a
loop diuretic, has pharmacokinetic properties that randomized clinical trial, which compared a 4-hour
make it an appealing functional tool. In contrast to infusion of furosemide to placebo as an intervention to
other drugs cleared by the kidney, furosemide is not promote renal recovery in patients who are on contin-
effectively filtered by the glomerulus. As an organic uous renal replacement therapy. In this post hocanalysis,
acid, furosemide is tightly bound to albumin and gains the authors assessed the intervention arm of the trial (i.e.,
access to the tubular lumen by active secretion via the the patients randomized to furosemide) and found that
human organic anion transporter system in the prox- the mean urine output was much higher in patients
imal convoluted tubule.27,28 Once in the tubular lumen, destined to recover (654 ml vs. 48 ml, P ¼ 0.007) and had
furosemide blocks luminal cation–chloride cotransport a diagnostic performance receiver operating character-
throughout the thick ascending limb of Henle, thereby istic area under the curve of 0.84. These 2 studies
preventing sodium reabsorption and resulting in demonstrate that the urine output response to furose-
natriuresis and increased urine flow.29–31 Based on mide is informative about renal tubular function
these properties, furosemide-induced increases in urine throughout the phases of AKI (progression and recov-
output represent a methodology to assess the integrity ery). Another advantage of the FST is that it does not just
of the renal tubular function in the setting of AKI. This measure the tubule’s secretion capacity, but is actually
methodology was developed by Chawla and col- an assessment of integrated renal function34 (Figure 4). In
leagues23 and is referred to as the furosemide stress test order for furosemide to increase urine output, furose-
(FST). mide must be actively secreted into the proximal lumen,
The FST has been prospectively assessed in a single and the thick ascending limb, luminal patency, and
cohort study of critically ill patients with AKI and was collecting duct function must all be intact.35 Because the
found to have good diagnostic performance. In that FST requires an intact nephron for full function, the FST
study, Chawla and colleagues23 administered a stan- does not readily identify the location of the defect in
dard dose of i.v. furosemide (1.0–1.5 mg/kg) to criti- cases in which the FST response is poor.
cally ill patients with Kidney Disease: Improving The aforementioned studies of FST are of modest size
Global Outcomes (KDIGO) stage I or stage II AKI and and are currently undergoing larger-scale validation
then assessed the urine output response. This study (NCT 01275729). However, the FST is based on the
showed that the 2-hour urine output response to a bedside practice of many clinicians, which involves
furosemide challenge was able to predict progression to challenging patients with a loop diuretic and assessing
KDIGO stage III within 14 days with a receiver oper- the clinical response. The FST, as currently devised, is
ating characteristic area under the curve of 0.87 simply a framework around this common bedside
(SE, 0.05). At a cutoff of 200 cm3 at 2 hours, the practice. The FST is also being assessed by the 0 by 25
sensitivity and specificity of the FST were 87.1% and
84.1%, respectively.23 In a follow-up study of the same
cohort, the same research group showed that FST
performed better than known AKI biomarkers.
Importantly, the follow-up study demonstrated that
the FST performance improves when utilized in pa-
tients with increased levels of AKI biomarkers.32 These
data suggest that the combination of AKI biomarkers
with tubular functional assessment is informative, and
can be used at the bedside to assist clinicians in
assessing the severity of AKI. It remains unclear
whether the FST reveals the severity of AKI, or the loss
of tubular functional capacity. An important caveat to Figure 4. Furosemide urinary response tests tubular integrity. TAL,
the FST is that the subject must be euvolemic for the thick ascending limb.

Kidney International Reports (2016) 1, 57–63 61


REVIEW LS Chawla and C Ronco: Renal Stress Testing in the Assessment of Kidney Disease

initiative spearheaded by the International Society of 7. Davies DF, Shock NW. Age changes in glomerular filtration
Nephrology. In an austere medical environment, simple rate, effective renal plasma flow, and tubular excretory ca-
pacity in adult males. J Clin Invest. 1950;29:496–507.
diagnostic tools like serum urea and creatinine are not
readily available. Thus, the use of FST in euvolemic 8. Barai S, Gambhir S, Prasad N, et al. Functional renal reserve
capacity in different stages of chronic kidney disease.
patients with oliguria may allow a thoughtful way to Nephrology. 2010;15:350–353.
triage patients who may need more advanced care.
9. Levey AS, Bosch JP, Lewis JB, et al. A more accurate method
Because furosemide is inexpensive and available to estimate glomerular filtration rate from serum creatinine: a
worldwide, this physiological assessment may allow for new prediction equation. Modification of Diet in Renal Dis-
broader use of this diagnostic approach. ease Study Group. Ann Intern Med. 1999;130:461–470.
10. Tsuda A, Ishimura E, Uedono H, et al. Comparison of the
Summary estimated glomerular filtration rate (eGFR) in diabetic pa-
Kidney stress testing can be accomplished by assessing tients, non-diabetic patients and living kidney donors. Kidney
Blood Press Res. 2016;41:40–47.
glomerular and tubular domains. These assessments are
11. Fliser D, Zeier M, Nowack R, Ritz E. Renal functional reserve in
safe and relatively inexpensive and can be done at the
healthy elderly subjects. J Am Soc Nephrol. 1993;3:1371–1377.
bedside or in the clinic. Importantly, these assessments
12. Bosch JP, Lew S, Glabman S, Lauer A. Renal hemodynamic
have been shown to be informative in both acute and changes in humans. Response to protein loading in normal
chronic kidney disease. However, neither of these and diseased kidneys. Am J Med. 1986;81:809–815.
stress tests is currently used routinely at the bedside or 13. Ronco C, Brendolan A, Bragantini L, et al. Renal functional
the clinic. Assessment of RFR-G can and, in the opinion reserve in pregnancy. Nephrol Dial Transplant. 1988;3:157–161.
of these authors, should be used to reveal the loss of 14. Pecly IM, Genelhu V, Francischetti EA. Renal functional
RFR in patients at risk for kidney disease. Tubular reserve in obesity hypertension. Int J Clin Pract. 2006;60:
functional testing has been less developed; early 1198–1203.
studies demonstrate good diagnostic performance, but 15. Zitta S, Stoschitzky K, Zweiker R, et al. Dynamic renal function
large validation studies are still needed. Because testing by compartmental analysis: assessment of renal
functional reserve in essential hypertension. Nephrol Dial
tubular testing may have the capacity to assess multiple
Transplant. 2000;15:1162–1169.
anatomic domains of the nephron, we believe that
16. Cachat F, Combescure C, Cauderay M, et al. A systematic
noninvasive kidney stress testing may allow clinicians
review of glomerular hyperfiltration assessment and defini-
to phenotype, prognosticate, and better follow patients tion in the medical literature. Clin J Am Soc Nephrol. 2015;10:
with kidney disease. Further research into the appro- 382–389.
priate use of these diagnostic techniques is warranted. 17. Bosch JP, Lauer A, Glabman S. Short-term protein loading in
assessment of patients with renal disease. Am J Med.
DISCLOSURE 1984;77:873–879.

LSC has received consulting fees from Astute Medical. The 18. De Nicola L, Blantz RC, Gabbai FB. Renal functional reserve in
treated and untreated hypertensive rats. Kidney Int. 1991;40:
other author declared no competing interests.
406–412.
19. Woods LL. Mechanisms of renal hemodynamic regulation in
REFERENCES response to protein feeding. Kidney Int. 1993;44:659–675.
1. Koopman MG, Koomen GC, Krediet RT, et al. Circadian 20. O’Hare AM, Batten A, Burrows NR, et al. Trajectories of kid-
rhythm of glomerular filtration rate in normal individuals. Clin ney function decline in the 2 years before initiation of long-
Sci. 1989;77:105–111. term dialysis. Am J Kidney Dis. 2012;59:513–522.
2. Brenner BM, Meyer TW, Hostetter TH. Dietary protein intake and 21. Bohle A, Grund KE, Mackensen S, Tolon M. Correlations
the progressive nature of kidney disease: the role of between renal interstitium and level of serum creatinine.
hemodynamically mediated glomerular injury in the pathogen- Morphometric investigations of biopsies in perimembranous
esis of progressive glomerular sclerosis in aging, renal ablation, glomerulonephritis. Virchows Arch A Pathol Anat Histol.
and intrinsic renal disease. N Engl J Med. 1982;307:652–659. 1977;373:15–22.
3. Graf H, Stummvoll HK, Luger A, Prager R. Effect of amino acid 22. Mackensen-Haen S, Bader R, Grund KE, Bohle A. Correlations
infusion on glomerular filtration rate. N Engl J Med. 1983;308: between renal cortical interstitial fibrosis, atrophy of the
159–160. proximal tubules and impairment of the glomerular filtration
4. Bosch JP, Saccaggi A, Lauer A, et al. Renal functional reserve rate. Clin Nephrol. 1981;15:167–171.
in humans. Effect of protein intake on glomerular filtration 23. Chawla LS, Davison DL, Brasha-Mitchell E, et al. Development
rate. Am J Med. 1983;75:943–950. and standardization of a furosemide stress test to predict the
5. Hostetter TH. Human renal response to meat meal. Am J severity of acute kidney injury. Crit Care. 2013;17:R207.
Physiol. 1986;250:F613–F618. 24. Herrera J, Rodriguez-Iturbe B. Stimulation of tubular secre-
6. Delanaye P, Schaeffner E, Ebert N, et al. Normal reference tion of creatinine in health and in conditions associated with
values for glomerular filtration rate: what do we really know? reduced nephron mass. Evidence for a tubular functional
Nephrol Dial Transplant. 2012;27:2664–2672. reserve. Nephrol Dial Transplant. 1998;13:623–629.

62 Kidney International Reports (2016) 1, 57–63


LS Chawla and C Ronco: Renal Stress Testing in the Assessment of Kidney Disease REVIEW

25. Rodriguez-Iturbe B, Herrera J, Marin C, Manalich R. Tubular 31. Brater DC, Anderson SA, Strowig S. Azosemide, a “loop”
stress test detects subclinical reduction in renal functioning diuretic, and furosemide. Clin Pharmacol Ther. 1979;25:
mass. Kidney Int. 2001;59:1094–1102. 435–439.
26. van der Voort PH, Boerma EC, Pickkers P. The furosemide 32. Koyner JL, Davison DL, Brasha-Mitchell E, et al. Furosemide
stress test to predict renal function after continuous renal stress test and biomarkers for the prediction of AKI severity.
replacement therapy. Crit Care. 2014;18:429. J Am Soc Nephrol. 2015;26:2023–2031.
27. Hasannejad H, Takeda M, Taki K, et al. Interactions of human 33. van der Voort PH, Boerma EC, Koopmans M, et al. Furose-
organic anion transporters with diuretics. J Pharmacol Exp mide does not improve renal recovery after hemofiltration for
Ther. 2004;308:1021–1029. acute renal failure in critically ill patients: a double blind
28. Bowman RH. Renal secretion of [35-S]furosemide and randomized controlled trial. Crit Care Med. 2009;37:533–538.
depression by albumin binding. Am J Physiol. 1975;229: 34. Powell TC, Warnock DG. The furosemide stress test and
93–98. predicting AKI outcomes. J Am Soc Nephrol. 2015;26:
29. Burg M, Stoner L, Cardinal J, Green N. Furosemide effect on 1762–1764.
isolated perfused tubules. Am J Physiol. 1973;225:119–124. 35. Berger BE, Warnock DG. Mechanisms of action and clinical
30. Dirks JH, Seely JF. Effect of saline infusions and furosemide uses of diuretics. In: Brenner BM, eds. The Kidney. 3rd ed.
on the dog distal nephron. Am J Physiol. 1970;219:114–121. Philadelphia: W.B. Saunders; 1986:433–456.

Kidney International Reports (2016) 1, 57–63 63


ARTIGO 9

GASOMETRIA ARTERIAL
[Downloaded free from http://www.ijccm.org on Tuesday, May 19, 2015, IP: 78.130.192.104]

Review Article

Interpretation of arterial blood gas


Pramod Sood, Gunchan Paul, Sandeep Puri1
Abstract

Disorders of acid–base balance can lead to severe complications in many disease states, and occasionally the abnormality
may be so severe as to become a life-threatening risk factor. The process of analysis and monitoring of arterial blood
gas (ABG) is an essential part of diagnosing and managing the oxygenation status and acid–base balance of the high-risk
patients, as well as in the care of critically ill patients in the Intensive Care Unit. Since both areas manifest sudden and
life-threatening changes in all the systems concerned, a thorough understanding of acid–base balance is mandatory for
any physician, and the anesthesiologist is no exception. However, the understanding of ABGs and their interpretation can
sometimes be very confusing and also an arduous task. Many methods do exist in literature to guide the interpretation
of the ABGs. The discussion in this article does not include all those methods, such as analysis of base excess or
Stewart’s strong ion difference, but a logical and systematic approach is presented to enable us to make a much easier
interpretation through them. The proper application of the concepts of acid–base balance will help the healthcare
provider not only to follow the progress of a patient, but also to evaluate the effectiveness of care being provided.

Keywords: Arterial blood gas interpretation, ABG analysis, rules for rapid ABG analysis, Anion gap,
Approach to mixed disorders

DOI: 10.4103/0972-5229.68215

Introduction concept of SID was introduced, which is defined as the


absolute difference between completely dissociated
Arterial blood gas (ABG) analysis is an essential part of
anions and cations. According to the principle of
diagnosing and managing a patient’s oxygenation status
electrical neutrality, this difference is balanced by the
and acid–base balance. The usefulness of this diagnostic
weak acids and CO2. The SID is defined in terms of weak
tool is dependent on being able to correctly interpret
acids and CO2 subsequently has been re-designated as
the results. Disorders of acid–base balance can create
effective SID (SIDe) which is identical to “buffer base.”
complications in many disease states, and occasionally
Similarly, Stewart’s original term for total weak acid
the abnormality may be so severe so as to become a
concentration (ATOT) is now defined as the dissociated
life-threatening risk factor. A thorough understanding
(A-) plus undissociated (AH) weak acid forms. This
of acid–base balance is mandatory for any physician,
is familiarly known as anion gap (AG), when normal
and intensivist, and the anesthesiologist is no exception.
concentration is actually caused by A-. Thus all the three
methods yield virtually identical results when they
The three widely used approaches to acid–base
are used to quantify acid–base status of a given blood
physiology are the HCO3- (in the context of pCO2),
sample.[1]
standard base excess (SBE), and strong ion difference
(SID). It has been more than 20 years since the Stewart’s
Why is it Necessary to Order an ABG
Analysis?
From:
Critical Care Division,1Department of Medicine, Dayanand Medical College The utilization of an ABG analysis becomes necessary
and Hospital, Ludhiana, Punjab, India in view of the following advantages:
Correspondence: • Aids in establishing diagnosis.
Dr. Sood Pramod, • Guides treatment plan.
Dayanand Medical College and Hospital, Ludhiana, Punjab, India
E-mail: soodpramod@yahoo.com • Aids in ventilator management.

Free full text available from www.ijccm.org

57
[Downloaded free from http://www.ijccm.org on Tuesday, May 19, 2015, IP: 78.130.192.104]

Indian J Crit Care Med April-June 2010 Vol 14 Issue 2

• Improvement in acid/base management; allows for than 30 minutes, use of glass syringes and ice slurry
optimal function of medications. is recommended.
• Acid/base status may alter electrolyte levels critical
to a patient's status. During preparation prior to sample transfer
• Visually inspect the sample for clots.
Accurate results for an ABG depend on the proper • Inadequate mixing of sample before analysis.
manner of collecting, handling, and analyzing the
specimen. Clinically important errors may occur at any of Insufficient mixing can cause coagulation of the sample.
the above steps, but ABG measurements are particularly It is recommended to mix the blood sample thoroughly
vulnerable to preanalytic errors. The most common by inverting the syringe 10 times and rolling it between
problems that are encountered include nonarterial the palms as shown in Figure 1. This prevents stacking
samples, air bubbles in the sample, inadequate or (such as coins or plates) of red blood cells.
excessive anticoagulant in the sample, and delayed
analysis of a noncooled sample.
During anticoagulation
Modern blood gas syringes and capillary tubes
Potential Preanalytical Errors are coated with various types of heparin to prevent
Preanalytical errors are caused at the following stages: coagulation in the sampler and inside the blood gas
analyzer:
During preparation prior to sampling • Liquid nonbalanced heparin
• Missing or wrong patient/sample identification; • Dry nonbalanced heparin
• Use of the incorrect type or amount of anticoagulant • Dry electrolyte-balanced heparin (Na+, K+, Ca2+)
• - dilution due to use of liquid heparin; • Dry Ca2+-balanced heparin
- insufficient amount of heparin;
- binding of electrolytes to heparin; Other anticoagulants, e.g., citrate and EDTA are both
• Inadequate stabilization of the respiratory condition slightly acidic which increase the risk of pH being falsely
of the patient; and lowered.
• Inadequate removal of flush solution in arterial lines
prior to blood collection. Liquid heparin
The use of liquid heparin as the anticoagulant causes
During sampling/handling a dilution of the sample, i.e., dilutes the plasma, but
• Mixture of venous and arterial blood during not the contents of the blood cells. As a consequence,
puncturing; parameters such as pCO2 and electrolytes are affected.
• Air bubbles in the sample. Any air bubble in the Only 0.05 mL of heparin is required to anticoagulate 1 mL
sample must be expelled as soon as possible after of blood. Dead space volume of a standard 5 mL syringe
withdrawing the sample and before mixing with with 1 inch 22 gauge needle is 0.2 mL; filling the syringe
heparin or before any cooling of the sample has been dead space with heparin provides sufficient volume to
done. An air bubble whose relative volume is up to
1% of the blood in the syringe is a potential source
of significant error and may seriously affect the pO2
value.
• Insufficient mixing with heparin.

During storage/transport
• Incorrect storage
• Hemolysis of blood cells

General Storage Recommendation


• Do not cool the sample.[2]
• Analyze within 30 min. For samples with high paO2,
e.g., shunt or with high leukocyte or platelet count
also analyze within 5 min. Figure 1: Correct method of mixing of the arterial sample with the
• When analysis is expected to be delayed for more anticoagulant in two dimensions to prevent stacking of red blood cells.

58
[Downloaded free from http://www.ijccm.org on Tuesday, May 19, 2015, IP: 78.130.192.104]

Indian J Crit Care Med April-June 2010 Vol 14 Issue 2

anticoagulate a 4-mL blood sample. If smaller sample Table 1: pH value and corresponding H+ ion concentration
volumes are obtained or more liquid heparin is left in pH H+ pH H+
the syringe, then the dilution effect will be even greater. 6.70 200 7.40 40
The dilution effect also depends on the hematocrit value. 6.75 178 7.45 35
Plasma electrolytes decrease linearly with the dilution of 6.80 158 7.50 32
the plasma along with pCO2, cGlucose, and ctHb values. 6.85 141 7.55 28
pH and pO2 values are relatively unaffected by dilution. 6.90 126 7.60 25
paO2 is only as little as 2% of the O2 physically dissolved 6.95 112 7.65 22
in the plasma, and so the oximetry parameters given in 7.00 100 7.70 20
7.05 89 7.75 18
fractions (or %) will remain unaffected.[3]
7.10 79 7.80 16
7.15 71 7.85 14
Syringes for blood gas analysis can have a wide range 7.20 63 7.90 13
of heparin amounts.[4] The units are typically given as IU/ 7.25 56 7.95 11
mL (international units of heparin per milliliter) blood 7.30 50 8.00 10
drawn into the syringe. In order to obtain a sufficient final 7.35 45
concentration of heparin in the sample, blood volume
recommended on the syringe must be drawn. Example:
Obtain a relevant clinical history
a syringe stated to contain 50 IU/mL when filled with
While making an interpretation of an ABG, never
1.5 mL of blood means that the syringe contains a total
comment on the ABG without obtaining a relevant
75 IU of dry heparin. If the user draws 2 mL of blood,
clinical history of the patient, which gives a clue to the
then the resulting heparin concentration will be too low
etiology of the given acid–base disorder. For example,
and the sample may coagulate. If the user draws only
a patient with a history of hypotension, renal failure,
1 mL, then the resulting heparin concentration will be
uncontrolled diabetic status, of treatment with drugs
higher than that aimed for, which may lead to producing
such as metformin is likely to have metabolic acidosis;
falsely low electrolyte results.
a patient, with a history of diuretic use, bicarbonate
administration, high-nasogastric aspirate, and vomiting,
Heparin binds to positive ions such as Ca2+, K+, and
is likely to have metabolic alkalosis. Respiratory acidosis
Na+. Electrolytes bound to heparin cannot be measured
would occur in COPD, muscular weakness, postoperative
by ion-selective electrodes, and the final effect will be
cases, and opioid overdose, and respiratory alkalosis is
measurement of falsely low values. The binding effect
likely to occur in sepsis, hepatic coma, and pregnancy.
and the resulting inaccuracy of results are especially
significant for corrected Ca2+. The use of electrolyte-
balanced heparin significantly reduces the binding effect Look at the oxygenation status of the patient
and the resulting inaccuracy.[5] The oxygenation status of the patient is judged by the
paO2; however, never comment on the oxygenation status
The following steps for rapid interpretation of ABG without knowing the corresponding FiO2. Calculate the
are recommended: expected paO2 (generally five times the FiO2).[6]

Based on the expected paO2 classify as mild, moderate,


Check for the consistency of ABG
and severe hypoxia.
While making an interpretation of an ABG always
check for the consistency of the report by the modified
Henderson equation. Ventilatory status
Look at paCO2.
[H ][HCO ] = 24
+_
3

PaCO2 Acid–base status


The hydrogen ion is calculated by subtracting the two Identify the primary disorder by looking at the pH
digits after the decimal point of pH from 80, e.g., if the
pH is 7.23 then pH > 7.40—Alkalemia: < 7.40—Acidemia
[H+] = 80 - 23 = 57
or Then look at paCO2 which is a respiratory acid, whether
[H+] = 10(9-pH) it is increased, i.e., >40 (acidosis) or decreased <40
(alkalosis) and if this explains the change of pH, then it
The hydrogen can be calculated from Table 1. is respiratory disorder; otherwise, see the trend of change

59
[Downloaded free from http://www.ijccm.org on Tuesday, May 19, 2015, IP: 78.130.192.104]

Indian J Crit Care Med April-June 2010 Vol 14 Issue 2

of HCO3- (whether increased in alkalosis or decreased ∆H +


in acidosis)—if it explains the change of pH, then it is a < 0.3 − Chronic
∆PaCO 2
metabolic disorder.

>0.8—acute
In a normal ABG 0.3–0.8—acute on chronic
• pH and paCO2 move in opposite directions.
• HCO3- and paCO2 move in same direction.
Alveolar Arterial Oxygen Gradient
It is calculated as follows:
1. When the pH and paCO2 change in the same direction
PAO2 = PiO2 – PaCO2
(which normally should not), the primary problem
is metabolic; when pH and paCO2 move in opposite R
directions and paCO2 is normal, then the primary
PiO2= FiO2 (PB-PH2O)
problem is respiratory.
2. Mixed Disorder—if HCO3- and paCO2 change in PAO2= FiO2 (PB-PH2O)- PaCO2
opposite direction (which they normally should R
not), then it is a mixed disorder: pH may be normal
with abnormal paCO2 or abnormal pH and normal where PAO2, alveolar partial pressure of oxygen; PiO2,
paCO2).[7] partial pressure of inspired oxygen; FiO2, fraction of
inspired oxygen; PB, barometric pressure (760 mmHg
If the trend of change in paCO2 and HCO3- is the same, at sea level); PH2O, water vapor pressure (47 mm Hg),
check the percent difference. The one, with greater PaCO2, partial pressure of carbon dioxide in blood; R,
% difference, between the two is the one that is the respiratory quotient assumed to be 0.8.
dominant disorder.
= FiO2 (760 - 47) - PaCO2
e.g.: pH = 7.25 HCO3 =16 -
paCO2 =60 0.8

Here, the pH is acidotic and both paCO2 and HCO3- Hypoxemic respiratory failure can be associated with
explain its acidosis: so look at the % difference normal (10–15 mmHg) or increased alveolar arterial
oxygen gradient. Figure 2 shows the alogrithim for
HCO3- % difference = (24 - 16)/24 = 0.33 approach in a patient with hypoxemic respiratory failure.
If this gradient is <20, then it indicates an extrapulmonary
paCO2 % difference = (60 - 40)/40 = 0.5 cause of respiratory failure.

Therefore, respiratory acidosis as the dominant Differentials of extrapulmonary causes of respiratory


disorder. failure:
(a) Central nervous system—Respiratory center
depression due to causes such as drug overdose,
Respiratory disorders primary alveolar hypoventilation, and myxedema.
After the primary disorder is established as respiratory, (b) Peripheral nervous system—Spinal cord diseases,
then the following points will help us to approach further Guillain-Barré syndrome, Amyotrophic lateral
with regard to the respiratory disorder).[8] sclerosis.
• Ratio of rate of change in H+ to change in paCO2 (c) Respiratory muscles—Hypophosphatemia, muscle
• Alveolar arterial oxygen gradient fatigue, myasthenia gravis, and polymyositis.
• Compensation (d) Chest wall diseases—Ankylosing spondylitis, flail
chest, thoracoplasty.
Ratio of rate of change in H+ to change in paCO2 (e) Pleural diseases—Restrictive pleuritis
The above ratio of rate of change in H+ to change in (f) Upper air way obstruction—Tracheal Stenosis, vocal
paCO2 helps in guiding us to conclude whether the cord tumor
respiratory disorder is acute, chronic, or acute on chronic.
As we have seen, the hydrogen can be calculated from Compensation
Table 1 and the change in H+ is calculated by subtracting Rules of compensation
the normal H+ from the calculated H+ ion.[9] (1) The compensatory response depends upon the

60
[Downloaded free from http://www.ijccm.org on Tuesday, May 19, 2015, IP: 78.130.192.104]

Indian J Crit Care Med April-June 2010 Vol 14 Issue 2

Figure 2: Flow diagram showing approach to hypoxemic respiratory failure

proper functioning of the organ system involved in mmHg decrease in paCO2 below 40.
the response (lungs or kidneys) and on the severity
of acid–base disturbance. For example, the likelihood Metabolic disorders
of complete compensation in chronic respiratory
In patients with metabolic acidosis, an excess of acid
acidosis is <15% when paCO2 exceeds 60 mmHg.
or loss of base is present. This causes the HCO3-:H2CO3
(2) Acute compensation occurs within 6–24 h and chronic
ratio and pH to fall while no change occurs in pCO2—
within 1–4 days. Respiratory compensation occurs
uncompensated metabolic acidosis.
faster than metabolic compensation.
(3) In clinical practice, it is rare to see complete
As a result of compensatory mechanisms, the lungs in
compensation. The maximum compensatory
the form of CO2 excrete H2CO3 and the kidneys retain
response in most cases is associated with only
HCO3-. pCO2 falls and HCO3-: H2CO3 ratio and pH rise
50–75% return of pH to normal. However, in chronic
toward normal even though concentrations of HCO3- and
respiratory alkalosis, the pH may actually completely
H2CO3 are less than normal. This is called compensated
return to normalcy in some cases.
metabolic acidosis and the expected paCO2 is calculated
as paCO2 = [1.5 × HCO3 + 8] ± 2.
Respiratory acidosis
Acute: [HCO3-] increase by 1 mEq/L for every 10 mmHg
Anion gap
increase in paCO2 above 40.
For more than 40 years, the AG theory has been used
by clinicians to exploit the concept of electroneutrality
Chronic: [HCO3-] increase by 3.5 mEq/L for every 10 and has evolved as a major tool for evaluating the acid–
mmHg increase in paCO2 above 40. base disorder. Anion gap is the difference between the
charges of plasma anions and cations, calculated from the
Respiratory alkalosis difference between the routinely measured concentration
Acute: [HCO3-] decrease by 2 mEq/L for every 10 of the serum cations (Na+ and K+) and anions (Cl- and
mmHg decrease in paCO2 below 40. HCO3-). Because electroneutrality must be maintained,
the difference reflects the unmeasured ions. Normally,
Chronic: [HCO3-] decrease by 5 mEq/L for every 10 this difference or the gap is filled by the weak acids (A-)

61
[Downloaded free from http://www.ijccm.org on Tuesday, May 19, 2015, IP: 78.130.192.104]

Indian J Crit Care Med April-June 2010 Vol 14 Issue 2

principally albumin, and to a lesser extent phosphates, • Helping to differentiate between causes of metabolic
sulfates, and lactates. acidosis: High AG versus normal AG metabolic
acidosis. In an inorganic metabolic acidosis (e.g., due
When the AG is greater than that produced by the to HCl infusion), the infused Cl- replaces HCO3-, and
albumin and phosphate, other anions (e.g., lactates the AG remains normal. In an organic acidosis, the
and ketones) must be present in higher than normal lost bicarbonate is replaced by the acid anion which
concentration. is not normally measured. This means that the AG
is increased.
Anion gap = (Na+ + K+) - [Cl- + HCO3-] • Providing assistance in assessing the biochemical
severity of the acidosis and follow the response to
Because of its low and narrow extracellular treatment.
concentration, K+ is often omitted from the calculation
The normal value ranges from 12 ± 4 when K + is Disorders that are associated with a low or negative
considered, and 8 ± 4 when K+ is omitted. Figure 3 shows serum AG are listed in Table 2.
the alogrithm for the approach to patients with normal
AG acidosis. Table 3 elaborates the species of the unaccounted
anions along with their sources of origin and diagnostic
The primary problem with AG is its reliance on the adjunts in case of high AG metabolic acidosis.
use of the normal range produced by the albumin and
to a lesser extent phosphate, the level of which may be In the patients with metabolic alkalosis, there is
grossly abnormal in critically ill patients. Because these an excess of base or a loss of acid which causes the
anions are not strong anions, their charges will be altered HCO3-:H2CO3 ratio and pH to rise, but with no change
by changes in pH.[10,11] occurring in pCO2, which is called uncompensated
metabolic alkalosis. However, the kidney has a large
Serum protein and phosphate capacity to excrete excess bicarbonate and so, for
Normal AG = 2{albumin(gm/L)} + 0.5 {phosphate sustaining the metabolic alkalosis, the elevated HCO3-
(mg/dL)} concentration must be maintained through an abnormal
Acid–base status renal retention of HCO3-.
In Acidemic state – Anion gap decreases by 1–3
In Alkalemic state – Anion gap increases by 2–5 Compensatory respiratory acidosis may be so marked
that pCO2 may rise higher than 55 mmHg. Expected paCO2
Major clinical uses of the anion gap is calculated as paCO2 = [0.7 × HCO3- + 21] ± 2 or 40 + [0.7
• For signaling, the presence of a metabolic acidosis ∆HCO3]. This is called compensated metabolic alkalosis.
and confirm other findings.
Most of the patients with metabolic alkalosis can be
+ + -
treated with chloride ions in the form of NaCl (saline
Urinary anion gap = (Urinary Na + Urinary K ) - Urinary Cl
responsive) rather than KCl (which is preferable). When
NaCl is given, Cl- ions are supplied, and so the blood
volume increases and the secretion of aldosterone in
Positive Negative excess decreases. Thus, excessive urinary loss of K+ and
renal extra renal
excessive reabsorption of HCO3- stops. When metabolic
(type I, II, IV Renal tubular acidosis)
alkalosis is due to the effects of excessive aldosterone or
other mineralocorticoids, the patient does not respond
Urinary pH to NaCl (saline resistant) and requires KCl.

Based on the urinary chloride, metabolic alkalosis is


divided into:
>6 <5.5
(Type I, renal tubular acidosis) Chloride responsive or extracellular volume depletion
(urinary chloride < 20)
• Vomiting
Hypokalemia Hyperkalemia
• Diuretic
(Type II renal tubular acidosis) (Type IV renal tubular A)
• Post hypercapnic
Figure 3: Approach to a patient with normal anion gap acidosis • Chronic diarrhea

62
[Downloaded free from http://www.ijccm.org on Tuesday, May 19, 2015, IP: 78.130.192.104]

Indian J Crit Care Med April-June 2010 Vol 14 Issue 2

Table 2: Disorders associated with low serum anion gap


Cause Comments
Laboratory error Most frequent cause of low anion gap
Hypoalbuminemia Second most common cause of low serum anion gap
Multiple myeloma Level of anion gap correlates with serum concentration of paraprotein
Halide intoxication Anion gap depends on serum halide concentration
(bromide, lithium, iodide) (low anion gap with lithium ≥4 mEq/L)
Hypercalcemia more likely in hypercalcemia associated with 10 hyperparathyroidism
Hypermagnesemia Theoretical cause but not documented in literature
Polymyxin B Anion gap depends on serum level; occurs with preparation with chloride
Underestimation of serum sodium Most frequent with hypernatremia or hypertriglyceridemia
Overestimation of serum chloride Rare with ion selective electrodes
Overestimation of serum bicarbonate Spurious ↑in serum HCO3 if cells not separated from sera

Table 3: Description of the species of unmeasured anions, source of origin, and diagnostic adjuncts in case of high anion gap
metabolic acidosis
Cause High serum anion gap
Comments
Species Origin Diagnostic adjuncts
Renal failure Phosphates, sulphates Protein metabolism BUN/creatinine
Ketocidosis Ketoacids Fatty acid metabolism Serum/urine ketones
Diabetic β Hydroxybutyrate
Alcoholic
Starvation Acetoacetate
Lactic acidosis Lactate Lactate levels
Exogenous poisoning Salicylate Salicylate Concomitant
Lactate Respiratory and metabolic alkalosis
ketoacids

Chloride resistant (urinary chloride > 20) evaluation of blood gas and acid–base disturbances in
• Severe potassium depletion the body, the following scheme is suggested:
• Mineralocorticoid excess—Primary
hypealdosteronism, Cushing's Syndrome, Ectopic 1. Look at pH – < 7.40 – Acidosis; > 7.40 – Alkalosis
ACTH
• Secondary hypereldosteronism—Renovascular 2. If pH indicates acidosis, then look at paCO2 and
disease, malignant hypertension, CHF, cirrhosis HCO3-

Aproach to mixed disorder 3. If paCO2 is ↑, then it is primary respiratory acidosis


Mixed metabolic disturbances (e.g., high AG from (a) To determine whether it is acute or chronic
diabetic ketoacidosis plus normal AG from diarrhea)
∆H+/∆paCO2 <0.3—chronic
can be identified using the relationship between AG
>0.8—acute
and HCO3-, which is called the gap–gap ratio. It is
0.3–0.8—acute on chronic
the ratio of change in anion gap (∆AG) to change in
(b) Calculate compensation by the respective methods
HCO3- (∆HCO3-). When hydrogen ions accumulate in
Acute: [HCO3-] ↑ by 1 mEq/L for every 10 mmHg
blood, the decrease in serum HCO3- is equivalent to the
increase in AG and the increase in AG excess/HCO3- ↑ in paCO2 above 40.
deficit ratio is unity, i.e., pure increase in AG metabolic Chronic: [HCO3-] ↑ by 3.5 mEq/L for every 10
acidosis. When a normal AG acidosis is present, the mmHg ↑ in paCO2 above 4
ratio approaches zero. When a mixed acidosis is present
(high AG + normal AG), the gap–gap ratio indicates the 4. If paCO2 ↓ and HCO3- is also ↓→ primary metabolic
relative contribution of each type to the acidosis. If it is acidosis
<1, then it suggests that there is a normal AG metabolic Calculate expected paCO2 as follows:
acidosis associated with it and if >2 it suggests that there paCO2 = [1.5 × HCO3 + 8] ± 2 metabolic acidosis only
is associated metabolic acidosis. paCO2 < expected paCO2 → concomitant respiratory
alkalosis.
Rules for rapid clinical interpretation of ABG paCO2 > expected paCO2 → concomitant respiratory
When required to make a proper approach towards the acidosis

63
[Downloaded free from http://www.ijccm.org on Tuesday, May 19, 2015, IP: 78.130.192.104]

Indian J Crit Care Med April-June 2010 Vol 14 Issue 2

5. If HCO3- is ↓, then AG should be examined. 13. If pH is normal ABG may be normal or mixed
disorder
6. If AG is unchanged → then it is hyperchloremic (a) ↑paCO2 and ↓HCO3- → respiratory and metabolic
metabolic acidosis. acidosis
(b) ↓paCO2 and↑ HCO3- → respiratory and metabolic
7. If AG is ↑ → then it is wide AG acidosis. alkalosis.
Calculate % difference (∆HCO 3- /HCO 3- and
8. Check gap–gap ratio ∆paCO 2 /paCO 2) to see which is dominant
∆AG/∆ HCO3- = 1, pure increased AG metabolic disorder.
acidosis
<1 normal anion gap metabolic acidosis References
>2 associated metabolic acidosis. 1. Kellum JA. Making Strong Ion Difference the "Euro" for Bedside Acid-
Base Analysis. Yearbook of Intensive Care and Emergency Medicine.
Spr Ber Heid Publ 2005;5:675.
9. If pH indicates alkalosis, then look at HCO3- and 2. Phillips B, Peretz DI. Blood Gas Pre-analytical considerations.
paCO2. Specimen collection, Calibration, and Controls (proposed guidelines).
In: National Committee for Clinical Laboratory Standards. NCCLS
publication. villanova PA, NCCLS; 1985.
10. If paCO 2 is ↓ → then it is primary respiratory 3. Börner U, Müller H, Höge R, Hempelmann G. The influence of
alkalosis. anticoagulation on acid-base status and blood-gas analysis. Acta
Anaesthesiol Scand 1984;28:277-9.
(a) Whether it is acute or chronic (with the same
4. Hutchison AS, Ralston SH, Dryburgh FJ, Small M, Fogelman I. Too
formula as above) much heparin: possible source of error in blood gas analysis. Br Med J
(b) Calculate compensation by the respective methods: 1983;287:1131-2.
Acute: [HCO3-]↓ by 2 mEq/L for every 10 mmHg 5. Toffaletti J, Ernst P, Hunt P, Abrams B. Dry electrolyte-balanced
heparinized syringes evaluated for determining ionized calcium and
↓ in paCO2 below 40. other electrolytes in whole blood. Clin Chem 1991;37:1730-3.
Chronic: [HCO 3 - ] ↓ by 5 mEq/L for every 6. Woolf CR. Letter: Arterial blood gas levels after oxygen therapy. Chest
10mmHg ↓ in paCO2 below 40. 1976;69:808-9.
7. Narins RG, Emmett M. Simple and mixed acid-base disorders: a
11. If paCO2 ↑ and HCO3- also ↑ → then it is primary practical approach. Medicine 1980;59:161-87.
metabolic alkalosis. 8. Marino PL. Arterial Blood Gas Interpretation. In; The ICU book, 2nd
Calculate the expected paCO2 edi. Lippincott, Williams and Wilkins Publishers;1998. P. 582-605.
9. Bartter TC, Abouzgheib WB, Pratter MR, Irwin RS. Respiratory
paCO2 = [0.7 × HCO3- + 21] ± 2 Or 40 + [0.7 ∆HCO3]
Failure- Part 1. In: Irwin and Rippe’s Intensive Care Medicine, 6th
→ metabolic alkalosis only edi. Lippincott, Williams and Wilkins Publishers; 2008. P. 485-9.
paCO2 < expected paCO2 → concomitant respiratory 10. Rao SM, Nagendranath V. Arterial Blood Gas Monitoring: Indian J
alkalosis. Anaesth 2002;46:289-97.
11. Chawla LS, Shih S, Davison D, Junker C, Seneff MG. Anion gap,
paCO2 > expected paCO2 → concomitant respiratory anion gap corrected for albumin, base deficit and unmeasured anions
acidosis in critically ill patients: implications on the assessment of metabolic
acidosis and the diagnosis of hyperlactatemia. BMC Emerg Med
2008;8:18.
12. Check urinary chloride
if urinary chloride < 20 → chloride responsive or ECV
depletion
Source of Support: Nil, Conflict of Interest: None declared.
if urinary chloride > 20→ chloride resistant

64
ARTIGO 10

TIPOS DE ACESSO VASCULAR


Vascular Access
for Hemodialysis
What is a vascular access? Two types of vascular access designed for

A
long-term use include the arteriovenous
vascular access is a hemodialysis
(AV) fistula and the AV graft. A third type
patient’s lifeline. A vascular access
of vascular access—the venous catheter—is
makes life-saving hemodialysis
for short-term use.
treatments possible. Hemodialysis is a
treatment for kidney failure that uses
a machine to send the patient’s blood What is an arteriovenous fistula?
through a filter, called a dialyzer, outside An AV fistula is a connection, made by a
the body. The access is a surgically vascular surgeon, of an artery to a vein.
created vein used to remove and return Arteries carry blood from the heart to the
blood during hemodialysis. The blood body, while veins carry blood from the
goes through a needle, a few ounces at a body back to the heart. Vascular surgeons
time. The blood then travels through a specialize in blood vessel surgery. The
tube that takes it to the dialyzer. Inside surgeon usually places an AV fistula in
the dialyzer, the blood flows through thin the forearm or upper arm. An AV fistula
fibers that filter out wastes and extra fluid. causes extra pressure and extra blood to
The machine returns the filtered blood flow into the vein, making it grow large
to the body through a different tube. A and strong. The larger vein provides easy,
vascular access lets large amounts of blood reliable access to blood vessels. Without
flow continuously during hemodialysis this kind of access, regular hemodialysis
treatments to filter as much blood as sessions would not be possible. Untreated
possible per treatment. About a pint of veins cannot withstand repeated needle
blood flows through the machine every insertions. They would collapse the way
minute. A vascular access should be in a straw collapses under strong suction.
place weeks or months before the first
hemodialysis treatment.

National Kidney and Urologic Diseases


Information Clearinghouse
Health care providers recommend an AV fistula over
the other types of access because it
■ provides good blood flow for dialysis To hemodialysis From hemodialysis
machine machine
■ lasts longer than other types of access
Arterial needle Venous needle
■ is less likely to get infected or cause blood clots
than other types of access Vein

Before AV fistula surgery, the surgeon may perform


a vessel mapping test. Vessel mapping uses Doppler
ultrasound to evaluate blood vessels that the surgeon
may use to make the AV fistula. Ultrasound uses a Fistula Artery
device, called a transducer, that bounces safe, painless
sound waves off organs to create an image of their
structure. A specially trained technician performs
AV fistula in forearm
the procedure in a health care provider’s office, an
outpatient center, or a hospital. A radiologist—a
doctor who specializes in medical imaging— At the start of a hemodialysis session, a health care
interprets the images. A patient does not need provider or the patient inserts two needles into the
anesthesia. A Doppler ultrasound shows how much vascular access. One needle carries blood from
and how quickly blood flows through arteries and the body to the dialyzer. The other carries filtered
veins so the surgeon can select the best blood vessels blood back to the body. To tell the needles apart,
to use. the needle that carries blood away from the body is
called the arterial needle. The needle that carries
A surgeon performs AV fistula surgery in an
blood back to the body is called the venous needle.
outpatient center or a hospital. The vascular access
Some patients prefer to insert their own needles into
procedure may require an overnight stay in the
the vascular access, which requires training to learn
hospital; however, many patients go home afterward.
how to prevent infection and protect the vascular
A health care provider uses local anesthesia to numb
access. No matter who inserts the needles, the
the area where the surgeon creates the AV fistula.
patient should know how to take care of the needle
An AV fistula frequently requires 2 to 3 months to insertion area to prevent infection.
develop, or mature, before the patient can use it for
If an AV fistula does not mature, an AV graft is the
hemodialysis. If an AV fistula fails to mature after
second choice for a long-lasting vascular access.
surgery, a surgeon must repeat the procedure.

2
What is an arteriovenous graft?
An AV graft is a looped, plastic tube that connects
Set Up the Vascular Access Well
an artery to a vein. A vascular surgeon performs before Starting Hemodialysis
AV graft surgery, much like AV fistula surgery, in an Patients should set up a vascular access well
outpatient center or a hospital. As with AV fistula before starting hemodialysis, as AV fistulas
surgery, the patient may need to stay overnight in and AV grafts both need time to mature before
the hospital, although many patients can go home they are ready for use. A health care provider
after the procedure. A health care provider uses can help schedule an appointment with a
local anesthesia to numb the area where the surgeon vascular surgeon well before the patient starts
creates the AV graft. hemodialysis, even if the patient is feeling fine.
Giving a vascular access time to mature can help
A patient can usually use an AV graft 2 to 3 weeks prevent problems with narrow veins, low blood
after the surgery. An AV graft is more likely than flow, and blood clots.
an AV fistula to have problems with infection and
clotting. Repeated blood clots can block the flow of Before the procedure, health care providers
blood through the graft. However, a well-cared-for should use the back of the patient’s hand for
graft can last several years. drawing blood to preserve the blood vessels in
the arm. A health care provider can teach the
patient simple exercises that help the blood
vessels grow larger for the surgeon’s use. The
same exercises help the AV fistula grow larger
Artery Vein
Looped graft after the procedure.

What is a venous catheter?


A venous catheter is a tube inserted into a vein in
Arterial needle the neck, chest, or leg near the groin, usually only
Venous needle for short-term hemodialysis. The tube splits in two
after the tube exits the body. The two tubes have
To From
hemodialysis hemodialysis caps designed to connect to the line that carries
machine machine blood to the dialyzer and the line that carries blood
from the dialyzer back to the body. A person must
AV graft in forearm close the clamps on each line when connecting and
disconnecting the catheter from the tubes.

3
If kidney disease has progressed quickly, a patient What problems could a vascular
may not have time for placement of an AV fistula or
AV graft before starting hemodialysis treatments. access cause?
All three types of vascular access—AV fistula,
A nephrologist—a doctor who specializes in kidney AV graft, and venous catheter—can cause problems
problems—or an interventional radiologist—a doctor that require further treatment or surgery. The most
who uses medical imaging equipment to perform common problems include access infection and low
operations—performs the venous catheter placement blood flow due to blood clotting in the access.
procedure in a hospital or an outpatient center. The
patient receives local anesthesia and sedation to stay Infection and low blood flow happen less frequently
calm and relaxed during the procedure. in properly formed AV fistulas than in AV grafts and
venous catheters. Still, having an AV fistula does not
Venous catheters are not ideal for long-term use. guarantee the access will be problem-free.
With a venous catheter, a patient may develop a
blood clot, an infection, or a scarred vein, causing AV grafts more often develop low blood flow, an
the vein to narrow. However, if a patient needs to indication of clotting or narrowing of the access. The
start hemodialysis right away, a venous catheter will AV graft may then require angioplasty, a procedure
work for several weeks or months until a surgeon can to widen the narrow part. Another option involves
perform a long-term access surgery and the AV fistula surgery on the AV graft to replace the narrow part.
or AV graft has time to mature. Venous catheters are the most likely to cause
If fistula or graft surgery is unsuccessful, then a infection and clotting problems. If these problems
patient will need a long-term venous catheter access. develop, medication may help. Antibiotics are
When a patient needs a venous catheter for more medications that fight bacteria that can cause
than 3 weeks, the surgeon will “tunnel” the catheter infection. Blood thinners such as warfarin keep
under the skin, rather than insert it directly into blood from clotting. If these treatments fail, a
the vein. A tunneled catheter is more comfortable nephrologist or an interventional radiologist will
and has fewer problems. Even tunneled catheters, need to replace the catheter.
however, may become infected.

Arterial line
Tunneled portion
of the catheter
Clamps

Arterial line to Venous line


hemodialysis
machine Caps

Venous line from


hemodialysis
machine

Tunneled venous catheter

4
How does a patient care for and protect Eating, Diet, and Nutrition
a vascular access? Researchers have not found that eating, diet, and
A patient can care for and protect a vascular access by nutrition play a role in causing or preventing
problems with a vascular access.
■ ensuring that the health care provider checks the
access for signs of infection or problems with Read more about eating well during hemodialysis
blood flow before each hemodialysis treatment, in Eat Right to Feel Right on Hemodialysis at
even if the patient is inserting the needles. www.kidney.niddk.nih.gov.

■ keeping the access clean at all times.


Points to Remember
■ using the access site only for dialysis. ■ A vascular access is a surgically created vein used
■ being careful not to bump or cut the access. to remove and return blood during hemodialysis.
■ checking the thrill in the access every day. The ■ An arteriovenous (AV) fistula is a connection,
thrill is the rhythmic vibration a person can feel made by a vascular surgeon, of an artery to a
over the vascular access. vein.
■ watching for and reporting signs of infection, ■ Health care providers recommend an AV fistula
including redness, tenderness, or pus. over the other types of access because it
■ not letting anyone put a blood pressure cuff on – provides good blood flow for dialysis
the access arm. – lasts longer than other types of access
■ not wearing jewelry or tight clothes over the – is less likely to get infected or cause blood
access site. clots than other types of access
■ not sleeping with the access arm under the head ■ An AV graft is a looped, plastic tube that
or body. connects an artery to a vein.
■ not lifting heavy objects or putting pressure on ■ A venous catheter is a tube inserted into a vein
the access arm. in the neck, chest, or leg near the groin, usually
only for short-term use.
■ Venous catheters are not ideal for long-term
hemodialysis. With a venous catheter, a patient
may develop a blood clot, an infection, or a
scarred vein, causing the vein to narrow.
■ All three types of vascular access can cause
problems that require further treatment or
surgery. The most common problems include
access infection and low blood flow due to blood
clotting in the access.

5
Hope through Research For More Information
The National Institute of Diabetes and Digestive People on hemodialysis can learn more about how
and Kidney Diseases (NIDDK) has many research to care for an access site from their health care
programs aimed at improving the treatment of provider. For a copy of the booklet Hemodialysis
kidney failure. One research program aimed at Access: What You Need to Know, contact
better understanding fistula maturation is the
National Kidney Foundation
Hemodialysis Fistula Maturation Study. This
30 East 33rd Street
multicenter study enrolled approximately 600 study
New York, NY 10016–5337
participants with newly placed fistulas to study the
Phone: 1–800–622–9010 or 212–889–2210
influence of vascular anatomy, vascular biology,
Fax: 212–689–9261
patient factors, and process of care on the fistulas’
Internet: www.kidney.org
usefulness for maintenance hemodialysis. The
results of this study will become available in 2015. For a copy of the booklet Understanding Your
Hemodialysis Access Options, contact
Clinical trials are research studies involving people.
Clinical trials look at safe and effective new ways to American Association of Kidney Patients
prevent, detect, or treat disease. Researchers also 2701 North Rocky Point Drive, Suite 150
use clinical trials to look at other aspects of care, Tampa, FL 33607
such as improving the quality of life for people with Phone: 1–800–749–2257 or 813–636–8100
chronic illnesses. To learn more about clinical trials, Fax: 813–636–8122
why they matter, and how to participate, visit the Email: info@aakp.org
NIH Clinical Research Trials and You website at Internet: www.aakp.org
www.nih.gov/health/clinicaltrials. For information
Life Options has developed an interactive
about current studies, visit www.ClinicalTrials.gov.
patient education website called Kidney School.
Module 8 of this program addresses vascular access
for hemodialysis. To view this module, go to
www.kidneyschool.org or contact
Life Options
c/o Medical Education Institute, Inc.
414 D’Onofrio Drive, Suite 200
Madison, WI 53719
Phone: 1–800–468–7777 or 608–833–8033
Fax: 608–833–8366
Internet: www.lifeoptions.org

6
The Centers for Medicare & Medicaid Services, About the Kidney Failure Series
the End-stage Renal Disease Networks, and the
The NIDDK Kidney Failure Series includes booklets
Institute for Healthcare Improvement launched
and fact sheets that can help the reader learn
the National Vascular Access Improvement
more about treatment methods for kidney failure,
Initiative in 2003. Materials from the Fistula
complications of dialysis, financial help for the
First Change Package are available by contacting
treatment of kidney failure, and eating right on
Fistula First Breakthrough Initiative hemodialysis. Free single printed copies of this series
IPRO ESRD Network Coordinating Center can be obtained by contacting the National Kidney
1979 Marcus Avenue, Suite 105 and Urologic Diseases Information Clearinghouse.
Lake Success, NY 11042
Phone: 516–209–5306 National Kidney Disease
Fax: 516–326–7805
Email: FFBI@ncc.esrd.net Education Program
Internet: www.fistulafirst.org 3 Kidney Information Way
Bethesda, MD 20892
Acknowledgments Phone: 1–866–4–KIDNEY (1–866–454–3639)
TTY: 1–866–569–1162
Publications produced by the Clearinghouse are Fax: 301–402–8182
carefully reviewed by both NIDDK scientists and Email: nkdep@info.niddk.nih.gov
outside experts. This publication was reviewed Internet: www.nkdep.nih.gov
by Michael Allon, M.D., University of Alabama
at Birmingham. The National Kidney Disease Education Program
(NKDEP) is an initiative of the National Institute
of Diabetes and Digestive and Kidney Diseases,
National Institutes of Health, U.S. Department of
Health and Human Services. The NKDEP aims to
raise awareness of the seriousness of kidney disease,
the importance of testing those at high risk, and the
availability of treatment to prevent or slow kidney
disease.

7
National Kidney and Urologic
Diseases Information Clearinghouse
3 Information Way
Bethesda, MD 20892–3580
Phone: 1–800–891–5390
TTY: 1–866–569–1162
Fax: 703–738–4929
Email: nkudic@info.niddk.nih.gov
Internet: www.kidney.niddk.nih.gov
The National Kidney and Urologic Diseases
Information Clearinghouse (NKUDIC) is a
service of the National Institute of Diabetes
and Digestive and Kidney Diseases (NIDDK).
The NIDDK is part of the National Institutes
of Health of the U.S. Department of Health
and Human Services. Established in 1987,
the Clearinghouse provides information
about diseases of the kidneys and urologic
system to people with kidney and urologic
disorders and to their families, health care
professionals, and the public. The NKUDIC
answers inquiries, develops and distributes
publications, and works closely with
professional and patient organizations and
Government agencies to coordinate resources
about kidney and urologic diseases.

This publication is not copyrighted. The Clearinghouse


encourages users of this publication to duplicate and
distribute as many copies as desired.
This publication is available at
www.kidney.niddk.nih.gov.

You may also find additional information about this


topic by visiting MedlinePlus at www.medlineplus.gov.
This publication may contain information about
medications and, when taken as prescribed,
the conditions they treat. When prepared, this
publication included the most current information
available. For updates or for questions about
any medications, contact the U.S. Food and Drug
Administration toll-free at 1–888–INFO–FDA
(1–888–463–6332) or visit www.fda.gov. Consult
your health care provider for more information.

NIH Publication No. 14–4554


May 2014

The NIDDK prints on recycled paper with bio-based ink.


ARTIGO 11

HEMODIÁLISE
DOI: 10.1111/sdi.12671

DIALYSIS EDUCATION:
ISSUES, INNOVATIONS AND IMPACT
Guest Editors: Bernard G. Jaar and Michael J. Choi

What the non-nephrologist needs to know about dialysis

Matt Foy1 | C. John Sperati2

1
Division of Nephrology, Department of
Medicine, Louisiana State University Health Abstract
Science Center, Baton Rouge, LA, USA The End-Stage Renal Disease (ESRD) program now serves approximately 675,000
2
Division of Nephrology, Department of
individuals in the United States at a cost of $26.1 billion to the Medicare system.
Medicine, Johns Hopkins University School
of Medicine, Baltimore, MD, USA Given the size of this population, healthcare providers from all disciplines will deliver
care to patients on dialysis. Mortality remains high among patients on chronic dialy-
Correspondence
C. John Sperati, MD, MHS, Johns Hopkins sis, with 42.3% surviving 5 years. As this is a vulnerable population, it is important
University School of Medicine, Baltimore,
in the care of ESRD patients that non-nephrologists have a working knowledge of
MD, USA.
Email: jsperati@jhmi.edu issues germane to dialysis. This review examines the physiology, mechanics, compli-
cations, and care delivery concerns of kidney dialysis modalities relevant to the non-
nephrologist. The majority of patients receive in-center hemodialysis thrice weekly,
with a small proportion on home-based therapies such as peritoneal dialysis or
home hemodialysis. Inpatients may undergo hemodialysis or peritoneal dialysis, and
in critically ill patients, continuous renal replacement therapies are utilized. Practical
aspects of each of these modalities are discussed.

Since the inception of the Medicare End-stage Renal Disease (ESRD) Perhaps one of the greatest concerns on how to safely conduct
program in 1972, the number of patients on dialysis has grown stea- HD centered on the permanent dialysis access. In 1960, Belding
dily. The prevalence of ESRD in the United States in 2014 was approxi- Scribner and Wayne Quinton devised an external device using
mately 675,000 patients, with close to 63% of these patients receiving Teflon tubing that could reliably be used for repeated HD access,
hemodialysis (HD). The annual cost for HD in the United States effectively becoming the first arteriovenous graft.4,6 Hemodialysis
approximates $87,500 per patient, with a total cost to Medicare of became more available through development of procedures to create
$26.1 billion.1 As healthcare providers of all disciplines will encounter arteriovenous fistulae, leading to a more permanent and longer last-
ESRD patients, the purpose of this study was to orient non-nephrolo- ing HD access.7 As HD use continued to grow in safety and applica-
gists to dialysis therapies and the issues relevant to their delivery. bility, its implementation faced challenges regarding who would
Though dialysis is considered a fairly commonplace procedure receive this precious resource.4,6 Access to this therapy changed
today, the beginnings of the therapy started a little more than immensely with passage of the Medicare ESRD Act.8,9
100 years ago. In 1912, John Jacob Abel experimented with a “vividif- Overall, survival trends on dialysis have improved over the past
fusion” apparatus in animals, with some degree of success.2 Clinical 15 years, although the adjusted 5-year survival for patients initiating
HD in humans began in 1943 under the work of Dr. Willem Kolff.3 all forms of dialysis in 2009 was 42.3%.1 The incidence of ESRD in
Though with limited success initially, the information gained from black patients is 3.1 times higher than Caucasians. Discrepancy in sur-
these early HD treatments helped pave the way for future exploration vival on HD exists based on ethnicity with older black patients with
of the artificial kidney. The first repeated hemodialysis treatments for ESRD having a lower mortality compared to white patients, with one
chronic kidney failure began at the University of Washington Hospital study noting a higher mortality in black patients with ESRD under the
in Seattle in 1960.4 Remarkably, this first patient lived another age of 50.10 In another study, an even more pronounced discrepancy
11 years on HD before dying of a myocardial infarction. Early imple- in survival in young black patients of lower socio-economic status was
mentation of peritoneal dialysis (PD) began in the early 1960s as well.5 demonstrated (65% higher risk of death compared to whites, 95% CI,

Seminars in Dialysis. 2018;1–10. wileyonlinelibrary.com/journal/sdi © 2018 Wiley Periodicals, Inc. | 1


2 | FOY AND SPERATI

1.38-1.97).11 Additionally, survival differences between men and negative pressure to the dialysis membrane, which subsequently
women exist, with men having a slightly higher adjusted mortality pulls off plasma water, or effluent, from the patient via ultrafiltra-
compared to women across all age groups (1.09, 95% CI 1.04-1.14).12 tion.17 It is worth noting that the fluid removed is not pure water,
but rather contains similar concentrations of solutes present within

1 | INTERMITTENT HEMODIALYSIS plasma that are capable of passing through the small pores of the
dialysis membrane. The amount removed is selected through volu-

1.1 | Indications metric controls on the dialysis machine. Selecting the amount of vol-
ume for removal is patient-dependent, and typically is determined by
Even though dialysis technology has evolved over the years, the indi- the patient’s estimated euvolemic weight (or “dry weight”), as well as
cations for initiating renal replacement therapy (RRT) have remained how much volume can be removed in a single dialysis session with-
largely constant (Table 1).13 On the most fundamental level, treatment out leading to complications such as cramping or hypotension
is meant to replicate essential functions of the kidney, including elimi- (Table 2). A patient’s interdialytic weight gain determines how much
nation of waste and clearance of small solutes from the bloodstream, volume to remove in an individual session. For example, if a patient’s
removal of excess body volume, and maintenance of blood pH.14 Addi- previous postdialysis session weight was 70 kg, and the current pre-
tionally, HD is useful in removing certain toxic ingestions. Timing of dialysis weight is 73 kg, then the ultrafiltration goal for the HD ses-
dialysis is patient-dependent, but data suggest that in the setting of sion will be 3 L since 1 L of water is 1 kg. Hypotension should
chronic kidney disease (CKD), there does not appear to be a benefit to prompt re-evaluation of UF goals, prescribed dry weight, use of pre-
starting dialysis early as compared to waiting for an indication for dial- dialysis antihypertensive medications, infection, cardiac dysfunction,
ysis initiation.15,16 The IDEAL study randomized adult patients with the presence of pericardial effusion, and adrenal insufficiency. In
CKD to start dialysis at an estimated glomerular filtration rate (eGFR) select patients, the use of midodrine, an a agonist, may be benefi-
of 10-14 mL/min/1.73 m2 or an eGFR of 5-7 mL/min/1.73 m2. Fol- cial.18 Albumin administered shortly before HD can sometimes facili-
lowing randomization, the delayed start group initiated dialysis tate ultrafiltration in the hospitalized, hypotensive, volume-
approximately 6 months later than the early start group, with no dif- overloaded patient, although its use is largely nonevidence-based.
ference in mortality or adverse events between the two.15

1.4 | Solute clearance


1.2 | Physiologic principles
Maintenance of small molecule homeostasis, such as potassium, cal-
As noted above, HD is designed to replace some of the normal
cium, and phosphorous balance, is an essential component of normal
physiology of the kidney. Blood is removed from the patient and fil-
renal function. With the loss of renal function, the kidney’s capacity
tered through a dialyzer consisting of over 10,000 small hollow
fibers constructed of a semipermeable membrane. These fibers have
small pores to facilitate exchange of substances between the blood T A B L E 2 Complications of renal replacement therapy
and dialysis fluid compartment that bathes the fibers and allows the Hemodialysis
necessary solute and fluid exchange to occur. Hypotension
Muscle cramping
Arrhythmia
1.3 | Volume removal Hemolysis
Air embolus
Retention of excess total body volume is one of the hallmarks of Thrombosis of filter and/or vascular access
advancing CKD. Unwanted body water is removed by applying a Infection of the bloodstream and/or vascular access
Vascular steal in patients with AVF/AVG
T A B L E 1 Indications for HD initiation Aneurysm or pseudoaneurysm of AVF/AVG
Heparin-induced thrombocytopenia
Indication Description Removal of water-soluble vitamins
Acidosis Sustained severe metabolic acidosis refractory to Continuous venovenous therapies
medical therapy. Typically pH <7.1
Air embolus
Electrolyte Sustained moderate to severe hyperkalemia despite Thrombosis of filter and/or vascular access
disturbance medical therapy and dietary modifications. Infection of the bloodstream and/or vascular access
Ingestion Dialyzable substances. Typically acute intoxication Thrombocytopenia
of salicylates, alcohols (ie, methanol and ethylene Hypophosphatemia
glycol), and lithium. Peritoneal dialysis
Volume overload Persistent excess total body volume impacting Peritonitis
pulmonary or cardiac function despite optimal Hyperglycemia
diuretic use. Bowel obstruction
Uremia Protein-calorie malnutrition, persistent nausea, Abdominal wall and inguinal hernias
confusion, uremic pericarditis, or uremic bleeding. Pleural effusion
FOY AND SPERATI | 3

to maintain normal levels of these substances is diminished. During pressures, pumps to inject heparin to prevent access clotting, and air
HD, exchange of these small molecules occurs through pores within detectors which can clamp the circuit and stop blood flow when air
the dialysis membrane by diffusion. Given the technical differences in the circuit is detected. Modern dialyzers are made of synthetic
between dialysis and ultrafiltration, patients can undergo solute clear- biocompatible material such as polysulfone; older more immunogenic
ance, volume removal, or both simultaneously. Though dialysis gener- filters of substituted cellulose are now rarely employed.22 Most dia-
ally results in removal of small molecules (such as potassium and lyzers in use today are considered high efficiency (large surface area)
phosphorous) from the patient, the movement is bidirectional and can and many are high flux (larger sized pores) to improve clearance dur-
include the movement of other small molecules (such as calcium and ing the dialysis treatment.23 Dialyzers are available in varying sizes
19
bicarbonate) from the dialysis fluid into the patient’s bloodstream. with different surface areas for exchange. While patient body sur-
Middle molecular weight and larger sized protein-bound mole- face area and treatment duration are considered in the dialysis pre-
cules in the bloodstream do not easily cross the dialysis membrane. scription, there tends to be little variation in the size of the dialysis
These substances are removed from the blood in a time-dependent membrane selected.
fashion which is augmented through the process of convection, also The water used to make the dialysate must undergo an extensive
known as solvent drag.20,21 In convective removal, larger molecules purification process with charcoal filtration, de-ionization, and
are pulled across the membrane as water is removed through ultrafil- reverse osmosis.24 Failure of the water purification system may
tration. It is important to recognize that dialysis does not selectively result in patient exposure to aluminum, fluoride, chloramine, copper,
clear the bloodstream of uremic toxins over more beneficial sub- and lead, which can lead to acute hemolysis or long-term toxicity
stances, such as water-soluble B vitamins. The concept of convec- such as dementia and osteomalacia (Table 2). Dialysate does not
tion is reviewed in Figure 1. have to be completely sterile in order to be used for a dialysis treat-
ment because the dialysis membrane acts as a barrier to bacteria. If
endotoxin is present within the dialysis fluid, pyrogenic reactions
1.5 | The dialyzer and dialysate
may occur.25
Blood is driven through the dialysis circuit using rotary pumps. Along Once the water meets standards for use in a dialysis treatment,
the course of the circuit are sensors to detect dialysis access adjustments are made to the dialysis bath to achieve the desired
concentrations of sodium, bicarbonate, potassium, and calcium.26
Typically, the concentrations in the dialysate fluid approximate nor-
mal serum concentrations, although adjustments are made based on

K+ patient needs. For example, if a patient has hyperkalemia of 6 mEq/


L, a 1 or 2 mEq/L potassium bath is selected to achieve a normal
postdialysis serum potassium concentration. It is worth noting that
K+
when patients present with profound electrolyte derangements (ie,
severe hyponatremia or severe hyperkalemia), careful consideration
K+
should be given to selection of dialysis fluid electrolytes to avoid
K+
overly rapid correction.
K+

1.6 | HD access and complications


K+ Given the technical components of HD, functional access to the
patient’s bloodstream is essential to long-term success. Three forms
of dialysis access predominate as follows: arteriovenous fistula
K+
(AVF), arteriovenous graft (AVG), and a dual lumen dialysis catheter.
K+ Though the least preferred access long-term, a dual lumen dialysis
catheter is often necessary in the setting of acute kidney injury
when no other access exists, or if an existing arteriovenous access is
not ready for use. Dialysis catheters may be tunneled or nontun-
neled.
Though the nontunneled dialysis catheters may be faster and
easier to place, they are associated with greater rates of infection
F I G U R E 1 Principles in hemodialysis. Diffusion: small molecules compared to tunneled catheters.27 Any long-term central vein dialy-
(K) move from the patient’s blood (PB) through pores in the dialysis
sis catheter is associated with increased risk of acute infection,
membrane (DM) into the dialysis fluid (DF). Ultrafiltration: water
molecules move via generation of negative pressure on the dialysate which in turn can lead to complications such as endocarditis. Surveil-
side of the DM. As the water moves across the DM, middle-sized lance data from US HD centers in 2014 demonstrated an 8.3-fold
molecules are pulled via convection (solvent drag) higher rate of blood stream infection with a central vein dialysis
4 | FOY AND SPERATI

catheter compared to an AVF.28 Dialysis catheter infections can be AVG may be associated with edema of the access arm and distal
challenging to eradicate without its removal. Consideration for steal phenomenon. Long-term patency rates are lower for AVGs
prompt removal of an infected dialysis catheter should be given to than AVFs, with most AVG having primary patency rates (ie, without
unstable patients with a suspected catheter infection.29 Additional intervention to maintain function) of under 18 months.35
concerns and complications from dialysis catheters include injury to HD is a remarkably well-tolerated procedure, although the previ-
surrounding anatomic structures during insertion (eg, arterial cannu- ously described hemodynamic, vascular, and dialysate complications
lation and pneumothorax), low blood flow rates with subsequent are of concern. In addition, equipment failures and inappropriate
inadequate clearance, and clotting. dialysis prescriptions may contribute to morbidity and potentially
The AVF is the preferred modality for long-term HD access, as it mortality (Table 2).36 The greatest concern with dialysis initiation is
has the greatest longevity and lowest complication rates. It is cre- dialysis disequilibrium syndrome (DES). DES results from rapid solute
ated surgically via ligation of a vein with anastomosis to an artery, shifts and pH changes from overly aggressive dialysis initiation,
preferably in the distal forearm (a radial-cephalic anastomosis), resulting in nausea, vomiting, headache, confusion, seizure, and pos-
though more proximal vessels may be selected (a brachial-cephalic sibly death.37 In addition, cerebral edema is more likely to occur in
30
anastomosis or brachial-basilic anastomosis). Generally, the non- patients undergoing HD within the first 24 hours of stroke or cere-
dominant arm is selected, and patients with CKD are advised to pro- bral hemorrhage. As such, the first 3 dialysis sessions are performed
tect this arm from venipuncture and blood pressure checks to at slower blood flows rate and for less time, and neurologically
preserve integrity of the blood vessels for future access use. It is unstable patients should be considered for continuous renal replace-
incumbent on all healthcare providers to preserve future HD access ment therapy (CRRT). Anaphylactic “Type A” dialyzer reactions have
sites in patients with CKD, especially in the acute care and hospital essentially been eliminated with the use of biocompatible mem-
setting. In general, peripherally inserted central catheters and subcla- branes and avoidance of ethylene oxide for sterilization, although
vian catheters should be avoided in patients with CKD.31 lesser “Type B” reactions such as back pain may occur on occasion
Because the AVF must undergo maturation prior to use, referral 30 or more minutes into treatment. Symptomatic management is
for access placement should occur in a timely fashion, though efforts sufficient for Type B reactions.
should be made to avoid unnecessary access placement. Generally Mechanical failure of the dialysis machine may result in air
speaking, ideal timing for referral occurs within 6-12 months of embolism or injury to the AV access; filter thrombosis may require
anticipated need for HD in a patient with CKD, as the AVF typically the use of anticoagulation. The prevalence of antibodies to platelet
requires 8-12 weeks to mature.31,32 The most common complication factor 4 is increased in HD patients, although uncertainty remains
from an AVF is failure for the access site to adequately develop, typ- regarding the risk for clinically relevant heparin-induced thrombocy-
ically due to proximal venous stenosis, insufficient arterial flow, or topenia.38 It is important to remember that heparin is often placed
30
collateral venous side branches. Additional complications associ- into the lumen of central venous catheters to maintain patency.
ated with the AVF may include edema in the access arm, aneurysmal
dilation of the vein, arterial steal phenomenon with distal ischemia,
1.7 | Dialysis dose adequacy
and bleeding from the access site. Unprovoked access bleeding (ie,
unrelated to recent cannulation or occurring after post-HD hemosta- The dialysis prescription should provide for adequate uremic solute
sis has been achieved) or breakdown of skin over the fistula war- removal. It is typically assessed by evaluating the kinetics of urea, a
rants prompt medical evaluation, as it may herald a more significant “marker” solute. It is quantified by two measurements in patients on
impending hemorrhage.33,34 Though not emergent, suspected clot- dialysis: Kt/V and the closely related urea reduction ratio (URR).39
ting of an AVF, demonstrated by a diminished or absent thrill and Kt/V is a unit-less measurement determined by (1) K, the clearance
bruit, pulsatile access, or prolonged postdecannulation bleeding, also of urea (mL/min) during a dialysis treatment; (2) t, the time (min) of
warrants prompt surgical evaluation to restore access flow and main- the treatment; and (3) V, the volume of distribution of urea (mL),
tain patency.30 which is approximately the total body water. URR is the fractional
If an AVF cannot be created, AVG placement is generally the reduction in the blood urea during a single dialysis treatment
next best choice for long-term hemodialysis access.31 The AVG typi- [URR = 1 ! (postdialysis BUN/predialysis BUN)]. Kt/V is the pre-
cally consists of polytetrafluorethylene (PTFE) and most commonly ferred method for assessing dialysis adequacy.
involves anastomosis of the brachial artery and basilic vein.30 One Though a number of factors can affect Kt/V, such as blood flow
benefit of an AVG over an AVF is that it can be used for HD much rate, dialysate flow rate, and the size of the dialysis membrane, the
sooner, typically within 2 weeks of placement.31 Because the AVG is most easily modifiable variable to achieve adequate dialysis is the
constructed of synthetic material, rates of stenosis, thrombosis, and time spent on treatment.
infection are higher than an AVF.30 Like AVF, prolonged or unpro- The minimum Kt/V was established by the HEMO trial.40 In this
voked bleeding from an AVG warrants prompt surgical investigation, study, patients were randomized to the either a lower clearance
especially if associated with aneurysmal dilation or skin breakdown, group with single treatment Kt/V 1.2 or a higher clearance group
as there may be little separating high vascular flow from the outside with single treatment Kt/V 1.65. At a mean of 2.8 years of follow-
world beyond a thin layer of skin or an eschar. Also like AVF, the up, there was no difference in overall patient mortality or
FOY AND SPERATI | 5

hospitalization rates. Current Kidney Disease Improving Global Out- abdominal surgeries or uncorrected hernias may preclude PD, and
comes (KDIGO) guidelines recommend a single treatment Kt/V the inherent biologic properties of the peritoneal membrane may
greater than 1.2 and a URR greater than 65%.16,39 Despite these render some patients less suitable for this modality.
recommendations, consideration of adequate clearance should still PD catheters are tunneled from an entry point from either below
be given on a case-by-case basis, taking into account patient residual the beltline or the presternal region and are coiled internally in the pel-
renal function, uremic symptoms, nutrition markers, and overall func- vis (Figure 2). Ideally, one should wait 2-4 weeks before instilling dialy-
tional status. sate to minimize leaks at the insertion site.46 Fundamentally, PD utilizes
the parietal peritoneum as a biological filter to exchange solute and
fluid. Solute exchange is described according to the 3-pore model, in
1.8 | Dialysis delivery
which ultrasmall pores (aquaporin-1) mediate water flux along osmotic
Outpatient HD predominantly occurs within a dialysis treatment facil- gradients, small pores (40-60 !
A) transport readily dialyzable molecules
ity, though home HD treatments have grown in popularity in recent such as sodium and urea, and large pores (100-200 !
A) are responsible
years, with 2.5-fold higher use of home HD in 2014 compared to for movement of macromolecules such as b-2 microglobulin.47 As the
1
2000. In-center HD typically consists of thrice weekly treatments biology of the peritoneum differs for each patient, so does the length of
lasting 3.5-4.5 hours, though alternative options such as extended time the dialysate must dwell to allow for efficient and effective dialy-
HD treatments and in-center nocturnal dialysis are also available in sis. A peritoneal equilibration test (PET) is performed to categorize
some settings. With home HD, patients may opt for thrice weekly, patients as low, low average, high average, or high transporters.
longer, or more frequent treatments. To perform home HD, patients Most patients perform automated peritoneal dialysis (APD) utiliz-
must be trained in self-cannulation of their dialysis access and the ing a cycler, in which a machine (Figure 3) performs approximately
basic functions of the dialysis machine. Selection of in-center versus 4-6 exchanges of 1.5-3 L of dialysate while the patient is sleeping.
home HD is dependent on regional availability, patient home support, Variations on the mode of delivery allow PD to be individualized to
functional status, and patient preference.16 While interest is growing
in home HD, rigorous outcome data comparing home HD to more
traditional RRT modalities do not exist. In the Frequent Hemodialysis
Network Nocturnal Trial, participants were randomized to thrice
weekly in-center HD versus 6 nights a week home nocturnal HD.41
This failed to demonstrate a survival difference but was limited by
lack of power; patients receiving frequent HD, however, have
reported a better health-related quality of life.42

2 | CONTINUOUS RENAL REPLACEMENT


THERAPY

2.1 | Peritoneal dialysis


There exist several forms of continuous renal replacement therapy
(CRRT), of which peritoneal dialysis (PD) is the only modality appro-
priate for outpatient management of ESRD. PD was also the CRRT
modality for unstable hospitalized patients with acute kidney injury F I G U R E 2 AP abdominal radiograph of a patient on peritoneal
dialysis. The tip (arrow) of the catheter is coiled in the pelvic cavity
until the introduction of continuous arteriovenous (CAV) therapy in
the 1980s. While there is a growing interest again in the use of
urgent-start PD for hospitalized patients, virtually all hemodynami-
cally unstable inpatients in the United States are currently managed
with continuous venovenous (CVV) therapy.43 PD is often continued
for hospitalized ESRD patients already on this modality, although
strong consideration should be given to the use of CVV in such
patients who are critically ill in the ICU.44,45
PD offers several advantages over in-center HD: (1) patients
have greater control over their therapy, (2) dialysis is performed on a
daily basis, (3) volume shifts are less as compared to thrice weekly
HD and as such potentially better tolerated in the setting of signifi-
cant cardiovascular disease, and (4) it is bloodless. At the same time,
successful PD requires a motivated, attentive patient. Prior intra- FIGURE 3 Automated peritoneal dialysis cycler with dialysate
6 | FOY AND SPERATI

T A B L E 3 Peritoneal dialysis modalities


Abbreviation Modality Description Comments
Automated PD (APD)
NIPD Nocturnal intermittent PD Machine performs automated exchanges Minimizes disruption to patient’s schedule while awake.
over 8-10 hr while patient is sleeping Best for “high transporters,” but may not achieve
adequate clearance of unmeasured “middle molecules.”
CCPD Continuous cycling PD At the conclusion of nocturnal cycling, the Convenience of nocturnal exchanges with the addition
machine leaves a “last fill” in the of at least 1 longer daytime exchange to augment
peritoneal cavity. The patient may or may middle molecule clearance. Fewer nocturnal exchanges
not manually drain the effluent prior to with longer daytime dwells are better suited to slower
the next night’s cycling. transporters.
Manual
CAPD Continuous ambulatory PD Patient performs multiple manual Provides best clearance for low transporters but
exchanges during waking hours. requires significant time commitment from patients.

the needs of the patient (Table 3). Ultrafiltration is achieved by the In 1-2% of patients, hydrothorax may develop due to transuda-
osmosis of water along osmotic gradients established by the dex- tion of dialysate through pleuroperitoneal connections.58 In such
trose concentration of dialysate. Patients adjust their ultrafiltration cases, the glucose concentration in the pleural fluid should approach
goal based on their weight and will typically refer to the dextrose that of the dialysate, and technetium-99 m scintigraphy may aid in
concentration by color—yellow (1.5%), green (2.5%), and red diagnosis. Lastly, patients should avoid submerging the catheter in a
(4.25%). The use of 4.25% dextrose will result in larger and more bath or swimming in freshwater. Swimming in the ocean or a chlori-
sustained ultrafiltration, although frequent use leads to sclerosis and nated pool is possible, although should be done with the approval of
failure of the peritoneal membrane over time. Less commonly, the PD program.
icodextrin rather than dextrose-based dialysate will be utilized; it can
falsely elevate blood glucose values with certain glucometers.48,49
2.2 | Continuous venovenous therapy
This effect may persist for 2 weeks after the discontinuation of
icodextrin-based PD. In critically ill patients requiring renal replacement therapy, CVV
Importantly, residual renal function (urine output > 100 mL/day) therapies are the modality of choice. By design, CVV therapies run
is critical for achieving adequate solute and fluid clearance in at slower blood and dialysate flow rates to permit slower solute
patients on PD and is associated with lower mortality.50,51 Renin- clearance with less destabilizing effects on patient hemodynamics.
angiotensin-aldosterone system antagonists, commonly prescribed, As shown in Table 4, dialysate flow rates with continuous venove-
may help slow the loss of residual renal function. Potential nephro- nous hemodialysis (CVVHD) may be 20-fold slower than typically
toxins such as NSAIDs and iodinated intravenous contrast should be prescribed in intermittent HD. CVVHD requires approximately
avoided when possible.52,53 24 hours of therapy to approach the small solute clearance achieved
PD is associated with several potential complications. Hyper- in 3-4 hours with intermittent HD. CVV therapies are particularly
glycemia from the high dextrose content (2.5% exchange =2500 mg/ well-suited for patients with unstable hemodynamics, increased
dL dextrose) of the dialysate may worsen or induce diabetes mellitus. intracranial pressure, severe hypo- or hypernatremia, acute liver fail-
Transient dysfunction of the catheter is common, often due to intralu- ure, and poisonings with dialyzable substances that possess a large
minal fibrin or patient constipation. Abdominal hernias develop at a volume of distribution. CVV therapies require the use of a central
rate of 0.04-0.08 hernias/patient/year and are more common in venous catheter, and while a small report describes successful use of
patients utilizing larger dwell volumes or performing ambulatory PD AVF/AVG for CVV delivery, it can lead to significant access compli-
while upright.54,55 In patients in whom the PD catheter may have been cations.59 Of note, in patients currently undergoing extracorporeal
internally sutured, bowel obstruction due to an internal hernia may membrane oxygenation (ECMO), the CVV machine can be placed in
develop, requiring a high index of suspicion and, at times, laparoscopic series such that a separate vascular access is not required.
evaluation for diagnosis. There exist several variations on CVV therapy. With CVVHD,
Exit site infections and peritonitis are worrisome complications. clearance is achieved by the diffusion of solutes between blood and
When performed with optimal technique, PD peritonitis rates should dialysate across the semipermeable filter. In contrast, continuous
be <0.39 per patient year.56 Abdominal pain and/or cloudy effluent venovenous hemofiltration (CVVH) does not utilize dialysate, but
should prompt evaluation for peritonitis.57 The catheter can often be rather achieves solute clearance by ultrafiltration of large volumes of
preserved, at least initially, with exit site infections and most cases plasma (500-5000 mL/h) with simultaneous administration of physio-
of peritonitis. Fungal peritonitis is one indication for which the PD logically balanced replacement fluid to maintain fluid balance. Solute
catheter should almost universally be removed. clearance is achieved through convection, or “solvent drag,” across
FOY AND SPERATI | 7

T A B L E 4 Continuous hemodialysis modalities


Ultra/
Blood flow Dialysate hemofiltration
Abbreviation Modality (mL/min) ratea,b (mL/h) rate (mL/h) Dialysis staff Nursing staff
CVVHD Continuous venovenous hemodialysis 200-300 1000-5000 Variable + +++
CVVH Continuous venovenous hemofiltration 200-300 n/a 1000-5000b + +++
b
CVVHDF Continuous venovenous hemodiafiltration 200-300 1000-5000 1000-5000b + +++
SCUF Slow continuous ultrafiltration 200-300 n/a Variable + +++
SLED Slow low-efficiency hemodialysis 200-300 1000-2000 Variable ++++ +
a
Typical dialysate rates are shown. Maximum rates vary by machine and clinical indication.
b
Typical prescription is 20-25 mL/kg/h. In CVVHDF, the total rate will be split between the dialysate and hemofiltration rates.

the filter. Both modalities may be combined to provide hemodiafil- renal replacement therapy (PIRRT).64 These modalities require less
tration (CVVHDF). No data support a meaningful difference in out- critical care nursing resources but limit the number of dialysis treat-
come across the different therapies.60 ments that can be provided on a daily basis.
In all modalities, the sodium, potassium, and calcium within the Hypophosphatemia develops in many patients and requires sup-
dialysate/replacement fluid can be altered to meet the clinical need. plementation or dialysis dose reduction. Thrombocytopenia presum-
In light of the slower blood flow rates characteristic of CVV thera- ably due to platelet-filter interactions may also be seen.65
pies, catheter and filter clotting are frequent complications. To
reduce clotting, intravenous heparin is often used. As many critically
ill patients are not candidates for systemic heparin therapy, alterna- 3 | OUTCOME DATA
tive interventions may be required. Frequent replacement of filters
and catheters, utilizing blood flows ≥300 mL/min, and the use of According to 2016 United States Renal Data System data, the unad-
citrate anticoagulation can be helpful interventions. Regional citrate justed 5-year survival rate of ESRD patients who initiated therapy
anticoagulation entails the prefilter administration of citrate followed on intermittent HD was 41.6% and 51.5% for patients on PD.1 It is
by chelation and inactivation by postfilter administration of intra- unclear if PD or HD offers a relative survival advantage, but in prop-
venous calcium. While this approach minimizes the risk for systemic erly selected candidates, the mortality rate appears similar between
anticoagulation and bleeding, it requires constant attention to avoid the modalities out to 5 years.56
61
citrate toxicity as well as hypo- or hypercalcemia. The use of regio- In critically ill patients, there is no compelling evidence that CVV
nal citrate anticoagulation is limited by the need for specialty solu- therapy versus intermittent HD impacts mortality or regain of renal
tions, complex protocols, and lack of familiarity at many institutions function, provided the patient is hemodynamically appropriate for
among nurses and nephrologists.62 the selected modality.66 Based on several randomized clinical trials,
Slow continuous ultrafiltration (SCUF) or continuous venovenous the dialysate rate in CVVHD or ultrafiltration rate in CVVH for
ultrafiltration (CVVU) is utilized for management of refractory volume patients with AKI is generally prescribed at 20-25 mL/kg/h, with
overload. As dialysate is not utilized and hourly UF rates are slower additional adjustments based on clinical circumstances.67-70 In criti-
than with CVVH, this results primarily in volume removal without cally ill patients receiving intermittent HD, thrice weekly HD was
appreciable convective clearance. In a randomized controlled trial associated with similar outcomes compared to a more intensive
comparing isolated UF to diuretics in the management of decompen- treatment strategy.67 A common problem with CVV therapy is treat-
sated congestive heart failure, isolated UF led to a higher creatinine ment interruption for medical procedures or system clotting. As
and similar weight loss.63 Thus, while isolated UF (and specifically such, the prescriber must be attentive to the cumulative “down-
SCUF in patients hemodynamically inappropriate for intermittent ther- time” and adjust the prescription as needed to ensure adequate
apy) has a role in the management of volume overload that has failed solute/fluid clearance. For critically ill patients with AKI, the optimal
maximal medical therapy, it is inappropriate as initial therapy. timing of dialysis initiation is unknown; a delayed initiation strategy
Importantly, CVV therapies are typically monitored by an ICU (ie, awaiting an indication for dialysis) may be as beneficial in regard
nurse, and as such, permit simultaneous delivery of RRT to more to mortality as an early start strategy.71 For AKI patients treated by
patients than could otherwise be accomplished with a limited dialysis intermittent HD, a typical prescription is 9-12 hours per week, often
nursing staff. This requires appropriate training of critical care nurs- delivered over 3-4 sessions.
ing staff and may impose limitations on nurse:patient ratios. An alter-
native is slow low-efficiency hemodialysis (SLED), in which a dialysis
nurse performs a 6-8 hour dialysis session at blood and dialysate 4 | MEDICATION DOSING
flows intermediate between CVVHD and intermittent HD. This is an
emerging arena within renal replacement therapy with multiple Appropriate dose adjustment for medications administered to
implementations, sometimes referred to as prolonged intermittent patients on dialysis is understudied and of significant importance to
8 | FOY AND SPERATI

clinical outcomes and patient safety. For many medications, guideli- 8. Institute of Medicine (US) Committee for the Study of the Medicare
nes for dosing on intermittent HD are often available, although not End-Stage Renal Disease Program; Rettig RA, Levinsky NG, editors.
Kidney Failure and the Federal Government. Washington, DC: National
always rigorously derived. For example, the package insert for apixa-
Academies Press (US); 1991.
ban does not recommend dose reduction for most patients on dialy- 9. Rettig RA. Special treatment—the story of Medicare’s ESRD entitle-
sis, a recommendation provided by a study of 8 patients receiving a ment. N Engl J Med. 2011;364:596-598.
dose lower than the prescribing instructions.72,73 A postmarketing 10. Kucirka LM, Grams ME, Lessler J, et al. Association of race and age
with survival among patients undergoing dialysis. JAMA. 2011;306:
study, however, has revealed significant drug accumulation at recom-
620-626.
mended doses, suggesting the dose on intermittent HD should 11. Johns TS, Estrella MM, Crews DC, et al. Neighborhood socioeco-
potentially be 50% less.74 nomic status, race, and mortality in young adult dialysis patients. J
For CRRT, dose adjustments are often not available. In the absence Am Soc Nephrol. 2014;25:2649-2657.
12. Hecking M, Bieber BA, Ethier J, et al. Sex-specific differences in
of guidelines, medications in patients on CVV therapies are often
hemodialysis prevalence and practices and the male-to-female mor-
dosed for a creatinine clearance equivalent to the dialysate (CVVHD) tality rate: the Dialysis Outcomes and Practice Patterns Study
or ultrafiltration (CVVH) rate. For example, at a dialysate flow rate of (DOPPS). PLoS Med. 2014;11:e1001750.
2000 mL/h on CVVHD, medications are often dosed for a creatinine 13. Sperati CJ. Hemodialysis: Initiation and Complications. In: Lerma EV,
Rosner M, eds. Clinical Decisions in Nephrology, Hypertension, and Kid-
clearance of approximately 30 mL/min (2000 mL/h " 60 min/
ney Transplantation. New York: Springer; 2013:333-348.
h = 33 mL/min). This assumes the medication is freely dialyzable, an
14. Daugirdas JT, Blake PG, Ing TS. Handbook of dialysis, 5th ed.
assumption that may not be accurate. As such, when possible, drug Philadelphia: Wolters Kluwer Health; 2015.
levels should be followed and doses adjusted accordingly. Significant 15. Cooper BA, Branley P, Bulfone L, et al. A randomized, controlled trial
potential exists for both over and under-dosing medications, in partic- of early versus late initiation of dialysis. N Engl J Med. 2010;363:
609-619.
ular antibiotics such as cefepime.75-77 Moreover, data are largely
16. National Kidney Foundation. KDOQI Clinical Practice Guideline for
nonexistent regarding the quantity of medication removed by convec- Hemodialysis Adequacy: 2015 update. Am J Kidney Dis. 2015;66:
tion typical of lower volume isolated ultrafiltration. Thus, while supple- 884-930.
mental doses are generally not required after an IUF session, the true 17. Neri M, Villa G, Garzotto F, et al. Nomenclature for renal replace-
ment therapy in acute kidney injury: basic principles. Crit Care.
impact on drug concentration is often unknown. Close coordination
2016;20:318.
with pharmacists and prescribers is required whenever the nephrolo- 18. Prakash S, Garg AX, Heidenheim AP, House AA. Midodrine appears
gist alters the dialysis prescription and/or modality. to be safe and effective for dialysis-induced hypotension: a system-
In summary, multiple strategies exist for the safe and life-saving atic review. Nephrol Dial Transplant. 2004;19:2553-2558.
19. Ronco C, Ghezzi PM, Brendolan A, Crepaldi C, La Greca G. The
delivery of dialysis therapies. While the specifics of dialysis prescrip-
haemodialysis system: basic mechanisms of water and solute trans-
tion are the responsibility of the nephrologist, providers from all dis- port in extracorporeal renal replacement therapies. Nephrol Dial
ciplines will potentially care for patients with ESRD. Familiarity with Transplant. 1998;13(Suppl 6):3-9.
dialysis modalities and the therapeutic concerns specific to these 20. Nistor I, Palmer SC, Craig JC, et al. Convective versus diffusive dialy-
sis therapies for chronic kidney failure: an updated systematic
technologies may facilitate improved multidisciplinary patient care.
review of randomized controlled trials. Am J Kidney Dis. 2014;63:
954-967.
21. Locatelli F, Manzoni C, Di Filippo S. The importance of convective
REFERENCES transport. Kidney Int Suppl. 2002;80:115-120.
22. Boure T, Vanholder R. Which dialyser membrane to choose? Nephrol
1. United States Renal Data System. 2016 USRDS annual data report: Dial Transplant. 2004;19:293-296.
epidemiology of kidney disease in the United States. Bethesda, MD: 23. Schiffl H. High-flux dialyzers, backfiltration, and dialysis fluid quality.
National Institutes of Health, National Institute of Diabetes and Semin Dial. 2011;24:1-4.
Digestive and Kidney Diseases; 2016. 24. Coulliette AD, Arduino MJ. Hemodialysis and water quality. Semin
2. Abel JJ, Rowntree LG, Turner BB. On the removal of diffusible sub- Dial. 2013;26:427-438.
stances from the circulating blood of living animals by dialysis. J 25. Bommer J, Jaber BL. Ultrapure dialysate: facts and myths. Semin Dial.
Pharmacol Exp Ther. 1914;5:275-316. 2006;19:115-119.
3. Kolff WJ, Berk HTJ. The artificial kidney: a dialyser with a great area. 26. ANSI/AAMI/ISO. Water for Hemodialysis and Related Therapies. ANSI/
Acta Med Scand. 1944;117:121-134. AAMI 13959: 2014. Arlington, VA: Association for the Advancement
4. Blagg CR. The early history of dialysis for chronic renal failure in the of Medical Instrumentation; 2014.
United States: a view from Seattle. Am J Kidney Dis. 2007;49:482- 27. Weijmer MC, Vervloet MG, ter Wee PM. Compared to tunnelled
496. cuffed haemodialysis catheters, temporary untunnelled catheters are
5. Boen ST, Mulinari AS, Dillard DH, Scribner BH. Periodic peritoneal associated with more complications already within 2 weeks of use.
dialysis in the management of chronic uremia. Trans Am Soc Artif Nephrol Dial Transplant. 2004;19:670-677.
Intern Organs. 1962;8:256-262. 28. Nguyen DB, Shugart A, Lines C, et al. National Healthcare Safety
6. Peitzman SJ. Chronic dialysis and dialysis doctors in the United Network (NHSN) dialysis event surveillance report for 2014. Clin J
States: a nephrologist-historian’s perspective. Semin Dial. 2001;14: Am Soc Nephrol. 2017;12:1139-1146.
200-208. 29. Mermel LA, Allon M, Bouza E, et al. Clinical practice guidelines for
7. Brescia MJ, Cimino JE, Appel K, Hurwich BJ. Chronic hemodialysis the diagnosis and management of intravascular catheter-related
using venipuncture and a surgically created arteriovenous fistula. N infection: 2009 Update by the Infectious Diseases Society of Amer-
Engl J Med. 1966;275:1089-1092. ica. Clin Infect Dis. 2009;49:1-45.
FOY AND SPERATI | 9

30. Rose DA, Sonaike E, Hughes K. Hemodialysis access. Surg Clin North a prospective, randomized, controlled trial. J Am Soc Nephrol.
Am. 2013;93:997-1012. 2002;13:1307-1320.
31. Vascular Access Work Group. Clinical practice guidelines for vascular 52. Htay H, Cho Y, Pascoe EM, et al. Predictors of residual renal func-
access. Am J Kidney Dis. 2006;48(Suppl 1):S176-247. tion decline in peritoneal dialysis patients: the balANZ Trial. Perit
32. Shechter SM, Skandari MR, Zalunardo N. Timing of arteriovenous Dial Int. 2017;37:283-289.
fistula creation in patients with CKD: a decision analysis. Am J Kid- 53. Zhang L, Zeng X, Fu P, Wu HM. Angiotensin-converting enzyme
ney Dis. 2014;63:95-103. inhibitors and angiotensin receptor blockers for preserving residual
33. Ellingson KD, Palekar RS, Lucero CA, et al. Vascular access hemor- kidney function in peritoneal dialysis patients. Cochrane Database
rhages contribute to deaths among hemodialysis patients. Kidney Int. Syst Rev. 2014;23:CD009120.
2012;82:686-692. 54. Del Peso G, Bajo MA, Costero O, et al. Risk factors for abdominal
34. Shrestha B, Boyes S, Brown P. Innocuous-looking skin scab over an wall complications in peritoneal dialysis patients. Perit Dial Int.
arteriovenous fistula: case report and literature review. World J 2003;23:249-254.
Nephrol. 2014;3:118-121. 55. Yang SF, Liu CJ, Yang WC, et al. The risk factors and the impact of
35. Huber TS, Carter JW, Carter RL, Seeger JM. Patency of autogenous hernia development on technique survival in peritoneal dialysis
and polytetrafluoroethylene upper extremity arteriovenous patients: a population-based cohort study. Perit Dial Int. 2015;35:
hemodialysis accesses: a systematic review. J Vasc Surg. 2003;38: 351-359.
1005-1011. 56. Mehrotra R, Devuyst O, Davies SJ, Johnson DW. The current state
36. Saha M, Allon M. Diagnosis, treatment, and prevention of hemodial- of peritoneal dialysis. J Am Soc Nephrol. 2016;27:3238-3252.
ysis emergencies. Clin J Am Soc Nephrol. 2017;12:357-369. 57. Li PKT, Szeto CC, Piraino B, et al. ISPD peritonitis recommendations:
37. Zepeda-Orozco D, Quigley R. Dialysis disequilibrium syndrome. Pedi- 2016 update on prevention and treatment. Perit Dial Int. 2016;36:
atr Nephrol. 2012;27:2205-2211. 481-508.
38. Davenport A. Antibodies to heparin-platelet factor 4 complex: 58. Nomoto Y, Suga T, Nakajima K, et al. Acute hydrothorax in continu-
pathogenesis, epidemiology, and management of heparin-induced ous ambulatory peritoneal dialysis—a collaborative study of 161
thrombocytopenia in hemodialysis. Am J Kidney Dis. 2009;54:361- centers. Am J Nephrol. 1989;9:363-367.
374. 59. Al Rifai A, Sukul N, Wonnacott R, Heung M. Safety of arteriovenous
39. Hemodialysis Adequacy Work Group. Clinical practice guidelines for fistulae and grafts for continuous renal replacement therapy: the
hemodialysis adequacy, update 2006. Am J Kidney Dis. 2006;48 Michigan experience. Hemodial Int. Mar 13 2017. https://doi.org/10.
(Suppl 1):S2-90. 1111/hdi.12550.
40. Eknoyan G, Beck GJ, Cheung AK, et al. Effect of dialysis dose and 60. Kidney Disease: Improving Global Outcomes (KDIGO) Acute Kidney
membrane flux in maintenance hemodialysis. N Engl J Med. Injury Work Group. KDIGO clinical practice guideline for acute kid-
2002;347:2010-2019. ney injury. Kidney Int Suppl 2012; 2: 1-138.
41. Rocco MV, Lockridge RS Jr., Beck GJ, et al. The effects of frequent 61. Bai M, Zhou M, He L, et al. Citrate versus heparin anticoagulation
nocturnal home hemodialysis: the Frequent Hemodialysis Network for continuous renal replacement therapy: an updated meta-analysis
Nocturnal Trial. Kidney Int. 2011;80:1080-1091. of RCTs. Intensive Care Med. 2015;41:2098-2110.
42. Garg AX, Suri RS, Eggers P, et al. Patients receiving frequent 62. Morabito S, Pistolesi V, Tritapepe L, Fiaccadori E. Regional citrate
hemodialysis have better health-related quality of life compared to anticoagulation for RRTs in critically ill patients with AKI. Clin J Am
patients receiving conventional hemodialysis. Kidney Int. Soc Nephrol. 2014;9:2173-2188.
2017;91:746-754. 63. Bart BA, Goldsmith SR, Lee KL, et al. Ultrafiltration in decompen-
43. Alkatheeri AM, Blake PG, Gray D, Jain AK. Success of urgent-start sated heart failure with cardiorenal syndrome. N Engl J Med.
peritoneal dialysis in a large Canadian renal program. Perit Dial Int. 2012;367:2296-2304.
2016;36:171-176. 64. Edrees F, Li T, Vijayan A. Prolonged intermittent renal replacement
44. Phu NH, Hien TT, Mai NT, et al. Hemofiltration and peritoneal dialy- therapy. Adv Chronic Kidney Dis. 2016;23:195-202.
sis in infection-associated acute renal failure in Vietnam. N Engl J 65. Bellomo R, Cass A, Cole L, et al. The relationship between
Med. 2002;347:895-902. hypophosphataemia and outcomes during low-intensity and high-
45. Gabriel DP, Caramori JT, Martim LC, Barretti P, Balbi AL. High vol- intensity continuous renal replacement therapy. Crit Care Resusc.
ume peritoneal dialysis vs daily hemodialysis: a randomized, con- 2014;16:34-41.
trolled trial in patients with acute kidney injury. Kidney Int Suppl. 66. Tonelli M, Manns B, Feller-Kopman D. Acute renal failure in the
2008;108:S87-93. intensive care unit: a systematic review of the impact of dialytic
46. Ranganathan D, John GT, Yeoh E, et al. A randomized controlled modality on mortality and renal recovery. Am J Kidney Dis. 2002;40:
trial to determine the appropriate time to initiate peritoneal dialysis 875-885.
after insertion of catheter (Timely PD Study). Perit Dial Int. 67. VI NIH, Acute Renal Failure Trial Network, Palevsky PM, Zhang JH,
2017;37:420-428. et al. Intensity of renal support in critically ill patients with acute
47. Perl J, Bargman JM. Peritoneal dialysis: from bench to bedside and kidney injury. N Engl J Med. 2008;359:7-20.
bedside to bench. Am J Physiol Renal Physiol. 2016;311:F999-F1004. 68. RENAL Replacement Therapy Study Investigators, Bellomo R, Cass
48. Wens R, Taminne M, Devriendt J, et al. A previously undescribed A, et al. Intensity of continuous renal-replacement therapy in criti-
side effect of icodextrin: overestimation of glycemia by glucose ana- cally ill patients. N Engl J Med. 2009;361:1627-1638.
lyzer. Perit Dial Int. 1998;18:603-609. 69. Prowle JR, Schneider A, Bellomo R. Clinical review: optimal dose of
49. Extraneal full prescribing information. http://www.baxterpi.com/pi- continuous renal replacement therapy in acute kidney injury. Crit
pdf/Extraneal_PI.pdf. Accessed July 6, 2017. Care. 2011;15:207.
50. Bargman JM, Thorpe KE, Churchill DN, CANUSA Peritoneal Dialysis 70. Fayad AI, Buamscha DG, Ciapponi A. Intensity of continuous renal
Study Group. Relative contribution of residual renal function and replacement therapy for acute kidney injury. Cochrane Database Syst
peritoneal clearance to adequacy of dialysis: a reanalysis of the Rev. 2016;10:CD010613.
CANUSA study. J Am Soc Nephrol. 2001;12:2158-2162. 71. Gaudry S, Hajage D, Schortgen F, et al. Initiation strategies for
51. Paniagua R, Amato D, Vonesh E, et al. Effects of increased peri- renal-replacement therapy in the intensive care unit. N Engl J Med.
toneal clearances on mortality rates in peritoneal dialysis: ADEMEX, 2016;375:122-133.
10 | FOY AND SPERATI

72. Eliquis full prescribing information. http://packageinserts.bms.com/ 77. Huwyler T, Lenggenhager L, Abbas M, et al. Cefepime plasma con-
pi/pi_eliquis.pdf. Accessed July 6, 2017. centrations and clinical toxicity: a retrospective cohort study. Clin
73. Wang X, Tirucherai G, Marbury TC, et al. Pharmacokinetics, pharma- Microbiol Infect. 2017;23:454-459.
codynamics, and safety of apixaban in subjects with end-stage renal
disease on hemodialysis. J Clin Pharmacol. 2016;56:628-636.
74. Mavrakanas TA, Samer CF, Nessim SJ, Frisch G, Lipman ML. Apixa-
ban pharmacokinetics at steady state in hemodialysis patients. J Am How to cite this article: Foy M, Sperati CJ. What the non-
Soc Nephrol. 2017;28:2241-2248. nephrologist needs to know about dialysis? Semin Dial.
75. Hobbs AL, Shea KM, Roberts KM, Daley MJ. Implications of aug-
2018;00:1–10. https://doi.org/10.1111/sdi.12671
mented renal clearance on drug dosing in critically ill patients: a
focus on antibiotics. Pharmacotherapy. 2015;35:1063-1075.
76. Nolin TD, Aronoff GR, Fissell WH, et al. Pharmacokinetic assessment
in patients receiving continuous RRT: perspectives from the Kidney
Health Initiative. Clin J Am Soc Nephrol. 2015;10:159-164.

Você também pode gostar