Content uploaded by Nihar Jena
Author content
All content in this area was uploaded by Nihar Jena on Jan 05, 2024
Content may be subject to copyright.
Advances in Renal
Diseases and Dialysis
Advances in Renal
Diseases and Dialysis
India . United Kingdom
Author(s)
Maheshwari Kata a*, Harkesh Arora b, Rajeshwari Ramachandran c,
Varun Victor d, Nihar Jena e, Abid Rizvi f, Prashanth Reddy Yella g,
Atul Bali h, Deepak Chandramohan i and Roopa Naik j
a Department of Internal Medicine, MNR Medical College and Hospital,
Telangana, India.
b Department of Internal Medicine, Lovelace Medical Center, Albuquerque, New
Mexico, USA.
c Department of Gastroenterology, Brooklyn Hospital Center, Brooklyn, New
York, USA.
d Department of Cardiology, Canton Medical Education Foundation/Aultman
Hospital, Canton, Ohio, USA.
e Department of Cardiology, Trinity Health Oakland/Wayne State University,
Pontiac, Michigan, USA.
f Department of Behavioural Health and Psychiatry, West Virginia University,
Morgantown, West Virginia, USA.
g Department of Internal Medicine, Yuma Regional Medical Center, Yuma,
Arizona, USA.
h Department of Internal Medicine/Nephrology, Geisinger Wyoming Valley
Medical Center, Wilkes-Barre, Pennsylvania, USA.
i Department of Internal Medicine/Nephrology, University of Alabama at
Birmingham, Birmingham, Alabama, USA.
j Department of Internal Medicine, Geisinger Wyoming Valley Medical Center,
Wilkes-Barre, Pennsylvania, USA.
*Corresponding author: E-mail: drmaheshwarikata@gmail.com;
FIRST EDITION 2023
ISBN 978-81-19491-84-1 (Print)
ISBN 978-81-19491-85-8 (eBook)
DOI: 10.9734/bpi/mono/978-81-19491-84-1
__________________________________________________________
© Copyright (2023): Author(s). The licensee is the publisher (B P International).
Contents
Preface
i
Abstract
1
Chapter 1
Acute Kidney Injury
Deepak Chandramohan, Juan J. Cintrón García,
Maheshwari Kata, Roopa Naik, Atul Bali,
Bushra Firdous Shaik, Harkesh Arora and Sreekant Avula
2-18
Chapter 2
Hepatorenal Syndrome
Roopa Naik, Atul Bali, Bushra Firdous Shaik,
Maheshwari Kata, Harkesh Arora, Deepak Chandramohan
and Rajeshwari Ramachandran
19-34
Chapter 3
Diabetic Kidney Disease
Roopa Naik, Rajeshwari Ramachandran,
Bushra Firdous Shaik and Atul Bali
35-53
Chapter 4
Continuous Renal Replacement Therapy
Atul Bali, Harkesh Arora, Roopa Naik,
Maheshwari Kata and Rajeshwari Ramachandran
54-67
Advances in Renal Diseases and Dialysis
Preface
i
PREFACE
As a physician who is interested in research related to kidney diseases, I always
felt the need to improve current knowledge and available research in this field. I
was fortunate to have met a group of physicians from different parts of the world
with a passion for research in medicine and especially in nephrology related
aspects. The feeling is amazing to have finally completed the project after
several dedicated preparation.
In this book we tried to cover as many key aspects related to the kidney diseases
as possible and provide updates regarding the latest research along with pointing
deficiencies in certain aspects for future. We aim to continue to contribute to the
field of medicine and nephrology in future with interesting topics. We hope this
book help to enhance knowledge among learners related to medicine and
research.
___________________________________________________________________________________
© Copyright (2023): Author(s). The licensee is the publisher (B P International).
________________________________________________________________________
a Department of Internal Medicine, MNR Medical College and Hospital, Telangana, India.
b Department of Internal Medicine, Lovelace Medical Center, Albuquerque, New Mexico, USA.
c Department of Gastroenterology, Brooklyn Hospital Center, Brooklyn, New York, USA.
d Department of Cardiology, Canton Medical Education Foundation/Aultman Hospital, Canton, Ohio,
USA.
e Department of Cardiology, Trinity Health Oakland/Wayne State University, Pontiac, Michigan, USA.
f Department of Behavioural Health and Psychiatry, West Virginia University, Morgantown, West Virginia,
USA.
g Department of Internal Medicine, Yuma Regional Medical Center, Yuma, Arizona, USA.
h Department of Internal Medicine/Nephrology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre,
Pennsylvania, USA.
i Department of Internal Medicine/Nephrology, University of Alabama at Birmingham, Birmingham,
Alabama, USA.
j Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre,
Pennsylvania, USA.
*Corresponding author: E-mail: drmaheshwarikata@gmail.com;
Print ISBN: 978-81-19491-84-1, eBook ISBN: 978-81-19491-85-8
Advances in Renal Diseases and Dialysis
Maheshwari Kata a*, Harkesh Arora b,
Rajeshwari Ramachandran c, Varun Victor d, Nihar Jena e,
Abid Rizvi f, Prashanth Reddy Yella g, Atul Bali h,
Deepak Chandramohan i and Roopa Naik j
DOI: 10.9734/bpi/mono/978-81-19491-84-1/CH0
ABSTRACT
This book primarily covers key areas of kidney diseases and recent
developments in dialysis. The contributions by the authors include acute kidney
injury, chronic kidney disease, newer biomarkers of AKI, epidemiology,
hepatorenal syndrome, cirrhosis, liver transplantation, acute tubular necrosis,
vasopressors, newer biomarkers of HRS, diabetic kidney disease, microvascular
damage, microalbuminuria, progression of CKD, RAS mechanism of action,
continuous renal replacement therapy, intermittent hemodialysis, hemofiltration,
hemodiafiltration, solute removal, electrolyte disorders, uremia,
hyperammonemia, diffusion. This book contains various materials suitable for
students, researchers and academicians in the field of medicine and medical
research.
Keywords: Acute kidney injury; chronic kidney disease; hepatorenal syndrome;
liver transplantation; acute tubular necrosis; vasopressors; newer
biomarkers of HRS; diabetic kidney disease; microvascular damage;
microalbuminuria; RAS mechanism of action; continuous renal
replacement therapy; intermittent hemodialysis; hemofiltration;
hemodiafiltration; solute removal; electrolyte disorders; uremia;
hyperammonemia; diffusion.
___________________________________________________________________________________
© Copyright (2023): Author(s). The licensee is the publisher (B P International).
________________________________________________________________________
a Department of Internal Medicine/Nephrology, University of Alabama at Birmingham, Birmingham,
Alabama, USA.
b Department of Internal Medicine/Nephrology, Loyola University Medical Center, Maywood, Illinois,
USA.
c Department of Internal Medicine, MNR Medical College and Hospital, Telangana, India.
d Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA,
USA.
e Department of Internal Medicine/Nephrology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre,
Pennsylvania, USA.
f Department of Internal Medicine, University Hospital and Clinics, Lafayette, Louisiana, USA.
g Department of Internal Medicine, Lovelace Medical Center, Albuquerque, New Mexico, USA.
h Department of Internal Medicine/Endocrinology, University of Minnesota, Minneapolis, Minnesota,
USA.
*Corresponding author: E-mail: drmaheshwarikata@gmail.com;
Chapter 1
Print ISBN: 978-81-19491-84-1, eBook ISBN: 978-81-19491-85-8
Acute Kidney Injury
Deepak Chandramohan a, Juan J. Cintrón García b,
Maheshwari Kata c*, Roopa Naik d, Atul Bali e,
Bushra Firdous Shaik f, Harkesh Arora g
and Sreekant Avula h
DOI: 10.9734/bpi/mono/978-81-19491-84-1/CH1
ABSTRACT
Acute kidney injury (AKI) is defined as the sudden loss of kidney function. AKI is
part of a group of diseases collectively called acute kidney disease and disorder
(AKD), in which a slow decline in kidney function or persistent kidney dysfunction
is associated with irreversible loss of nephrons (functional units of kidney), which
can eventually lead to chronic kidney disease (CKD). New biomarkers to detect
injury before loss of function await clinical application. AKI and AKD is a major
concern worldwide. Infections and hypovolemic shock are the main causes of
AKI in low- and middle-income countries. In high-income countries, AKI occurs
most often in hospitalized elderly patients and is associated with sepsis,
medications, or invasive procedures. Infections and trauma-induced AKI are
common in all regions. The broad spectrum of AKI involves different
pathophysiological mechanisms. Management of AKI in the intensive care setting
is complex, including appropriate volume management, careful monitoring of
nephrotoxic drugs, and choice of renal replacement therapies. Fluid and
electrolyte management is essential. Because AKI can be fatal, renal
replacement therapy is often necessary. AKI generally has a poorer prognosis in
critically ill patients due to various reasons. Long-term consequences of AKI and
AKD include chronic kidney disease and morbidity due to cardiovascular disease.
Therefore, early detection and prevention of AKI are quintessential.
Keywords: Acute kidney injury; chronic kidney disease; dialysis; nephrotoxins;
renal replacement therapy.
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
3
1. INTRODUCTION
Acute kidney injury (AKI) remains a major problem in clinical medicine today [1].
It occurs in about 1-5% of all hospitalized patients. The incidence increases
significantly as the severity of the underlying cause progresses: up to 50% of
patients treated in the intensive care unit develop AKI, in many cases secondary
to disseminated infection or sepsis [2,3]. The term acute kidney injury replaced
acute renal failure, emphasizing that the progression of kidney damage begins
long before it is possible to measure adequate excretion of renal function by
standard laboratory tests. The term also refers to a continuum of predictions
where an increase in mortality is associated with even a small increase in serum
creatinine and a further increase in mortality as creatinine levels rise. The
prognosis has not significantly improved in the last 20-30 years, although
significant progress has been made in the field of intensive care and dialysis
treatment [4,5]. In the mid-1970s, 70% of all patients with AKI died. Mortality
decreased moderately in the early 1990s (30-50%) and has remained stable for
the last 20 years. The poor prognosis is due in part to the disease itself that
causes AKI, but also to the complications associated with AKI. Thus, the
invention of more effective therapeutic interventions remains an important goal of
the field of nephrology research.
2. DEFINITION OF AKI
AKI is defined as an acute impairment of renal function manifested by an
increase in serum creatinine [6]. In most patients (70%), urine production also
decreases. The definition of the syndrome is refined from time to time, and
according to the latest KDIGO guidelines, AKI can be diagnosed if the following
criteria are met: (I) an increase in serum creatinine of more than 0.3 mg/dL within
48 hours, or (II) a 1.5-fold increase in serum creatinine within a seven day period
(compared to a known or suspected baseline) and/or (III) a decrease in urine
output below 0.5 ml/kg per day for at least 6 hours [7,8]. It should not be
forgotten that serum creatinine is a poor parameter of renal function, because its
concentration begins to increase late in AKI, when at least 60% of renal function
is lost. The severity of AKI varies, and several scores can be used to distinguish
certain degrees of acute kidney failure. The RIFLE criteria distinguish risk (R),
injury (I), failure (F), loss (L), and end-stage kidney disease (E) depending on the
relative increase in serum creatinine and/or relative decrease in urine production
[9,10]. It should be noted that stage E can only be diagnosed if the renal
dysfunction lasts more than 3 months. Such criteria may not be important from a
therapeutic point of view, but from a prognostic point of view, since the prognosis
significantly worsens with the severity of kidney damage.
3. EPIDEMIOLOGY
The global mortality burden of AKI far exceeds mortality from breast cancer,
heart failure, or diabetes, and mortality has remained high for the past 50 years
[11,12]. Generally, the incidence of AKI is reported as either community-acquired
or hospital-acquired AKI. In high-income countries (HICs), AKI is mostly hospital-
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
4
acquired, while community-acquired AKI is more common in low-income
countries. In HIC, AKI patients are generally elderly, have multiple co-
morbidities, and have access to dialysis and intensive care when needed. The
main causes of AKI in HIC are postoperative or diagnostic interventions or
iatrogenic factors. However, there are many community-acquired causes in poor
environments, including sepsis, body volume depletion, poisons (bites, drugs)
and pregnancy [13,14]. Patients tend to be younger than those with HIC, access
to treatment is more difficult, and women are underrepresented in the patient
population.
4. RISK FACTORS OF AKI
Risk factors for AKI include environmental, socioeconomic and/or cultural factors,
as well as factors related to the treatment process, acute exposure and patients
themselves [15,16]. Environmental factors include inadequate drinking water and
sewage systems, inadequate infectious disease control, and inadequate health
care delivery systems. Patient-related factors may be modifiable, such as volume
depletion, hypotension, anemia, hypoxia, and use of nephrotoxic medications, or
nonmodifiable, such as chronic kidney, heart, liver, or gastrointestinal disease,
diabetes, and serious infections and sepsis [17-19]. Less common causes are
genetic predisposition to myoglobinuria, hemoglobinuria and urolithiasis. Other
important risk factors for AKI are serious illness, acute infections, sepsis, malaria,
severe trauma, hypovolemia, old age, pre-existing chronic kidney disease, acute
organ failure, major surgery (including heart surgery), intensive care unit stay
with exposure to nephrotoxic medications and acquiring opportunistic infections,
cancer chemotherapy, delayed kidney transplant graft function, autoimmune
diseases with rapidly progressive kidney damage, cholesterol crystal embolism,
and urinary tract obstruction [20-23]. Although severe AKI occurs more often in
association with hospital-associated risk factors such as major surgery,
hemorrhage, septic shock, or drug intoxication in elderly patients with multiple
diseases, milder forms of AKI can also occur in the community. In contrast, in
low- and middle-income countries (LMICs), community-acquired AKI affects
younger, previously healthy individuals, and the incidence of sepsis and obstetric
complications is relatively high. COVID-19 remains a major risk factor for AKI
worldwide [24,25].
5. KIDNEY PHYSIOLOGY AND MECHANISMS OF AKI
Renal physiology and renal lifespan: The kidneys maintain the homeostasis of
body fluids, electrolytes, osmolality and pH, excrete metabolic waste products
and secrete hormones and bioactive molecules. Because AKI disrupts
homeostasis, severe AKI is potentially fatal if renal replacement therapy (RRT)
does not maintain homeostasis until renal function is restored. AKI in multiorgan
failure is often fatal despite RRT.
The kidneys consist of nephrons, small independent functional units whose
glomerular part filters fluids and small molecules from the blood, and a single
tubule that reabsorbs most of the filtered molecules and excretes metabolic
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
5
waste products, producing 1-2 liters of urine each day. The number of nephrons
is determined at birth and decreases gradually with age after about 25 years [26].
Because metabolic activity also declines with age, healthy 70-year-olds can do
well with only half the original number of nephrons without adaptation. However,
low nephron count at birth
or loss of nephrons after normal aging shortens renal lifespan. Therefore, the
incidence of chronic kidney disease and kidney failure requiring RRT increases in
the elderly. AKI and CKD are linked because AKI can cause irreversible nephron
loss at any stage of life, thereby shortening kidney life [27,28]. Thus, AKI is an
important risk factor for chronic diseases, especially in the aging population.
The pathogenesis of inflammatory diseases of the renal parenchyma, such as
glomerulonephritis and vasculitis, is complex and involves almost all aspects of
the innate inflammatory system and mechanisms mediated by antibodies and
immune cells [29-31]. In this review, we focus on acute kidney injury due to
prerenal factors, because this form is the most common in developed countries,
in hospitals, and especially in critically ill patients. Much of our understanding of
the pathophysiology of prerenal acute kidney injury comes from animal studies
[32]. Studies in models of acute ischemia induced by acute renal artery occlusion
reveal the many pathways and mechanisms of organ injury likely involved. The
coagulation system is locally activated, leukocytes infiltrate the kidney, the
endothelium is damaged and adhesion molecules are expressed, cytokines are
released, toll-like receptors are induced, intrarenal vasoconstrictor pathways are
activated, and apoptosis is induced. It appears that kidney injury can trigger
organ injury elsewhere (so-called organ crosstalk) through obscure pathways,
further emphasizing the complexity of the biological response to acute kidney
injury.
Unfortunately, this ischemic model has little clinical relevance in diseases such
as sepsis. Sepsis is the most common cause of acute kidney injury in hospitalized
and intensive care patients [33]. The model also has little relevance for periods of
reduced perfusion that may occur during major surgery, as 80% renal artery
occlusion for 2 h does not cause permanent renal dysfunction. Thus, many of the
principles that physicians use to understand acute kidney injury are questionable
for patients in modern hospitals or intensive care units. The most common
causes of acute kidney injury in such patients are sepsis, major surgery
(especially open heart surgery), and acute decompensated heart failure [34,35].
The renal artery is not blocked in any of these situations. More appropriate
models are needed.
Neurohormonal mechanisms: Activation of the sympathetic system and
neurohormonal responses specific to the kidney are activated in acute kidney
injury [36]. The renin-angiotensin-aldosterone system, the renal sympathetic
system and the tubulo-glomerular feedback system are activated. Knowing these
changes led to further understanding of how people can develop acute kidney
injury. This framework suggests that in sepsis-like situations, infection leads to
nitric oxide synthase induction and nitric oxide-mediated vasodilation, which in
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
6
turn leads to arterial hypoperfusion and baroreceptor activation [37]. These
circulatory changes trigger activation of the sympathetic system, which increases
renin- angiotensin-aldosterone system (RAAS) activity and renal
vasoconstriction. At the same time, arginine vasopressin is released and
promotes water retention.
Hepatorenal syndrome (HRS) is perhaps the most widely studied form of
acute kidney injury in terms of neurohormonal changes and provides useful
mechanistic insights [38]. In this syndrome, as in experimental sepsis, acute
kidney injury occurs without histopathological renal changes and is thus
functional in nature. Severe renal vasoconstriction associated with significant
RAAS activation is a characteristic finding in patients with hepatorenal syndrome,
suggesting that neurohormonal events drive the development of the disease.
Although the mechanisms leading to such activation are debated, the central
event is thought to be a decrease in systemic blood pressure due to splanchnic
vasodilation [39]. The neurohormonal response to such vasodilation supports
systemic blood flow, but renal blood flow may be adversely affected. It is not
known whether a similar condition occurs in other diseases associated with
hypotension and systemic vasodilation (eg, inflammation and sepsis). Thus,
elevations in noradrenaline, renin, and angiotensin II may contribute to other
forms of acute kidney injury, suggesting that, at least in some settings,
neurohormonal renal vasoconstriction may underlie the loss of excretory function.
6. CLINICAL MANIFESTATIONS
AKI patients do not suffer from clinical symptoms more or less specific to the
disease. On the other hand, they may indicate symptoms of some underlying
disease (eg heart failure, sepsis, systemic vasculitis, thrombotic
microangiopathy). Urine production is reduced in 70% of AKI and can result in
fluid retention with worsening blood pressure and heart failure with pulmonary
edema [40]. Due to the reduction in the secretion of electrolytes and
endogenous/exogenous residues, the whole body is affected. The term uremia
describes such poisoning and is associated with a variety of heterogeneous
symptoms, including pruritus, neurological manifestations, nausea and vomiting,
diarrhea, anorexia and loss of appetite, cardiac arrhythmias, and insomnia
[41,42]. In addition, patients have an increased risk of infections and abnormal
bleeding complications (due to platelet dysfunction). The presence of uremia is
important because in most cases dialysis therapy becomes mandatory. Polyuria
after initial oliguria usually indicates the onset of kidney function, but can cause
significant loss of water, sodium and potassium. The latter can cause heart
rhythm abnormalities [43]. If the kidney recovery takes more than 3 months, it
means AKI is transformed to CKD.
7. LONG-TERM CONSEQUENCES OF AKI
Irreversible nephron loss, fibrosis and CKD: Depending on the severity of
AKI, few, many, or most nephrons remain irreversibly destroyed and lost,
resulting in post-AKI CKD and shortened renal lifespan [44]. Albuminuria after
AKI is a clinical indicator of CKD, even when GFR appears to have fully
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
7
recovered. The impact of AKI on renal life expectancy is most evident in older
adults, where AKI-related nephron loss increases age-related nephron loss and
often chronic kidney disease from previous injuries or chronic nephropathies,
known as AKI in CKD. Therefore, since ATN means nephron loss, the severity of
ATN determines the effect on kidney lifespan. In extreme cases, severe ATN can
lead to kidney failure and continuous need for RRT.
Hypertension and risk of cardiovascular disease: AKI survivors may have
hypertension, which may be a sign of subclinical CKD. A retrospective cohort
study showed a 22% increased risk of blood pressure >140/90 mmHg in patients
with AKI compared to those without AKI [45]. CKD after AKI is associated with
increased cardiovascular and cerebrovascular morbidity and mortality [46-48]. It
is not clear whether this increase is related to effects on the cardiovascular
system during an episode of AKI or to an increased risk of CKD after AKI.
Mortality: AKI survivors face increased post-hospital mortality. Long-term
mortality in AKI patients may also be increased. In a study of heart surgery
patients, the increase in mortality risk was independent of improvement in
kidney function at hospital discharge and did not begin until 4 to 5 years after
surgery [49]. The most common causes of death are cardiovascular disease
(28%) and cancer (28%), and the corresponding standardized mortality rates are
nearly six and eight times higher than those in the general population. Cancers
were mostly hematological or urogenital. An episode of AKI predicts the risk of
later development of kidney cancer, and an episode of AKI after partial
nephrectomy for kidney cancer increases the risk of cancer recurrence, probably
because kidney damage causes DNA damage and clonal expansion of mutated
cells during the healing phase [50,51]. Indeed, renal progenitors that leads to
tubular regeneration can become tumor cells after ischemic ATN and trigger
monoclonal lesions in the papillary renal cell adenoma-carcinoma sequence.
8. DIAGNOSIS, SCREENING AND PREVENTION
Unlike myocardial infarction and other acute organ failure, AKI does not cause
immediately alarming symptoms such as chest pain, shortness of breath,
paralysis or blindness; therefore, diagnosis requires special technical
assessments. The best overall index of kidney function is GFR, but direct
measurement of GFR is difficult. Generally, GFR is estimated using serum levels
of endogenous filtration markers such as creatinine. Several studies have shown
that small increases in serum creatinine are associated with worse outcomes in
AKI [52]. In addition, urine output is a sensitive parameter of renal function and a
biomarker of tubular damage. However, the relationship between urine output,
GFR and tubular injury is very complex.
Diagnostic and classification criteria: There is evidence to suggest that acute
and mild declines in renal function, as manifested by changes in blood chemistry
and urine output, are associated with worse outcome in AKI. In contrast to the old
term acute kidney failure, the Risk, Injury, Failure, Loss of kidney function and
End-Stage Renal Disease (RIFLE) and Acute Kidney Injury Network (AKIN)
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
8
classifications have provided updated definitions of AKI that cover the entire
spectrum of syndrome from slight increase in serum creatinine to the need for
active treatment [53]. The RIFLE and AKIN classifications have three degrees of
severity based on changes in serum creatinine or urine output, with the worse of
the two criteria being used to determine the grade. RIFLE and AKIN thus
provided a conceptual framework for the diagnosis and staging of AKI, but
further modifications were necessary to respond to the clinical complexity of AKI,
especially outside the intensive care unit (ICU) or hospital settings.
The latest KDIGO guidelines defined the diagnostic criteria for AKI. Contrary to
previous recommendations, the KDIGO criteria no longer require adequate fluid
resuscitation and exclusion of urinary tract obstruction before using the criteria.
Patients with CKD are particularly prone to AKI, as CKD is an independent risk
factor for AKI. However, the diagnosis of AKI in patients with CKD is difficult
because these patients have impaired renal function and changes in serum
creatinine levels after AKI are partially confounded with baseline renal function
levels. The KDIGO criteria use decreased urine output, but decreased urine
output is also a physiological mechanism in response to decreased fluid intake
or fluid loss that readily responds to fluid intake and usually does not indicate
tubular damage. Damaged tubules no longer respond to diuretics due to the loss
of necessary sodium transporters; thus, a single bolus of a loop diuretic that is
not followed by a significant increase in urine output, called the furosemide stress
test, indicates tubular injury [54]. In fact, the incidence of AKI is significantly
higher when both urine output and serum creatinine are abnormal, compared
with abnormal serum creatinine alone (62.1% vs. 17.7%).
9. SCREENING AND RISK ASSESSMENT WITH BIOMARKERS
About half of patients with stage 1 acute kidney injury have elevated serum
biomarkers and histological abnormalities on renal biopsy, whereas most of
those with stage 3 AKI have both [55]. Serum creatinine level and urine output
are two functional biomarkers with several limitations. The specificity of urine
production is low because several factors can affect this parameter, including
hypovolemia and the use of diuretics [56]. In contrast, serum creatinine sensitivity
in previously healthy kidneys is low because serum creatinine levels rise only
when at least 50% of functional nephrons are lost. In patients with a low baseline
GFR, small changes in kidney function can lead to a increase in serum creatinine
of 0.3 mg/dL, which defines AKI. Although novel biomarkers such as IL-18 or
kidney injury molecule 1 (KIM-1) are available and most of them have a very
good predictive value, there are limitations, including poor predictive power when
the timing of kidney injury is unknown; therefore, they are only inconsistently
applied in clinical practice.
10. PREVENTIVE MEASURES
In LMIC, prevention of volume depletion alone is thought to have a large impact
on the incidence of AKI. In addition to preventing volume depletion and
nephrotoxin exposure or overdose, new biomarkers may identify patients at high
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
9
risk of AKI. This approach can be used to stratify patient populations and
implement different interventions to prevent the development of AKI.
Implementation of the "KDIGO bundle" consisting of volume and hemodynamic
optimization, avoidance of nephrotoxic drugs and prevention of hyperglycemia in
patients with high risk of AKI identified by biomarkers can prevent AKI after
cardiac surgery [57-59]. In a Quality Initiative program, implementation of
supportive measures in biomarker-positive patients reduced the incidence of
moderate and severe AKI after cardiac surgery and the incidence of AKI in
abdominal surgery patients [60]. Despite these data, only about 5% of high-risk
patients receive these supportive measures.
11. MANAGEMENT
Management of AKI is often suboptimal. The first step in managing AKI is to
identify its causes, such as prerenal causes (hypovolemia), intrinsic renal
causes, or postrenal causes (outflow obstruction). Further treatment is influenced
by the clinical situation, location and history of the patient.
Volume status: Volume depletion may cause impairment of kidney function, but
does not damage the kidney by itself unless it is severe and persistent. However,
volume depletion can contribute to several causes of AKI, and fluid resuscitation
is the cornerstone of treatment. Patients with AKI in the community may become
dehydrated, as may hospitalized patients receiving diuretics or dehydrating from
injuries or drainage. A hospitalized patient should never become severely
dehydrated; however, correcting dehydration by giving inappropriate fluids
without properly assessing patients with AKI can lead to fluid overload, which can
have significant detrimental effects [61]. Patients requiring intravenous fluid
resuscitation should be under direct physician supervision and treatment will
benefit from guidelines for hemodynamic monitoring. In addition, the sudden
need for fluid resuscitation requires measures to determine its cause (for
example, occult bleeding or sepsis). It should be noted that patients can develop
oliguria from AKI and then become fluid overloaded with injudicious
administration of intravenous fluids in addition to fluids from medications and
nutritional support. Importantly, fluid overload has been identified as a major
cause of AKI, as venous congestion can compromise circulation and cause direct
damage to the renal parenchyma.
Hemodynamic control: Management of blood pressure and cardiac function in
the setting of AKI (eg, septic shock or cardiac surgery) is complex and involves
context-specific considerations, depending on the type of circulatory shock the
patient is experiencing. Under normal conditions, most organs, including the
kidneys, are adequately perfused with a mean arterial pressure (MAP) of 65
mmHg or above [62]. Studies examining whether MAP targets should be used in
ICU patients have produced mixed results. Patients with severe (and perhaps
poorly controlled) hypertension may benefit from a higher MAP in shock, but a
fixed target cannot be recommended. Some findings suggest individualized blood
pressure control by adjusting MAP targets based on the patient's typical blood
pressure. Also, patients with elevated venous pressure (for example due to right-
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
10
sided heart failure) may not achieve adequate renal perfusion pressure at a MAP
of 65 mmHg. Studies using functional hemodynamic monitoring to guide
hemodynamic management have shown promise in both cardiac surgery and
sepsis. Noradrenaline is the first choice as a vasopressor in vasodilatory shock
[63,64]. The other agents are generally reserved for shock requiring therapy or
emergency situations, and none are generally more "kidney friendly."
Nephrotoxic drugs and substances: The risk of AKI increases with the number
of nephrotoxic drugs used, and all potentially nephrotoxic agents that can be
discontinued should be stopped [65,66]. If possible, careful monitoring of drug
concentration is also mandatory (for example, in the case of vancomycin). The
use of arteriolar contrast agents should be limited to situations where the
therapeutic benefit outweighs the risk, and they should be used in the smallest
amounts possible.
Stepwise management of AKI: The KDIGO AKI Guidelines emphasize the
importance of AKI staging as a management guide. Prognosis is strongly
correlated with the peak phase of AKI and the duration of AKI (transient versus
persistent), so the urgency and invasiveness of diagnostic and therapeutic
measures increase with the phase of AKI [67]. However, the stages of AKI must
be interpreted in the context of baseline kidney function. For patients with
preexisting normal renal function and stage 1 AKI, treatment primarily involves
prompt identification of the likely cause of AKI and avoidance of secondary
insults. Depending on baseline GFR, drug dosage adjustments usually become
clinically relevant in stage 2 AKI. In stage 3 AKI, symptoms can be caused by a
disturbance in the acid-base balance and electrolyte levels, as well as the
accumulation of uremic toxins [68]. For example, patients may develop
tachypnea not only due to fluid overload, but also due to metabolic acidosis.
Acidosis removes potassium from cells, further exacerbating hyperkalemia. Even
relatively moderate uremia can impair platelet function and increase the risk of
bleeding. If treatment is ineffective or if the disorders are life-threatening, RRT is
necessary.
In all stages of AKI, it is recommended to stop all potentially nephrotoxic drugs as
soon as possible, because all drugs cause or contribute to AKI in most cases and
are probably the most modifiable risk factors for AKI [69,70]. Volume control and
hemodynamic monitoring are also required in all stages of AKI. Avoidance of
hyperglycemia is important because filtered glucose increases tubular
reabsorption workload and oxidative stress, a process that sensitizes renal
tubules to damage. An important yet unresolved question so far is whether the
various forms of acute kidney injury can be treated with approaches targeting the
underlying causes. This strategy is possible in conditions such as obstructive
uropathy or atypical hemolytic uremic syndrome, but other forms of AKI often
have an unknown etiology. Current knowledge indicates that risk factors and risk
modifiers such as medications, contrast agents, low cardiac output and overload
should be reduced or eliminated. Even if the cause of an AKI episode is
identifiable, this awareness may come too late to prevent the eventual common
pathway of tubular toxicity, ischemia, and inflammation.
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
11
Management of the trajectory of AKI: Most patients with AKI who receive
medical care and whose injury resolves on its own (such as surgery) or whose
cause is resolved (such as stopping a nephrotoxic drug or treating an infection)
begin to improve kidney function usually within 24-48 hours [71]. However, in 25–
35% of patients, AKI persists for ≥72 hours. These patients have significantly
worse outcomes. Thus, persistent AKI should prompt the clinician to reconsider
their working diagnosis regarding the cause of AKI. For example, in a patient
who develops AKI after cardiac surgery, the volume status, hemodynamics, and
medication list should be carefully reviewed and any problems corrected [72].
Recurrence of AKI is common, especially in ICU patients [73]. Whether
recurrence is due to multiple insults to kidney or recurrent renal failure due to
damage from a single injury varies. It is best to assume that the recurrence may
be due to a new cause that needs to be identified. Recovery after AKI is best
assessed after hospital discharge, but follow-up of these patients has historically
been poor. Patients may be discharged with unstable renal function and are
therefore at greater risk of drug-related adverse events, since most receive drugs
that are excreted by the kidneys [74,75]. Common reasons for hospital
readmission are both treatment failure due to under dosing in patients with
improving renal function and toxic effects due to overdose in patients with
declining renal function. Therefore, patients should be referred to a nephrologist
for evaluation of kidney function immediately after discharge from the hospital.
Finally, even patients who appear to have fully recovered from AKI may be at
increased risk of developing kidney damage over an unknown period of time.
Renal replacement therapy: The various aspects of RRT have advanced
significantly over the years, making extracorporeal therapy safer and easier to
implement. The time and choice of initiation of therapy remain controversial,
often due to the heterogeneity of the populations studied. In a patient-centered
evaluation based on the principles of precision medicine, life- threatening
conditions are not the only indicators of the need to RRT but the prevention of
clinical complications should also be considered.
Peritoneal dialysis has been used for many years and is still used in areas where
access to more advanced techniques is limited or unavailable [76,77]. In the
absence of evidence to support specific techniques, the choice of method should
be based on pathophysiological considerations. In unstable and critically ill
patients, continuous RRT is often preferred [78]. After the patient is discharged
from the intensive care unit, intermittent methods such as continuous low-dose
dialysis or daily intermittent hemodialysis can be safely used. Continuous veno-
venous hemofiltration, continuous veno-venous hemodialysis, or continuous
veno-venous hemodiafiltration are used according to center experience and staff
training rather than evidence-based technique differences. A conceptual model of
sequential extracorporeal therapy with early removal of endotoxins by polymyxin-
B hemoperfusion followed by removal of cytokines and proinflammatory and anti-
inflammatory mediators by absorbent devices has been proposed for patients
with sepsis-related AKI [79,80]. The best time to initiate RRT in critically ill
patients remains controversial, in part because of conflicting renal outcomes in
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
12
relevant studies. In fact, best practices for RRT can vary in many ways,
especially for certain populations.
12. QUALITY OF LIFE
There is no information on the quality of life of AKI patients in the intensive care
unit, where many patients receive medication while on respiratory support. AKI-
specific aspects would also be difficult to assess because the diseases are often
complex. Existing research focuses on the long-term effects of AKI on
health-related quality of life
(HRQL) and functional status in critically ill survivors [81-83]. Most studies
consistently show that survivors of AKI have significantly reduced HRQL
compared to survivors of critical illness without AKI or the general population.
HRQL was lower in patients who required intensive care for severe AKI than in
those who did not. Among AKI survivors, 20–40% developed a new disability in
at least one activity of daily living, and only 28–69% of AKI survivors with
preexisting severe disease were able to return to work [84]. After one year of AKI
requiring dialysis, 81.8% of survivors would agree to readmission to the ICU if
necessary, but after four years that number dropped to 71.4.
13. CURRENT AND FUTURE PERSPECTIVE
To date improvements in the field of diagnosis and treatment of acute kidney
injury remain unsatisfactory. From a diagnostic point of view, the introduction of a
marker of kidney damage in clinical practice remains a priority compared to the
current approach based on kidney function. Measurement of serum creatinine
does not allow early diagnosis of AKI, which is essential to improve patient
outcomes. Creatinine assessment also does not elucidate the extent to which
subclinical episodes of AKI contribute to shortened renal lifespan and CKD.
Recommendations from the 23rd ADQI Consensus Conference suggest that
combining definitions of AKI based on serum creatinine and urinary excretion
with biomarkers of kidney injury would improve the accuracy of predicting the
course of AKI.
Marlies et al. [85] Future studies have yet to show the extent to which new
biomarkers can help improve short- and long-term outcomes.
14. CONCLUSION
Due to a variety of factors, AKI often has a worse prognosis in critically ill
patients. Chronic renal disease and morbidity from cardiovascular disease are
long-term effects of AKI and AKD. Early AKI detection and prevention are so
crucial.
COMPETING INTERESTS
Authors have declared that no competing interests exist.
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
13
REFERENCES
1. Renske Wiersema, Ruben J Eck, Mikko Haapio, Jacqueline Koeze, Meri
Poukkanen, Frederik Keus, Iwan C C van der Horst, et al. Burden of acute
kidney injury and 90-day mortality in critically ill patients. BMC Nephrol.
2019;21(1):1.
2. James Case, Supriya Khan, Raeesa Khalid, Akram Khan. Epidemiology of
acute kidney injury in the intensive care unit. Crit Care Res Pract.
2013;2013:479730.
3. Abshiro H Mayow, Fatima Ahmad, Muhammad Sohaib Afzal, Muhammad
Usama Khokhar, Daneyal Rafique, Sai Krishna Vallamchetla, et al. A
Systematic Review and Meta-Analysis of Independent Predictors for
Acute Respiratory Distress Syndrome in Patients Presenting With Sepsis.
Cureus. 2023;15(4):e37055.
4. Z Ricci, D Cruz, C Ronco. The RIFLE criteria and mortality in acute kidney
injury: a systematic review. Kidney Int. 2008;73(5):538-46.
5. Sreekant Avula, Sandesh Dewan, Bushra Firdous Shaik, Atul Bali.
Diabetic Kidney Disease. Int Clinc Med Case Rep Jour. 2023;2(11):1-4.
6. Mark E Thomas, Caroline Blaine, Anne Dawnay, Mark A J Devonald,
Saoussen Ftouh, Chris Laing, et al. The definition of acute kidney injury
and its use in practice. Kidney Int. 2015;87(1):62-73.
7. Khwaja Arif. KDIGO clinical practice guidelines for acute kidney injury.
Nephron Clin Pract. 2012;120(4): c179-c184.
8. Palleti, Sujith Kumar, Sreekanth Avula, and Sandesh Dewan. Kidney: A
Review on End Stage Renal Disease, Dialysis and Transplant. Int Clinc
Med Case Rep Jour. 2023;2 (10): 1-8.
9. Venkataraman Ramesh, John A. Kellum. Defining acute renal failure: the
RIFLE criteria. J Intensive Care Med. 2007;22(4):187-93.
10. Hanif, Muhammad O, Atul Bali, Kamleshun Ramphul. Acute renal tubular
necrosis. StatPearls [Internet]. StatPearls Publishing. 2022.
11. Sawhney Simon, Simon D. Fraser. Epidemiology of AKI: utilizing large
databases to determine the burden of AKI. Adv Chronic Kidney Dis.
2017;24(4):194-204.
12. Palleti, Sujith Kumar, Sreekant Avula, Roopa Naik. Chronic Kidney
Disease. Int Clinc Med Case Rep Jour. 2023;2(11):1-15.
13. Eric A J Hoste, Sean M Bagshaw, Rinaldo Bellomo, Cynthia M Cely, Roos
Colman, Dinna N Cruz, et al. Epidemiology of acute kidney injury in
critically ill patients: the multinational AKI-EPI study. Intensive Care Med.
2015;41(8):1411-23.
14. Barkhane Zineb, et al. Independent Predictors of Mortality in Systemic
Sclerosis-Associated Pulmonary Arterial Hypertension: A Systematic
Review and Meta-Analysis. Cureus. 2023;15(5).
15. Sandra L Kane-Gill, Florentina E Sileanu, Raghavan Murugan, Gregory S
Trietley, Steven M Handler, John A Kellum. Risk factors for acute kidney
injury in older adults with critical illness: a retrospective cohort study. Am J
Kidney Dis. 2015;65(6):860-869.
16. Abdulmaleek Idanesimhe Sado, Muhammad S Afzal, Lavanya
Kannekanti, Hrushikesh Reddy Pamreddy, Jorge Pimentel Campillo,
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
14
Vaishnavi Kandukuri, et al. A Meta-Analysis on Predictors of Mortality
Among Patients Hospitalized for Acute Exacerbation of Asthma. Cureus.
2023;15(2):e35225.
17. Sasa Nie, Li Tang, Weiguang Zhang, Zhe Feng, Xiangmei Chen. Are
there modifiable risk factors to improve AKI?. Biomed Res Int.
2017;2017:5605634.
18. Rakesh Pilla, Sujith Kumar Palleti, Renuka Rayana, Satish Reddy Skss,
Aminah Abdul Razzack, Sruti Kalla. Glycated haemoglobin (HbA1c)
variations in nondiabetics with nutritional anemia. Cureus.
2020;12(11):e11479.
19. Sai Kiran Kondabolu, Yasitha Kakarlapudi, Haider Malik, Hamza Malik,
Saima Khan, Praveen Kumar Komminni, et al. Beneficial Impacts of
Fenoldopam on Patients with or at Risk for Acute Renal Failure and
Undergoing Surgery: A Meta-Analysis of Randomized Clinical Trials.
Cureus. 2023;15(2):e34584.
20. Rodrigo Cartin-Ceba, Markos Kashiouris, Maria Plataki, Daryl J Kor,
Ognjen Gajic, Edward T Casey. Risk factors for development of acute
kidney injury in critically ill patients: a systematic review and meta-
analysis of observational studies. Crit Care Res Pract. 2012;2012:691013.
21. Robin Rauniyar, Rahul Rauniyar, Ankita Agrawal, Shikha Yadav, Sreekant
Avula. Severe late‐onset neutropenia induced by ocrelizumab in a multiple
sclerosis patient: A case report. Clin Case Rep. 2022;10(1):e05299.
22. Fareeha Naseer Syed, Nadia Kashif, Mubashir Razzaq Khan, Fayaz
Ahmed Memon, Muhammad Zubair Jabbar, Sujith Kumar Palleti. Hepatitis
C Virus Infection in Patients with Beta Thalassemia after Multiple
Transfusions at a Tertiary Care Hospital. Pakistan Journal of Medical &
Health Sciences. 2023;17(1):363- 363.
23. Vaillant Angel A. Justiz, Roopa Naik. HIV-1 associated opportunistic
infections. StatPearls [Internet]. StatPearls Publishing. 2021.
24. Matthieu Legrand, Samira Bell, Lui Forni, Michael Joannidis, Jay L
Koyner, Kathleen Liu, et al. Pathophysiology of COVID-19-associated
acute kidney injury. Nat Rev Nephrol. 2021;17(11):751-764.
25. Benaicha Karima, et al. Efficacy of Molnupiravir for the Treatment of
Mild or Moderate COVID-19 in Adults: A Meta-Analysis. Cureus.
2023;15(5).
26. Lindeman Robert D. Overview: renal physiology and pathophysiology of
aging. Am J Kidney Dis. 1990;16(4):275-282.
27. Belayev Linda Y, Paul M. Palevsky. The link between AKI and CKD. Curr
Opin Nephrol Hypertens. 2014;23(2):149.
28. Bali, Atul, et al. “: Atul Bali, Sandesh Dewan, Roopa Naik, Maheshwari
Kata, Sreekant Avula. Continuous Renal Replacement Therapy.”
International Clinical and Medical Case Reports Journal Review Article.
2023;2(12):1-13.
29. Jennette J. Charles. Rapidly progressive crescentic glomerulonephritis.
Kidney Int. 2023;63(3):1164-1177.
30. Palleti, Sujith K, et al. Sjögren’s Syndrome Overlap with Lupus Nephritis:
A Case Report and Literature Review of a Rare Entity. Cureus.
2023;15(5).
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
15
31. Palleti Sujith K. et al. Subarachnoid Hemorrhage in Autoimmune
Vasculitis: A Rare Presentation of Systemic Lupus Erythematosus-
Antineutrophil Cytoplasmic Autoantibody-Associated Vasculitis Overlap
Syndrome. Cureus. 2023;15(5).
32. P Meneton, I Ichikawa, T Inagami, J Schnermann. Renal physiology of the
mouse. Am J Physiol Renal Physiol . 2000;278(3):F339-51.
33. Zarjou Abolfazl, Anupam Agarwal. Sepsis and acute kidney injury. J Am
Soc Nephrol. 2011;22(6):999- 1006.
34. Singbartl, Kai, John A. Kellum. AKI in the ICU: definition, epidemiology,
risk stratification, and outcomes. Kidney Int. 2012;81(9):819-25.
35. Muhammad Ali Sameer, Bilal Aziz Malik, Muhammad Obaid Ullah
Choudry, Muhammad Shoaib Anwar, Muhammad A Nadeem, Fizza
Mahmood, et al. Comparison of Coronary Artery Bypass Grafting
Combined With Mitral Valve Repair Versus Coronary Artery Bypass
Grafting Alone in Patients With Moderate Ischemic Mitral Regurgitation: A
Meta-Analysis. Cureus. 2023;15(4):e37238.
36. A Sigurdsson, P Held, K Swedberg. Short-and long-term neurohormonal
activation following acute myocardial infarction. Am Heart J.
1993;126(5):1068-76.
37. Vallon Volker. Tubuloglomerular feedback and the control of glomerular
filtration rate. News Physiol Sci. 2003;18:169-74.
38. Pere Ginès , Elsa Solà, Paolo Angeli , Florence Wong 8, Mitra K Nadim,
Patrick S Kamath. Hepatorenal syndrome. Nature Rev Dis Primers.
2018;4(1):23.
39. Hani M Wadei, Martin L Mai, Nasimul Ahsan, Thomas A Gonwa.
Hepatorenal syndrome: pathophysiology and management. Clin J Am Soc
Nephrol. 2006;1(5):1066-79.
40. John R Prowle, Yan-Lun Liu, Elisa Licari, Sean M Bagshaw, Moritoki Egi,
Michael Haase, et al. Oliguria as predictive biomarker of acute kidney
injury in critically ill patients. Critical Care. 2011;15(4):1-10.
41. Timothy W Meyer 1, Thomas H Hostetter. Uremia. N Engl J Med.
2007;357(13):1316-25.
42. Atul Bali, John Kevin Hix, Peter Kouides. Safe and effective use of chronic
transdermal estradiol for life- threatening uremic bleeding in a patient with
coronary artery disease. Nephron Extra. 2014;4(2):134-137.
43. Yuki Sato, Masahiro Takahashi, Motoko Yanagita. Study on Recognition
of Heart Arrhythmias by Deep Learning Approach of the ECG Signals
using Edge Devices.
44. Sato, Yuki, Masahiro Takahashi, and Motoko Yanagita. Pathophysiology
of AKI to CKD progression. Semin Nephrol. 2020;40(2):206-215.
45. Ayodele Odutayo, Christopher X Wong, Michael Farkou, Douglas G
Altman, Sally Hopewell, Connor A Emdin, et al. AKI and long-term risk for
cardiovascular events and mortality. J Am Soc Nephrol. 2017;28(1):377-
387.
46. Matthew T James, William A Ghali, Merril L Knudtson, Pietro Ravani,
Marcello Tonelli, Peter Faris, et al. Associations between acute kidney
injury and cardiovascular and renal outcomes after coronary angiography.
Circulation. 2011;123(4):409-416.
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
16
47. Karthik Gonuguntla, Komal Ejaz, Chaitanya Rojulpote, Ayesha Shaik,
Abhijit Bhattaru, Tapan Buch, et al. A nationwide cohort analysis to
determine the prevalence of sinus node dysfunction and rates of
pacemaker implantation in systemic lupus erythematosus. Am J
Cardiovasc Dis. 2021;11(4):478-483.
48. Karedath Jithin, et al. Comparison of Effectiveness and Safety of Low-
Dose Versus Standard-Dose Intravenous Recombinant Tissue
Plasminogen Activator in Patients with Acute Ischemic Stroke: A Meta-
Analysis. Cureus. 2023;15(2): e35571.
49. Sophie Perinel, François Vincent, Alexandre Lautrette, Jean Dellamonica,
Christophe Mariat, Fabrice Zeni, et al. Transient and persistent acute
kidney injury and the risk of hospital mortality in critically ill patients:
results of a multicenter cohort study. Crit Care Med. 2015;43(8):e269-75.
50. Marianne Schmid, Nandita Krishna, Praful Ravi, Christian P Meyer,
Andreas Becker, Deepansh Dalela, et al. Trends of acute kidney injury
after radical or partial nephrectomy for renal cell carcinoma. Urologic
Oncology: Seminars and Original Investigations. Urol Oncol.
2016;34(7):293.e1-293.e10.
51. Kar, Saikat, et al. A two-center, retrospective cohort trial of a tumor
necrosis factor inhibitor coupled with intravenous immunoglobulin and
heparin for the treatment of recurrent spontaneous abortion.
52. Rajit K Basu, Anne Andrews, Catherine Krawczeski, Peter Manning,
Derek S Wheeler, Stuart L Goldstein, et al. Acute kidney injury based on
corrected serum creatinine is associated with increased morbidity in
children following the arterial switch operation. Pediatr Crit Care Med.
2013;14(5):e218-24.
53. José António Lopes, Sofia Jorge. The RIFLE and AKIN classifications for
acute kidney injury: a critical and comprehensive review. Clin Kidney J.
2013;6(1):8-14.
54. Jay L Koyner, Danielle L Davison, Ermira Brasha-Mitchell, Divya M
Chalikonda, John M Arthur, Andrew D Shaw, et al. Furosemide stress test
and biomarkers for the prediction of AKI severity. J Am Soc Nephrol.
2015;26(8):2023-31.
55. Edelstein, Charles L. Biomarkers in acute kidney injury. Biomarkers of
Kidney Disease. 2017;241-315.
56. John A Kellum, Florentina E Sileanu, Raghavan Murugan, Nicole Lucko,
Andrew D Shaw, Gilles Clermont. Classifying AKI by urine output versus
serum creatinine level. J Am Soc Nephrol. 2015;26(9):2231-8.
57. Mira Küllmar, Alexander Zarbock, Daniel T Engelman, Subhasis
Chatterjee, Nana-Maria Wagner. Prevention of acute kidney injury. Critical
Care Clinics. 2020;36(4):691-704.
58. Raza A Khan, Nidhi Patel, Atunde Folajimi, Bansari Raveena Bai, Vrushak
Patel, Praveen Kumar Komminni, et al. Comparison of the Efficacy and
Safety of Metformin-Based Combination Therapy Versus Metformin Alone
in Children and Adolescents with Type 2 Diabetes Mellitus: A Meta-
Analysis. Cureus. 2023;15(2):e35014.
59. Eyth Emily, Roopa Naik. Hemoglobin A1c. StatPearls [Internet]. StatPearls
Publishing. 2022.
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
17
60. Mitra K Nadim, Lui G Forni, Azra Bihorac, Charles Hobson, Jay L Koyner,
Andrew Shaw, et al. Cardiac and vascular surgery–associated acute
kidney injury: the 20th international consensus conference of the ADQI
(acute disease quality initiative) group. J Am Heart Assoc.
2018;7(11):e008834.
61. Rolando Claure-Del Granado, Ravindra L Mehta. Fluid overload
in the ICU: evaluation and management. BMC Nephrol. 2016;17(1)
:109.
62. Julie Badin, Thierry Boulain, Stephan Ehrmann, Marie Skarzynski, Anne
Bretagnol, Jennifer Buret, et al. Relation between mean arterial pressure
and renal function in the early phase of shock: a prospective, explorative
cohort study. Crit Care. 2011;15(3):R135.
63. Gunnar Gamper, Christof Havel, Jasmin Arrich, Heidrun Losert, Nathan
Leon Pace, Marcus Müllner, et al. Vasopressors for hypotensive shock.
Cochrane Database Syst Rev. 2016;2(2):CD003709.
64. Zaid Munir, Muhammad Akash, Fnu Jaiprada, Bilal Abu Tarboush, Osama
Ijaz, Anan Bseiso, et al. Evaluation of the Effects of Extracorporeal
Shockwave Therapy in Patients with Peripheral Arterial Disease: A Meta-
Analysis of Randomized Control Trials. Cureus. 2023;15(2):e34729..
65. Naughton, Cynthia A. Drug-induced nephrotoxicity. American family
physician. 20085;78(6):743-750.
66. Sujith K Palleti, Anuradha Wadhwa, Abdul H Rasheed. The First Case of
Ceftazidime-Induced Antibiomania: A Case Report and Literature Review.
Cureus. 2022;14(12):e32904.
67. Patrick T Murray, Prasad Devarajan, Andrew S Levey, Kai U Eckardt,
Joseph V Bonventre, Raul Lombardi, et al. A framework and key research
questions in AKI diagnosis and staging in different environments. Clin J
Am Soc Nephrol. 2008;3(3):864-8.
68. Alexandre Braga Libório, Tacyano Tavares Leite, Fernanda Macedo de
Oliveira Neves, Flávio Teles, Candice Torres de Melo Bezerra. AKI
complications in critically ill patients: association with mortality rates and
RRT. Clin J Am Soc Nephrol. 2015;10(1):21-8.
69. E J Filippone, W K Kraft, J L Farber. The nephrotoxicity of vancomycin.
Clin Pharmacol Ther. 2017;102(3):459-469.
70. Palleti, Sujith K., Anuradha Wadhwa, Abdul H. Rasheed. Ceftazidime-
induced Antibiomania: A Rare Case Study and Review of Literature.
Research Highlights in Disease and Health Research Vol. 2023;6:
56-61.
71. Harkesh Arora, Sathishkumar Ramalingam, Rajvee Sanghavi, Adedayo
Balogun, Maheshwari Muruganandam, et al. Disseminated Nocardia
infection-A rare presentation. Clin Case Rep. 2021;9(8):e04673.
72. Robert H Thiele, James M Isbell, Mitchell H Rosner. AKI associated with
cardiac surgery. Clin J Am Soc Nephrol. 2015;10(3):500-14.
73. Edward D Siew, Sharidan K Parr, Khaled Abdel-Kader, Svetlana K Eden,
Josh F Peterson, Nisha Bansal, et al. Predictors of recurrent AKI. J Am
Soc Nephrol. 2016;27(4):1190-200.
74. Fatemeh Ghane Shahrbaf, Farahnak Assadi. Drug-induced renal
disorders. J Renal Inj Prev. 2015;4(3):57- 60.
Advances in Renal Diseases and Dialysis
Acute Kidney Injury
18
75. Matthew A Gorris, Harkesh Arora, David C Lieb, Joseph A Aloi. A word of
caution when prescribing high-dose vitamin D. Am J Med.
2017;130(4):e129-e130.
76. Daniela Ponce, Marina Berbel Buffarah, Cassiana Goes, André Balbi.
Peritoneal dialysis in acute kidney injury: trends in the outcome across
time periods. PLoS One. 2015;10(5):e0126436.
77. Sujith K Palleti, Santhoshi R Bavi, Margaret Fitzpatrick, Anuradha
Wadhwa. First case report of Sphingobium lactosutens as a human
pathogen causing peritoneal dialysis-related peritonitis. Cureus.
2022;14(7): e27293.
78. Antoine G Schneider, Rinaldo Bellomo, Sean M Bagshaw, Neil J
Glassford, Serigne Lo, Min Jun, et al. Choice of renal replacement therapy
modality and dialysis dependence after acute kidney injury: a systematic
review and meta-analysis. Intensive Care Med. 2013;39(6):987-97.
79. Romagnoli, Stefano, Zaccaria Ricci, and Claudio Ronco. CRRT for sepsis-
induced acute kidney injury. Curr Opin Crit Care. 2018;24(6):483-492.
80. Sathishkumar Ramalingam, Harkesh Arora, Susan Lewis, Kulothungan
Gunasekaran, Maheswari Muruganandam, Sivakumar Nagaraju, et al.
COVID-19 vaccine-induced cellulitis and myositis. Cleve Clin J Med.
2021;88(12):648-650.
81. Pierre-Marc Villeneuve, Edward G Clark, Lindsey Sikora, Manish M Sood,
Sean M Bagshaw. Health- related quality-of-life among survivors of acute
kidney injury in the intensive care unit: a systematic review. Intensive
care medicine. Intensive Care Med. 2016;42(2):137-46.
82. Palleti SK. Fellowship in the United States as an exceptionally qualified
applicant. Postgraduate Medical Journal. 2023.
83. Sean M Bagshaw, Michael Darmon, Marlies Ostermann, Fredric O
Finkelstein, Ron Wald, Ashita J Tolwani, et al. Current state of the art for
renal replacement therapy in critically ill patients with acute kidney injury.
Intensive Care Med. 2017;43:841-854.
84. Zhang Ting. Hospital readmissions after acute kidney injury: A systematic
review and meta-analysis. Int J Clin Exp Med. 2016;9(7):13456-13465.
85. Marlies Ostermann, Alexander Zarbock, Stuart Goldstein, Kianoush
Kashani, Etienne Macedo, Raghavan Murugan, et al. Recommendations
on acute kidney injury biomarkers from the acute disease quality initiative
consensus conference: a consensus statement. JAMA Netw Open.
2020;3(10):e2019209.
___________________________________________________________________________________
© Copyright (2023): Author(s). The licensee is the publisher (B P International).
DISCLAIMER
This chapter is an extended version of the article published by the same author(s) in the following journal.
Annal of Clin Med & Med Res, 1(1): 1-17, 2023. DOI: https://doi.org/10.5281/zenodo.8076523
________________________________________________________________________
a Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA,
USA.
b Department of Internal Medicine/Nephrology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre,
Pennsylvania, USA.
c Department of Internal Medicine, University Hospital and Clinics, Lafayette, USA.
d Department of Internal Medicine, MNR Medical College and Hospital, Telangana, India.
e Department of Internal Medicine, Lovelace Medical Center, Albuquerque, New Mexico, USA.
f Department of Internal Medicine/Nephrology, University of Alabama at Birmingham, Birmingham,
Alabama, USA.
g Department of Internal Medicine/Gastroenterology, Brooklyn Hospital Center, Brooklyn, New York,
USA.
*Corresponding author: E-mail: drmaheshwarikata@gmail.com;
Chapter 2
Print ISBN: 978-81-19491-84-1, eBook ISBN: 978-81-19491-85-8
Hepatorenal Syndrome
Roopa Naik a, Atul Bali b, Bushra Firdous Shaik c,
Maheshwari Kata d*, Harkesh Arora e,
Deepak Chandramohan f and Rajeshwari Ramachandran g
DOI: 10.9734/bpi/mono/978-81-19491-84-1/CH2
ABSTRACT
Hepatorenal syndrome is a serious complication of end-stage cirrhosis
characterized by increased splanchnic blood flow, a hyperdynamic state, reduced
central volume, activation of vasoconstrictor systems, and extreme renal
vasoconstriction leading to a decrease in GFR. In recent years, the role of
systemic inflammation, a key feature of cirrhosis, in the development of
hepatorenal syndrome has been emphasized. The mechanisms by which
systemic inflammation induces changes in renal blood flow during hepatorenal
syndrome remain to be elucidated. Early diagnosis is central to treatment, and
recent changes in the definition of hepatorenal syndrome help to identify patients
at an earlier stage. Vasoconstrictor agents such as terlipressin and albumin are
the first-line treatment options. Several controlled studies have shown that
terlipressin is effective in reversing hepatorenal syndrome and may improve
short-term survival. Not all patients respond, and even those who do have a very
high early mortality rate without liver transplantation. Liver transplantation is the
only definitive treatment for hepatorenal syndrome. In the long term, transplant
patients with hepatorenal syndrome usually have a lower GFR than patients
without hepatorenal syndrome. Differentiating hepatorenal syndrome from acute
tubular necrosis (ATN) is often a difficult but important step, as vasoconstrictor
drugs are not warranted in the treatment of ATN. Hepatorenal syndrome and
ATN can be considered a continuum rather than separate entities. Emerging
biomarkers may help distinguish between these two conditions and provide
prognostic information about renal recovery after liver transplantation and
potentially influence the decision for simultaneous liver and kidney
transplantation.
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
20
Keywords: Hepatorenal syndrome; liver transplantation; biomarkers of HRS;
acute kidney injury; vasopressors.
1. INTRODUCTION
Advanced liver cirrhosis is a condition characterized by impaired liver function,
portal hypertension, increased splanchnic circulation, a hyperdynamic state with
increased cardiac output, systemic vasodilatation, reduced central blood volume,
and a systemic inflammatory response. Acute kidney injury (AKI) is one of the
most important complications of cirrhosis, occurring in up to 50% of hospitalized
patients and associated with higher mortality that increases with the severity of
AKI [1]. Hepatorenal syndrome (HRS) is a type of AKI that occurs in advanced
cirrhotic patients and is mainly characterized by impaired renal perfusion
unresponsive to volume expansion. Hepatorenal syndrome is associated with
significant use of health care resources, with estimated direct medical costs of
approximately $4 billion annually in the United States [2]. Advancements in
definitions helped diagnose hepatorenal syndrome in earlier stages of liver
cirrhosis. Recent advances in the understanding of the pathophysiology of
hepatorenal syndrome indicate that, in addition to systemic and splanchnic
circulatory changes, systemic inflammation and renal circulatory changes are
also involved [3]. Although treatment of hepatorenal syndrome in combination
with vasoconstrictors and albumin has improved outcomes, the prognosis is poor
without liver transplantation. This recent literature review focuses on providing an
updated version of the HRS, with particular attention to some important changes.
2. DEFINITION OF HRS AND ACUTE KIDNEY INJURY IN CIRRHOSIS
The definition of cirrhotic AKI has changed significantly in recent years [4]. A
common theme among definitions is the use of relative changes in serum
creatinine rather than absolute cutoffs (eg, >1.5 mg/dL) and the identification of
patients at greatest risk of short-term and long-term mortality based on these
stages within each criterion. In 2012, the Acute Dialysis Quality Initiative (ADQI)
recommended adapting the AKI Network serum creatinine criteria for defining
AKI [5]. These criteria were independent of the cause of AKI, and as such,
hepatorenal syndrome type 1 was classified as a specific type of AKI and
hepatorenal syndrome type 2 was classified as a form of CKD. The International
Ascites Club (ICA) further modified the definition of AKI based on serum
creatinine criteria developed by Kidney Disease Improving Global Outcomes
(KDIGO) using baseline serum creatinine in the last 3 months [6]. Although
oliguria is not included in the current definition of AKI in cirrhotic patients, urine
output has been found to be a sensitive and early marker of AKI in critically ill
cirrhotic patients and is associated with adverse events. Therefore, despite an
increase in serum creatinine in cirrhotic patients, a decrease in urine output or
development of anuria should be considered as AKI in cirrhosis until proven
otherwise.
Changes in the definition of AKI in cirrhotic patients led to changes in the
definition of hepatorenal syndrome, so that the serum creatinine cut-off was
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
21
removed and replaced by the ICA AKI criteria, which allows earlier diagnosis and
treatment of patients with hepatorenal syndrome. An important limitation of the
criteria for hepatorenal syndrome is that it does not allow patients with liver
disease to have other forms of acute or chronic kidney disease at the same
time, such as diabetic nephropathy or glomerular disease [7-9]. However,
patients with concomitant renal disease may develop "hepatorenal physiology".
Thus, ADQI proposed the term "hepatorenal diseases" to describe all patients
with advanced cirrhosis and concomitant renal failure, taking into account the
appropriate classification and management of these patients, retaining the term
hepatorenal syndrome.
3. PATHOPHYSIOLOGY OF HEPATORENAL SYNDROME
Cirrhosis is characterized by a decrease in systemic vascular resistance due to
splanchnic artery vasodilation [10]. In the early stages of the disease, splanchnic
vasodilatation is moderate and decreased systemic vascular resistance is
balanced by increased cardiac output. In advanced stages, vasodilatation is
more pronounced because the synthesis of vasodilatory factors increases and
cannot be balanced by an increase in cardiac output. The result is effective
arterial hypovolemia due to the difference in intravascular blood volume and the
greatly expanded arterial circulation [11,12]. Cirrhotic cardiomyopathy is a
disease entity that combines diastolic dysfunction, a dull increase in cardiac
output after stimulation and various electromechanical abnormalities [13,14]. The
inflammatory response during cirrhosis, where circulating TNF-α is elevated,
may contribute to impaired cardiac response. In advanced cirrhosis with ascites,
reduced cardiac output appears to precede hepatorenal syndrome. Decreased
cardiac output can reduce blood flow to the kidneys. Finally, changes in renal
hemodynamics and autoregulation of renal blood flow contribute to a decrease in
GFR.
To maintain arterial pressure, systemic vasoconstrictor systems (renin-
angiotensin-aldosterone system, sympathetic nervous system and arginine
vasopressin) are activated, which, together with the increased cardiac output
associated with the hyperdynamic state, help to maintain renal blood flow.
Although activation of these systems has a positive effect by increasing arterial
pressure, they cause renal vasoconstriction, sodium retention leading to edema
and ascites, and secretion of insoluble water leading to hyponatremia and
decreased GFR [15]. In the most advanced stages of cirrhosis, severe renal
vasoconstriction occurs, and renal perfusion is no longer compensated by
increased cardiac output and GFR decreases, eventually leading to hepatorenal
syndrome. Recently, the concept of systemic inflammatory disease has emerged
in patients with cirrhosis, and there is increasing evidence that inflammation
plays a role in hepatorenal syndrome [16,17]. Cirrhosis is associated with
systemic inflammation that correlates with the severity of liver disease and portal
hypertension. The main mechanism is the translocation of bacterial and/or
pathogen-associated molecular patterns from the gut due to altered intestinal
permeability [18,19]. Inflammatory components can reach the systemic
circulation and peripheral organs, causing extrahepatic organ dysfunction,
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
22
including the kidneys. Inflammation can affect systemic circulation disorders and
impair renal perfusion. Patients with bacterial translocation have increased levels
of pro-inflammatory cytokines (TNF-α and IL-6) and vasodilators (such as nitric
oxide) [20,21]. Bacterial infections are a typical trigger for HRS; however,
approximately 30% of these patients develops systemic inflammatory response
syndrome (SIRS) without documented evidence of bacterial infection [22,23].
4. INCIDENCE OF HRS
HRS is thought to be a common complication in patients with advanced cirrhosis.
However, most classic studies on the prevalence of HRS in cirrhotic patients
were conducted many years ago and used atypical diagnostic criteria. Therefore,
the current incidence of HRS or its incidence relative to other causes of renal
failure in cirrhosis is unknown.
5. CLINICAL FINDINGS AND INVESTIGATIONS
In the setting of cirrhosis, HRS usually occurs in the late stages of the disease,
when patients have already had several episodes of some of the more serious
complications of cirrhosis, especially ascites. Patients with renal sodium retention
and dilutional hyponatremia with ascites are at high risk for HRS. The
predominant finding of HRS is renal failure, although many patients have other
manifestations, such as electrolyte imbalances, cardiovascular and infectious
complications, and complications related to liver disease [24-26]. Currently, when
frequent biochemical monitoring is widely used, the most common diagnosis of
HRS is an increase in serum creatinine or blood urea nitrogen concentration. In
some patients, serum creatinine and urea nitrogen concentrations rapidly rise to
very high values. Most of these patients have progressive oligo-anuria. In other
patients, the elevations in serum creatinine and blood urea nitrogen are
moderate and have no (or very little) tendency to progress over time, at least in
the short term. These two different patterns of renal failure progression define
two different clinical types of HRS [27,28]. The rate of progression used to define
type 1 HRS is arbitrarily set as a 100% increase in serum creatinine to a value
greater than 221 μmol/L (2.5 mg/dL) in less than 2 weeks. Patients with type 1
HRS have a very low GFR, usually less than 20 mL/min, and a very high serum
creatinine concentration (average approximately 356 μmol/L). In contrast, most
patients with type 2 HRS have milder GFR and creatinine (average 178 μmol/L).
An important clinical difference between the two types of HRS is that type 1
patients have a very poor short-term outcome compared to type 2 patients
[29,30].
In addition to renal failure, patients with HRS have sodium retention with salt and
water overload. In most cases, sodium retention is present and evident before
the development of HRS, but renal sodium excretion may further deteriorate as
renal failure occurs due to decreased GFR and activation of anti-natriuretic
systems [31,32]. As a result, increased positive sodium balance leads to weight
gain due to increased ascites volume and peripheral edema. Hyponatremia is
almost universal in HRS, so if serum sodium is normal in a patient with cirrhosis
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
23
and renal failure, the diagnosis of HRS is highly unlikely and the patient should
be investigated for another cause of renal failure. Hyponatremia is caused by a
decreased ability of the kidneys to excrete insoluble water, resulting in
disproportionate water retention relative to sodium (dilutional hyponatremia).
Severe metabolic acidosis is rare in HRS, except in patients who develop severe
infection [33,34].
Cardiovascular function is severely impaired in patients with HRS. Systemic total
vascular resistance is greatly reduced and arterial pressure is low in most
cases, despite strong activation of important vasoconstrictor mechanisms such
as renin-angiotensin and the sympathetic nervous system [35,36]. Cardiac output
is increased in most patients, while arterial pressure is usually low but stable
(mean arterial pressure approximately 70 mm Hg). In case of hemodynamic
instability, there is reason to suspect an infectious complication. Apart from
arterial pressure, the other cardiovascular abnormalities are not detected in the
clinical situations, unless invasive vascular monitoring is performed. However,
these procedures are not usually necessary in the clinical management of HRS
patients. Pulmonary edema, a common and serious complication of acute renal
failure, is very rare in patients with HRS unless treated aggressively with plasma
expanders [37]. Serious bacterial infections, particularly septicemia (either
spontaneous or indwelling catheter-related), spontaneous bacterial peritonitis,
and pneumonia are common complications in patients with HRS and are major
causes of death [38-40]. Both kidney failure and advanced liver disease are
believed to increase susceptibility to infection.
Finally, most patients with HRS have signs and symptoms of advanced liver
failure and portal hypertension, particularly jaundice, coagulopathy, malnutrition,
and hepatic encephalopathy, although HRS develops in some patients with only
moderate liver failure [41-43]. Ascites is common in patients with HRS, so the
absence of ascites in patients with cirrhosis and renal failure argues against HRS
as a cause of renal failure and points to other causes, particularly prerenal failure
due to volume depletion from excessive diuresis.
6. PRECIPITATING FACTORS
In some patients, HRS develops spontaneously without an obvious precipitating
event, while in others it occurs in close chronological association with a number
of precipitating factors that can cause circulatory disturbances and subsequent
renal hypo-perfusion. Known precipitating factors include bacterial infections,
large paracentesis without plasma expansion, and gastrointestinal bleeding
[44,45]. Among the different types of bacterial infections with cirrhosis, a clear
chronological and pathogenic relationship between infection and HRS was found
only in spontaneous bacterial peritonitis. This disorder is characterized by
spontaneous ascites infection, most often with gram-negative intestinal bacteria,
without intra-abdominal infection or intestinal perforation. Approximately 20% of
patients with spontaneous bacterial peritonitis develop HRS during or shortly
after infection - mostly type 1.
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
24
Whether HRS can also occur as a result of other serious bacterial infections has
not been studied. Another known cause of HRS is large-volume paracentesis
without plasma expansion [46]. Up to 15% of patients with ascites develop HRS
when large volumes of ascitic fluid (more than 5 L) are removed without
administration of a plasma expander. This association is one of the reasons why
large amounts of intravenous albumin should be administered during
paracentesis. Finally, renal failure occurs in approximately 10% of patients with
cirrhosis and gastrointestinal bleeding. However, a significant proportion of
episodes of renal failure after gastrointestinal bleeding are due to acute tubular
necrosis associated with hypovolemic shock rather than HRS. Intravascular
volume depletion (ie, diuretic-induced extrarenal fluid loss) has traditionally been
considered the trigger for HRS. However, conclusive evidence to support this
pathogenic association has not yet been reported.
7. PROGNOSIS
Of all the complications of cirrhosis, HRS has the worst prognosis [47]. Survival
is very low and spontaneous recovery is extremely rare. The main determinant of
survival is the type of HRS. Type 1 has an in-hospital survival rate of less than
10% and an expected median survival time of only 2 weeks. In contrast, type 2
patients have a much longer median survival time (about 6 months). Another
determinant of survival is the severity of liver disease. Patients with severe
hepatic impairment (Child-Pugh class C) have a much worse outcome than
patients with moderate hepatic impairment (class B). For many years, the
development of kidney failure was not considered to contribute to the dismal
results of HRS, and the cause of death was largely thought to be liver disease.
However, recent research suggests that kidney failure is an important factor in
outcomes, with patients whose kidney function improves after treatment living
longer than those who do not.
8. APPROACH TO DIAGNOSIS
The first step in diagnosing HRS is to demonstrate renal failure (low GFR).
Serum creatinine is generally considered a better marker of GFR [48]. However,
serum creatinine is not an ideal marker of GFR in cirrhosis, as it is usually lower
than any GFR expected due to low endogenous creatinine production associated
with reduced muscle mass present in most patients with advanced cirrhosis.
However, because the use of more sensitive elimination methods to measure
GFR is expensive and not available in all situations, serum creatinine is currently
the most popular method to estimate GFR in cirrhosis. In patients receiving
diuretics with elevated serum creatinine, serum creatinine should be remeasured
after the diuretic is discontinued, as diuretic use may be associated with a mild
and reversible increase in serum creatinine.
Because there are no specific diagnostic tests, the diagnosis of HRS should
always be made after other conditions that can cause kidney failure in cirrhosis
have been ruled out. Acute renal failure of prerenal origin due to gastrointestinal
losses (vomiting, diarrhea, bleeding, nasogastric tube) or renal related losses
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
25
(excessive diuresis) should be investigated in all patients with renal failure based
on history and physical examination [49]. When renal failure is secondary to
volume deficit, renal function improves rapidly after volume expansion, whereas
no improvement occurs in patients with HRS. The presence of shock before the
onset of renal failure excludes the diagnosis of HRS and suggests the diagnosis
of acute tubular necrosis. Hypovolemic shock due to gastrointestinal bleeding is
common and easily recognized in cirrhosis. However, septic shock can be more
difficult to diagnose because some patients with cirrhosis have no symptoms of
bacterial infection, and arterial hypotension caused by sepsis can be confused
with advanced liver disease, at least in the early stages [50,51]. Therefore,
bacterial infection (leukocyte count, ascitic fluid examination, cultures, C-reactive
protein) must always be ruled out before diagnosis of HRS. In contrast, some
patients with cirrhosis and bacterial infection develop transient renal failure,
which in most cases improves after the infection resolves. Therefore, HRS
should only be diagnosed if renal failure persists after complete resolution of the
infection. Cirrhotic patients are at high risk of developing kidney failure when
using nephrotoxic agents such as NSAIDs, aminoglycosides or other drugs
[52,53]. Therefore, treatment with these nephrotoxic drugs in the days or weeks
before the onset of renal failure must always be excluded. Renal failure can also
occur after the administration of radiocontrast agents. However, whether cirrhotic
patients are at high risk for this complication has never been assessed. Finally,
patients with cirrhosis may also develop renal failure due to parenchymal kidney
disease, particularly glomerulonephritis [54-56]. It can occur with any cause of
cirrhosis, but is particularly common with chronic hepatitis B or C infection or
chronic alcoholism. These cases can be recognized by proteinuria, hematuria or
both. In some cases, the diagnosis can be confirmed with a kidney biopsy.
9. PREVENTION OF HEPATORENAL SYNDROME
Strategies to prevent the development of hepatorenal syndrome include
preventing progression of liver disease in the well-compensated patient, reversing
decompensation in patients with advanced cirrhosis, avoiding agents that
aggravate AKI, and avoiding factors that further impair blood flow and reduce
renal perfusion [57,58]. Prophylactic antibiotics to prevent spontaneous bacterial
peritonitis and intravenous albumin after variceal bleeding in patients with
spontaneous bacterial peritonitis and patients undergoing large paracentesis (> 5
L) have been shown to reduce the incidence of hepatorenal syndrome. There is
no evidence that albumin in addition to antibiotics reduces the incidence of AKI in
patients with bacterial infection other than spontaneous bacterial peritonitis. In a
controlled trial, long-term administration of albumin to patients with
decompensated cirrhosis was associated with improvements in spontaneous
bacterial peritonitis, hepatorenal syndrome, and survival [59].
β-blockers are very effective in preventing variceal bleeding and are widely used
in patients with cirrhosis and significant portal hypertension. A recent meta-
analysis indicates that the use of beta-blockers is not associated with a significant
increase in mortality in patients with ascites or refractory ascites [60]. However,
some series have shown increased mortality in patients with refractory ascites
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
26
receiving β-blockers compared with patients not receiving beta-blockers. It has
been suggested that the reduction in cardiac output caused by β-blockers may
lead to AKI. Clinicians must individually weigh the risks and benefits of
continuous nonselective beta-blockers in patients with refractory ascites.
10. MANAGEMENT OF HEPATORENAL SYNDROME
The etiology of AKI should be urgently investigated to prevent further worsening
of AKI, as progression to advanced AKI is associated with higher mortality. This
is especially important for those with hepatorenal syndrome, because early
treatment can increase the likelihood of improvement in hepatorenal syndrome,
which can improve short-term survival. Albumin administration is a crucial step in
the treatment and diagnosis of hepatorenal syndrome; However, it is important
to be cautious when administering fluids to patients with AKI to avoid significant
fluid retention and pulmonary edema, as renal excretion of sodium and water is
reduced in cirrhotic patients.
11. PHARMACOLOGICAL TREATMENT
Vasoconstrictive agents along with albumin are the main choice in the treatment
of hepatorenal syndrome. Terlipressin is the most commonly used vasopressin
analog; however, it is not accepted in all countries [61]. The effectiveness of
terlipressin and albumin in the treatment of hepatorenal syndrome has been
demonstrated in several studies with a response rate of 25-75%. The most
important side effects of terlipressin are related to vasoconstriction, with a risk of
myocardial infarction and intestinal ischemia [62,63]. Baseline serum creatinine
and degree of acute chronic liver failure are associated with response to
terlipressin. However, there have been no studies on the use of vasoconstrictors
with lower serum creatinine in the early stages of hepatorenal syndrome.
Other vasoconstrictors have been recommended in combination with albumin.
Norepinephrine (administered intravenously at a dose of 0.5-3 mg/h) is an
alternative agent that has been shown to be effective in increasing arterial
pressure and reversing renal insufficiency in patients with hepatorenal syndrome
in small studies; however, a recent controlled trial indicates that norepinephrine is
inferior to terlipressin in terms of hepatorenal syndrome, need for renal
replacement therapy (RRT), and overall survival [64]. A combination of midodrine
and octreotide, used in countries where terlipressin is not yet available, was
shown to be less effective than terlipressin in a study [65].
12. TRANSJUGULAR INTRAHEPATIC PORTOSYSTEMIC SHUNT
Theoretically, a transjugular intrahepatic portosystemic shunt may improve renal
function in hepatorenal syndrome by reducing portal hypertension and by
reducing and reversing the circulatory changes (and possibly systemic
inflammation) that cause hepatorenal syndrome [66]. Small studies have shown
that transjugular intrahepatic portosystemic shunt is associated with a reduction
in serum creatinine that may improve survival in patients with hepatorenal
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
27
syndrome, but patients with advanced liver disease have a high incidence and
further exacerbation of hepatic encephalopathy.
13. RENAL REPLACEMENT THERAPY
Initiation of RRT in patients with hepatorenal syndrome is controversial and has
generally been considered a bridge in patients scheduled for transplantation [67].
Recent studies have shown that disease severity and degree of organ failure are
better predictors of 28-day mortality than the cause of AKI in patients with acute
and chronic liver failure. Therefore, it seems reasonable to consider a RRT trial in
selected patients, regardless of the transplant candidate. The ideal time of RRT
initiation in cirrhotic patients has not been studied, so it should be individualized
and based on clinical reasons, such as renal impairment with electrolyte
imbalance unresponsive to medical therapy or diuretic intolerance/resistance.
RRT should also be considered to prevent fluid retention if daily fluid balance
cannot be maintained or is negative regardless of their urine output.
14. LIVER SUPPORT SYSTEM
Although initial results suggested that albumin dialysis with a molecular
adsorptive circulation system may improve outcomes in patients with hepatorenal
syndrome, this has not been confirmed in larger randomized trials [68]. A
randomized trial of patients with acute or chronic liver failure found no significant
difference in 28-day mortality between patients with hepatorenal syndrome who
underwent molecular adsorptive recirculation compared with conventional
medical therapy. There is currently no evidence that albumin dialysis is better
than conventional filtration in patients requiring CRT.
15. LIVER TRANSPLANT ALONE VERSUS SIMULTANEOUS LIVER-
KIDNEY TRANSPLANT
Predicting the recovery of renal function and its extent of improvement after liver
transplantation is difficult because it is difficult to delineate the relative impact of
preexisting comorbidities, undetected intrinsic renal disease, perioperative
events, and post transplant immunosuppression on renal failure after liver
transplantation. Onset of AKI before liver transplantation has been shown to be
associated with a higher risk of CKD and ESKD after liver transplantation and
has also been associated with an increased risk of mortality [69]. Liver
transplantation is the primary form of treatment for patients with hepatorenal
syndrome, and in theory, kidney function is fully reversible after transplantation.
Renal recovery and patient survival after liver transplantation were significantly
higher in patients with hepatorenal syndrome than in patients with acute tubular
necrosis and comparable to patients without AKI or stage 1 AKI, regardless of
their dialysis status before transplantation.
The introduction of organ allocation based on the end-stage liver disease model
in 2002 led to a sharp increase in the number of simultaneous liver-kidney
transplants, as liver transplant candidates with kidney failure were preferred. At
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
28
present simultaneous liver and kidney transplants account for 10% of all liver
transplants in the United States, and approximately 5% of transplanted
deceased donor kidneys are removed from kidney-only transplant candidates,
causing concern in the kidney transplant community, especially because of the
uncertain benefits in case of simultaneous liver and kidney transplantation
[70,71]. The decision to perform simultaneous liver-kidney transplantation and
liver transplantation is not only a concern for post-transplant mortality, but also a
concern for lack of renal recovery, which is thought to increase mortality. Studies
have shown that liver transplant patients on the waiting list have a higher
mortality compared to patients waiting only for a kidney transplant. The Organ
Procurement and Transplantation Network recently developed criteria for
simultaneous liver and kidney transplantation based on previous consensus
recommendations that include factors such as AKI and duration of dialysis and
evidence of chronic kidney disease. Age, comorbidities or cause of AKI, which
may influence renal recovery, are not currently in the criteria [72].
16. NEWER BIOMARKERS
Early diagnosis and recognition of the AKI phenotype is crucial, as treatment
varies depending on the different causes. Conventional measures such as
urinary excretion or sodium or urea fraction have been shown to be significantly
limited in patients with advanced cirrhosis and to correlate poorly with biopsy
findings. Several novel biomarkers have recently been investigated, the most
studied of which are neutrophil-gelatinase-associated lipocalin, kidney molecule-
1, liver fatty acid-binding protein, and IL-18 [73-75]. These specific biomarkers
usually reflect early signs of ischemia-related events and may play a role in the
diagnosis of AKI before liver transplantation. These biomarkers are not specific
for kidney damage, can be influenced by inflammation or infection, and have not
been validated using kidney biopsy as the gold standard. Furthermore,
considerable overlap between the different phenotypes was observed, and no
clear cut-off value distinguishes hepatorenal syndrome from acute tubular
necrosis. Biomarkers that predict recovery from AKI after liver transplantation
may improve decision algorithms regarding the need for liver-kidney
transplantation or kidney-sparing therapies.
17. IMAGE STUDIES
Renal ultrasonography is a useful noninvasive test to rule out structural causes of
AKI, such as obstructive uropathy and intrinsic parenchymal renal disease, which
preclude the diagnosis of hepatorenal syndrome [76]. Assessment of renal artery
resistive indices by Doppler ultrasound, contrast-enhanced ultrasound, and
magnetic resonance elastography has been shown in very small studies to be
associated with the development of hepatorenal syndrome. Whether these
methods help in the early diagnosis of hepatorenal syndrome, distinguish
hepatorenal syndrome from other AKI phenotypes, or predict vasoconstrictor
response, need to be further investigated in larger studies.
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
29
18. CONCLUSIONS AND PERSPECTIVES
In recent years, significant advances have been made in the diagnosis and
treatment of hepatorenal syndrome. Even with vasoconstrictors and albumin,
three-month mortality is particularly high without liver transplantation. In addition
to splanchnic and systemic circulatory changes, inflammation may also play
significant role in the development of hepatorenal syndrome. Therapeutic
interventions designed to control inflammation may help prevent or reverse
hepatorenal syndrome. A new biomarker combined with imaging studies may
improve the diagnostic performance of AKI in cirrhotic patients. Irreversible renal
changes are probably underestimated; In the future, new biomarkers and
imaging studies may provide additional information on the healing potential of the
kidney after liver transplantation and may influence the decision for simultaneous
liver and kidney transplantation.
COMPETING INTERESTS
Authors have declared that no competing interests exist.
REFERENCES
1. Raseen Tariq, Yousaf Hadi, Khusdeep Chahal, Sivani Reddy, Habeeb
Salameh, Ashwani K Singal. Incidence, mortality and predictors of acute
kidney injury in patients with cirrhosis: A systematic review and meta-
analysis. J Clin Transl Hepatol. 2020;8(2):135-142.
2. Ashwani K Singal, Yong-Fang Kuo, K Rajender Reddy, Ramon Bataller,
Paul Kwo. Healthcare burden and outcomes of hepatorenal syndrome
among cirrhosis‐related hospitalisations in the US. Aliment Pharmacol
Ther. 2022;56(10):1486-1496.
3. Ahmed Adel Amin, Eman Ibrahim Alabsawy, Rajiv Jalan, Andrew
Davenport. Epidemiology, pathophysiology, and management of
hepatorenal syndrome. Semin Nephrol. 2019;39(1):17-30.
4. Wong Florence. Acute kidney injury in liver cirrhosis: new definition and
application. Clin Mol Hepatol. 2016;22(4):415-422.
5. Mitra K Nadim, Lui G Forni, Ravindra L Mehta, Michael J Connor Jr,
Kathleen D Liu, Marlies Ostermann, et al. COVID-19-associated
acute kidney injury: consensus report of the 25th Acute Disease
Quality Initiative (ADQI) Workgroup. Nat Rev Nephrol. 2020;16(12):747-
764.
6. Paolo Angeli, Pere Gines, Florence Wong, Mauro Bernardi, Thomas D
Boyer, Alexander Gerbes, et al. Diagnosis and management of acute
kidney injury in patients with cirrhosis: revised consensus
recommendations of the International Club of Ascites. Gut.
2015;64(4):531-7.
7. J P Mulkay, H Louis, V Donckier, N Bourgeois, M Adler, J Deviere, O Le
Moine. Long-term terlipressin administration improves renal function in
cirrhotic patients with type 1 hepatorenal syndrome: a pilot study. Acta
Gastroenterol Belg. 2001;64(1):15-9.
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
30
8. Palleti, Sujith Kumar, Sreekanth Avula, and Sandesh Dewan. Kidney: A
Review on End Stage Renal Disease, Dialysis and Transplant. Int Clinc
Med Case Rep Jour. 2023;2(10):1-8.
9. Avula Sreekant, et al. Diabetic Kidney Disease. Int Clinc Med Case Rep
Jour. 2023;2(11):1-4.
10. Yasuko Iwakiri, Roberto J Groszmann. Groszmann. Vascular endothelial
dysfunction in cirrhosis. J Hepatol. 2007;46(5):927-34.
11. J Clària, W Jiménez, J Ros, M Asbert, A Castro, V Arroyo, F Rivera, J
Rodés. Pathogenesis of arterial hypotension in cirrhotic rats with ascites:
role of endogenous nitric oxide. Hepatology. 1992;15(2):343-9.
12. Abshiro H Mayow, Fatima Ahmad, Muhammad Sohaib Afzal, Muhammad
Usama Khokhar, Daneyal Rafique, Sai Krishna Vallamchetla, et al. A
Systematic Review and Meta-Analysis of Independent Predictors for
Acute Respiratory Distress Syndrome in Patients Presenting with Sepsis.
Cureus. 2023;15(4):e37055.
13. Møller, Søren, Jens H. Henriksen. Cirrhotic cardiomyopathy. Journal of
Hepatology. 2021;53(1):179-190.
14. Andrés Cárdenas, Vicente Arroyo. Study on Recognition of Heart
Arrhythmias by Deep Learning Approach of the ECG Signals using Edge
Devices.
15. Cárdenas, Andrés, and Vicente Arroyo. Mechanisms of water and sodium
retention in cirrhosis and the pathogenesis of ascites. Best Pract Res Clin
Endocrinol Metab. 2003;17(4):607-22.
16. Vicente Arroyo, Paolo Angeli, Richard Moreau, Rajiv Jalan, Joan Clària,
Jonel Trebicka, et al. The systemic inflammation hypothesis: towards a
new paradigm of acute decompensation and multiorgan failure in
cirrhosis. J Hepatol. 2021;74(3):670-685.
17. Ramalingam, Sathishkumar, et al. Acute pancreatitis and hepatitis: A rare
presentation of Epstein–Barr virus.
18. Reiner Wiest, Guadalupe Garcia-Tsao. Bacterial translocation (BT) in
cirrhosis. Hepatology. 2005;41(3):422-33.
19. Vaillant, Angel A. Justiz, and Roopa Naik. HIV-1 associated opportunistic
infections. StatPearls [Internet]. StatPearls Publishing, 2021.
20. Reiner Wiest, Melissa Lawson, Markus Geuking. Pathological bacterial
translocation in liver cirrhosis. J Hepatol. 2014;60(1):197-209.
21. Sujith K Palleti, Santhoshi R Bavi, Margaret Fitzpatrick, Anuradha
Wadhwa. First case report of Sphingobium lactosutens as a human
pathogen causing peritoneal dialysis-related peritonitis. Cureus.
2002;14(7).
22. Puneeta Tandon, Guadalupe Garcia-Tsao. Bacterial infections, sepsis,
and multiorgan failure in cirrhosis." Seminars in liver disease. Semin Liver
Dis. 2008;28(1):26-42.
23. Fareeha Naseer Syed, Nadia Kashif, Mubashir Razzaq Khan, Fayaz
Ahmed Memon, et al. Hepatitis C Virus Infection in Patients with Beta
Thalassemia after Multiple Transfusions at a Tertiary Care Hospital.
Pakistan Journal of Medical & Health Sciences. 2023;17(01):363-363.
24. Pere Ginès, Mónica Guevara, Vicente Arroyo, Juan Rodés. Hepatorenal
syndrome. Lancet. 2003;362(9398):1819-1827.
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
31
25. Sai Kiran Kondabolu, Yasitha Kakarlapudi, Haider Malik, Hamza Malik,
Saima Khan, Praveen Kumar Komminni, et al. Beneficial Impacts of
Fenoldopam on Patients with or at Risk for Acute Renal Failure and
Undergoing Surgery: A Meta-Analysis of Randomized Clinical Trials.
Cureus . 2023;15(2):e34584.
26. Palleti Sujith K, et al. Study about Sphingobium Lactosutens: A Human
Pathogen Causing Peritoneal Dialysis Related Peritonitis. Perspective of
Recent Advances in Medical Research. BP International 9. 2023:31-35.
27. Vicente Arroyo, Carlos Terra, Pere Ginès. Advances in the pathogenesis
and treatment of type-1 and type-2 hepatorenal syndrome. J Hepatol.
2007;46(5):935-46.
28. Palleti, Sujith Kumar, Sreekant Avula, and Roopa Naik. Chronic Kidney
Disease. Int Clinc Med Case Rep Jour. 2023;2(11):1-15.
29. Florence Wong, Wesley Leung, Mohammed Al Beshir, Max Marquez,
Eberhard L Renner, et al. Outcomes of patients with cirrhosis and
hepatorenal syndrome type 1 treated with liver transplantation. Liver
Transpl. 2015;21(3):300-7.
30. Sreekant Avula. Continuous Renal Replacement Therapy. Int Clinc Med
Case Rep Jour. 2023;2(12):1-13.
31. Pere Ginès, Mónica Guevara. Hyponatremia in cirrhosis: pathogenesis,
clinical significance, and management. Hepatology. 2008;48(3):1002-
10.
32. Hanif Muhammad O, Atul Bali, Kamleshun Ramphul. Acute renal tubular
necrosis. StatPearls. StatPearls Publishing, 2022.
33. Martin-Walter Welker, Stefan Luhne, Christian M Lange, Johannes
Vermehren, Harald Farnik, Eva Herrmann, et al. Lactic acidosis in patients
with hepatitis C virus cirrhosis and combined ribavirin/sofosbuvir
treatment. J Hepatol. 2016;64(4):790-9.
34. Bali Atul, Roopa Naik. Pseudo Metabolic Acidosis in the Setting of
Hypertriglyceridemia: A Case Report. 2020.
35. S Møller, J H Henriksen. Cardiovascular complications of cirrhosis. Gut.
2008;57(2):268-78.
36. Sameer, Muhammad Ali, et al. Comparison of Coronary Artery Bypass
Grafting Combined With Mitral Valve Repair Versus Coronary Artery
Bypass Grafting Alone in Patients With Moderate Ischemic Mitral
Regurgitation: A Meta-Analysis. Cureus. 2023;15(4).
37. Barkhane, Zineb, et al. Independent Predictors of Mortality in Systemic
Sclerosis-Associated Pulmonary Arterial Hypertension: A Systematic
Review and Meta-Analysis. Cureus. 2023;15(5).
38. Douglas A Simonetto, Laura Piccolo Serafim, Alice Gallo de Moraes,
Ognjen Gajic, Patrick S Kamath. Management of sepsis in patients with
cirrhosis: current evidence and practical approach. Hepatology.
2019;70(1):418-428.
39. Abdulmaleek Idanesimhe Sado, Muhammad S Afzal, Lavanya
Kannekanti, Hrushikesh Reddy Pamreddy, Jorge Pimentel Campillo,
Vaishnavi Kandukuri, et al. A Meta-Analysis on Predictors of Mortality
Among Patients Hospitalized for Acute Exacerbation of Asthma. Cureus.
2023;15(2).
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
32
40. Sathishkumar Ramalingam, Kulothungan Gunasekaran, Harkesh Arora,
Maheswari Muruganandam, Priyesh Padmanabhan. Clostridium difficile
and noncirrhotic hyperammonnemia in a patient with COVID- 19 infection.
Cureus. 2021;13(4):e14533.
41. Scott W Biggins, Paulo Angeli, Guadalupe Garcia-Tsao, Pere Ginès,
Simon C Ling, Mitra K Nadim, et al. Diagnosis, evaluation, and
management of ascites, spontaneous bacterial peritonitis and hepatorenal
syndrome: 2021 practice guidance by the American Association for the
Study of Liver Diseases. Hepatology. 2021;74(2):1014-1048.
42. Rakesh Pilla, Sujith Kumar Palleti, Renuka Rayana, Satish Reddy Skss,
Aminah Abdul Razzack, Sruti Kalla. Glycated haemoglobin (HbA1c)
variations in nondiabetics with nutritional anemia. Cureus. 2020;12(11).
43. Atul Bali, Roopa Naik. The Impact of a Vegan Diet on Many Aspects of
Health: The Overlooked Side of Veganism. Cureus. 2023;15(2).
44. Bataller, Ramón, et al. Hepatorenal syndrome. Seminars in liver disease.
1997;17(03).
45. Jithin Karedath, Fnu Avanteeka, Muhammad Nouman Aslam, Ahmad
Nadeem, Rao Ahmed Yousaf, Sandesh Shah, et al. Comparison of
Effectiveness and Safety of Low-Dose Versus Standard-Dose Intravenous
Recombinant Tissue Plasminogen Activator in Patients with Acute
Ischemic Stroke: A Meta- Analysis. Cureus. 2023;15(2).
46. Diaconescu, Diana, et al. Hepatorenal syndrome: a review. Arch Balk Med
Union. 2018;53(2):239-245.
47. Thomas D Boyer, Arun J Sanyal, Guadalupe Garcia-Tsao, Frederick
Regenstein, Lorenzo Rossaro, Beate Appenrodt, et al. Impact of liver
transplantation on the survival of patients treated for hepatorenal
syndrome type 1. Liver Transpl. 2011;17(11):1328-32.
48. Stefan Herget-Rosenthal, Arend Bökenkamp, Walter Hofmann. How to
estimate GFR-serum creatinine, serum cystatin C or equations?. Clin
Biochem. 2007;40(3-4):153-61.
49. Kar, Saikat, et al. A two-center, retrospective cohort trial of a tumor
necrosis factor inhibitor coupled with intravenous immunoglobulin and
heparin for the treatment of recurrent spontaneous abortion.
50. Djillali Annane, Eric Bellissant, Jean-Marc Cavaillon. Septic shock. Lancet.
2005;365(9453):63-78.
51. Palleti Sujith K., et al. The First Case of Ceftazidime-Induced
Antibiomania: A Case Report and Literature Review. Cureus. 2022;14(12).
52. Naughton, Cynthia A. Drug-induced nephrotoxicity. Am Fam Physician.
2008;78(6):743-750.
53. Palleti Sujith K, Anuradha Wadhwa, Abdul H. Rasheed. Ceftazidime-
induced Antibiomania: A Rare Case Study and Review of Literature.
Research Highlights in Disease and Health Research. 2023;6:56-61.
54. V M Campese, N Mitra, D Sandee. Hypertension in renal parenchymal
disease: why is it so resistant to treatment?. Kidney Int. 2006;69(6):967-
73.
55. Palleti Sujith K, Maria M. Picken, Anuradha Wadhwa. Sjögren's Syndrome
Overlap with Lupus Nephritis: A Case Report and Literature Review of a
Rare Entity. Cureus. 2023;15(5).
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
33
56. Sujith K Palleti, Hanna Larson, Sreekant Avula, Maria M Picken, Anuradha
Wadhwa. Subarachnoid hemorrhage in autoimmune vasculitis: a rare
presentation of systemic lupus erythematosus-antineutrophil cytoplasmic
autoantibody-associated vasculitis overlap syndrome. Cureus. 2023;15(5).
57. El-Sayed Ibrahim, Ayman Alsebaey, Hassan Zaghla, Sabry Moawad
Abdelmageed, Khalid Gameel, Eman Abdelsameea. Long-term rifaximin
therapy as a primary prevention of hepatorenal syndrome. Eur J
Gastroenterol Hepatol. 2017;29(11):1247-1250.
58. Atul Bali, John Kevin Hix, Peter Kouides. Safe and effective use of chronic
transdermal estradiol for life- threatening uremic bleeding in a patient with
coronary artery disease. Nephron Extra. 2014;4(2):134-7.
59. Mauro Bernardi, Paolo Angeli, Joan Claria, Richard Moreau, Pere Gines,
Rajiv Jalan, et al. Albumin in decompensated cirrhosis: new concepts and
perspectives. Gut. 2020;69(6):1127-1138.
60. Mattias Mandorfer, Thomas Reiberger. Beta blockers and cirrhosis, 2016.
Dig Liver Dis. 2017;49(1):3-10.
61. Wong, Florence, et al. Terlipressin plus albumin for the treatment of type 1
hepatorenal syndrome. New England Journal of Medicine.
2021;384(9):818-828.
62. Sashidhar V Sagi, Sahil Mittal, Krishna S Kasturi, Gagan K Sood.
Terlipressin therapy for reversal of type 1 hepatorenal syndrome: a
meta‐analysis of randomized controlled trials. J Gastroenterol Hepatol.
2010;25(5):880-5.
63. Zaid Munir, Muhammad Akash, Fnu Jaiprada, Bilal Abu Tarboush, Osama
Ijaz, Anan Bseiso, et al. Evaluation of the Effects of Extracorporeal
Shockwave Therapy in Patients with Peripheral Arterial Disease: A Meta-
Analysis of Randomized Control Trials. Cureus. 2023;15(2):e34729.
64. Antonio Paulo Nassar Junior, Alberto Queiroz Farias, Luiz Augusto
Carneiro D' Albuquerque, Flair José Carrilho, Luiz Marcelo Sá
Malbouisson. Terlipressin versus norepinephrine in the treatment of
hepatorenal syndrome: a systematic review and meta-analysis. PLoS One.
2014;9(9):e107466.
65. Marta Cavallin, Patrick S Kamath, Manuela Merli, Silvano Fasolato,
Pierluigi Toniutto, Francesco Salerno, et al. Terlipressin plus albumin
versus midodrine and octreotide plus albumin in the treatment of
hepatorenal syndrome: a randomized trial. Hepatology. 2015;62(2):567-
74.
66. Tingxue Song, Martin Rössle, Fuliang He, Fuquan Liu, Xiaozhong Guo,
Xingshun Qi. Transjugular intrahepatic portosystemic shunt for
hepatorenal syndrome: A systematic review and meta-analysis. Dig Liver
Dis. 2018;50(4):323-330.
67. Achuthan Sourianarayanane, Rupesh Raina, Gaurav Garg, Arthur J
McCullough, Robert S O'Shea. Management and outcome in hepatorenal
syndrome: need for renal replacement therapy in non- transplanted
patients. Int Urol Nephrol. 2014;46(4):793-800.
68. Evangelos Tsipotis, Asim Shuja, Bertrand L Jaber. Jaber. Albumin dialysis
for liver failure: a systematic review. Adv Chronic Kidney Dis.
2015;22(5):382-90.
Advances in Renal Diseases and Dialysis
Hepatorenal Syndrome
34
69. Ashwani K Singal, Song Ong, Sanjaya K Satapathy, Patrick S Kamath,
Russel H Wiesner. Simultaneous liver kidney transplantation. Transpl Int.
2019;32(4):343-352.
70. M K Nadim, R S Sung, C L Davis, K A Andreoni, S W Biggins, G M
Danovitch, S Feng, et al. Simultaneous liver-kidney transplantation
summit: current state and future directions. Am J Transplant.
2012;12(11):2901-8.
71. Karima Benaicha, Raja Ram Khenhrani, Maha Veer, Sapna Devi, Usman
Shahbaz, Qais M Salah, et al. Efficacy of Molnupiravir for the Treatment of
Mild or Moderate COVID-19 in Adults: A Meta-Analysis. Cureus.
2023;15(5).
72. Erik L Lum, Andrés Cárdenas, Paul Martin, Suphamai Bunnapradist.
Current status of simultaneous liver‐ kidney transplantation in the United
States. Liver Transpl. 2019;25(5):797-806.
73. Kapil Gupta, Abhishek Bhurwal, Cindy Law, Scott Ventre, Carlos D
Minacapelli, Savan Kabaria, et al. Acute kidney injury and hepatorenal
syndrome in cirrhosis. World J Gastroenterol 2021;27(26):3984.
74. Raza A Khan, Nidhi Patel, Atunde Folajimi, Bansari Raveena Bai, Vrushak
Patel, Praveen Kumar Komminni, et al. Comparison of the Efficacy and
Safety of Metformin-Based Combination Therapy Versus Metformin Alone
in Children and Adolescents with Type 2 Diabetes Mellitus: A Meta-
Analysis. Cureus. 2023;15(2):e35014.
75. Rohan Vijay Yewale, Balakrishnan Siddartha Ramakrishna. Novel
biomarkers of acute kidney injury in chronic liver disease: Where do we
stand after a decade of research?. Hepatol Res. 2023;53(1):3-17.
76. Brian Erly, William D Carey, Baljendra Kapoor, J Mark McKinney, Mathew
Tam, Weiping Wang. Hepatorenal syndrome: a review of pathophysiology
and current treatment options. Semin Intervent Radiol. 2015;32(4):445-54.
___________________________________________________________________________________
© Copyright (2023): Author(s). The licensee is the publisher (B P International).
DISCLAIMER
This chapter is an extended version of the article published by the same author(s) in the following journal.
Annal of Clin Med & Med Res, 1(1): 1-15, 2023. DOI: https://doi.org/10.5281/zenodo.8118544
________________________________________________________________________
a Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre,
Pennsylvania, USA.
b Department of Gastroenterology, Brooklyn Hospital Center, Brooklyn, New York, USA.
c Department of Internal Medicine, University Hospital and Clinics, Lafayette, Louisiana, USA.
d Department of Internal Medicine/Nephrology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre,
Pennsylvania, USA.
*Corresponding author;
Chapter 3
Print ISBN: 978-81-19491-84-1, eBook ISBN: 978-81-19491-85-8
Diabetic Kidney Disease
Roopa Naik a*, Rajeshwari Ramachandran b,
Bushra Firdous Shaik c and Atul Bali d
DOI: 10.9734/bpi/mono/978-81-19491-84-1/CH3
ABSTRACT
The kidney is probably the most important target of microvascular damage in
diabetes. A significant proportion of people with diabetes develop kidney disease
because of their disease and/or other comorbidities, including hypertension and
age-related kidney loss. The presence and severity of chronic kidney disease
(CKD) identifies individuals at increased risk for adverse health outcomes and
premature mortality. Therefore, the prevention and treatment of CKD in diabetics
is now a central goal of their general care. Intensive care of diabetic patients
includes blood sugar and blood pressure control and blockade of the renin-
angiotensin-aldosterone system; these approaches reduce the incidence of
diabetic kidney disease and slow its progression. In fact, the significant reduction
in the incidence of diabetic kidney disease (DKD) over the past 30 years and the
improvement in patient prognosis is largely due to improvements in diabetes
management. However, there is a need for innovative treatment strategies to
prevent and control the progression of DKD. In this review, we summarize what
is currently known about the pathogenesis of CKD in patients with diabetes and
the key pathways and targets involved in its progression. In addition, we discuss
the current evidence that supports the prevention and treatment of DKD and
some of the controversies. Finally, we will explore ways to develop new
interventions by making urgently needed investments in targeted and focused
research.
Keywords: Diabetes; hypertension; chronic kidney disease; albuminuria; SGLT
inhibitors.
1. INTRODUCTION
Diabetic kidney disease (DKD) remains by far the most common cause of end-
stage renal disease (ESRD) in the United States and most parts of the world [1,2].
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
36
The involvement of the kidneys directly and indirectly increases the effects on
other organs and increases the morbidity and mortality of diabetics. With the
increasing industrialization, increased inactivity and changes in diet and lifestyle,
as well as the increasing prevalence of obesity, insulin resistance and type 2
diabetes increases. Since many of the classic diabetic symptoms of type 2
diabetes are absent, unlike type 1 diabetes, the importance of screening
methods to identify these patients for kidney involvement is very important [3,4].
Our understanding of the pathogenesis of DKD has also advanced significantly.
In addition, several new drugs have undergone clinical trials with some success.
2. EPIDEMIOLOGY
Diabetes affects 30.3 million people of all ages, which is 9.4 percent of the US
population and approximately 149 million people and 10.9 percent of the Chinese
population and 415 million worldwide [5]. DKD is the most common cause of end
stage renal disease (ESRD) worldwide and is associated with increased
morbidity and mortality especially in diabetic patients. Both types of diabetes can
lead to CKD and eventually ESRD. However, because the prevalence of type 2
diabetes is much higher than type 1 diabetes, people with ESRD often have type
2 diabetes [6,7]. The overall incidence is approximately 4-17% 20 years after
diagnosis and approximately 16% after 30 years. Although the incidence of
chronic kidney disease (CKD) in diabetics has decreased in the United States in
recent years, the prevalence remains high. In 2018, United States Renal Data
System (USRDS) annual data reported that diabetes accounted for 36 percent of
chronic diseases in the National Health and Nutrition Examination Survey
(NHANES) population from 2013 to 2016, up from 44 percent from 2001 to 2004.
However, the total number of DKD patients continued to increase due to the
increase in the number of diabetic patients. The number of adults with diabetes
over 18 years of age who started treatment for ESRD also increased
significantly, from more than 40,000 in 2000 to more than 50,000 in 2014 [8,9]. If
type 1 diabetics have diabetic nephropathy, they are likely to reach end-stage
diabetes. Although the risk of type 2 diabetes after the onset of clinical
nephropathy is approximately 20%. Perhaps one of the reasons is that many
people with type 2 diabetes die of cardiovascular events before they reach ESRD
[10,11]. About 30% of patients with type 2 diabetes who have kidney disease
actually have some other kidney disease, but not diabetic nephropathy, as
evidenced by the absence of retinopathy and albuminuria.
3. DEFINITION OF DIABETIC KIDNEY DISEASE
Diabetic kidney disease (also called "chronic kidney disease" due to diabetes
(CKD) or diabetic nephropathy) is defined as persistently elevated albuminuria
>300 mg/24 h (or >200 µg) in both type 1 and type 2 diabetes or albumin-to-
creatinine ratio (ACR) > 300 mg/g, confirmed in at least two of three samples,
without evidence of concomitant diabetic retinopathy or other forms of kidney
disease [12]. The normal range for albuminuria is 30 mg/g, but values in either
range may be associated with an increased risk of kidney and cardiovascular
disease. Moderately increased urinary albumin excretion (microalbuminuria) (30-
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
37
300 mg/g) is widely considered to be a precursor to diabetic nephropathy,
indicating both early risk and a target for intervention [13].
4. MECHANISMS/PATHOPHYSIOLOGY
DKD has traditionally been considered a microvascular disease grouped together
with retinopathy and neuropathy, distinct from macrovascular disease that
contributes to coronary artery disease, peripheral vascular disease, and
cerebrovascular disease [14,15]. However, each disease can be considered a
tissue-specific manifestation of the same pathogenic process, and DKD is a renal
manifestation of the same glucose-driven process that occurs elsewhere in the
body at sensitive sites. Although all cells in diabetes are exposed to chronically
high plasma glucose levels, only some show progressive dysfunction, the best
example being the endothelial cells that line blood vessels [16]. In particular,
the inability of endothelial cells to regulate their glucose transport in response
to high glucose levels leads to an overwhelming intracellular glucose flux, which
triggers the generation of pathogenic mediators that contribute to diabetic
complications, including DKD.
5. REACTIVE OXYGEN SPECIES
Excessive glucose flux leads to the generation of toxic intermediates, the most
important of which are probably reactive oxygen species (ROS) [17,18]. Excess
glucose production can generate reactive oxygen species in various ways.
Increased mitochondrial substrate oxidation and the resulting increased
mitochondrial membrane potential lead to overproduction of superoxide. The
centrality of ROS in triggering these processes is illustrated by the fact that they
can be prevented if hyperglycemia-mediated ROS production is limited.
6. PATHWAYS OF NUTRIENT RECOGNITION
Each cell has pathways that recognize nutrient abundance and respond
specifically to it to ensure efficient use of the substrate [19]. The best known of
these nutrient sensors are rapamycin (mTOR), 5'AMP-activated protein kinase
(AMPK) and sirtuins. From a kidney perspective, diabetes directly causes
changes in the expression and activity of AMPK, sirtuins, and mTOR37, as well
as downstream signaling in cellular homeostasis, including reductions in
autophagy, regeneration, mitochondrial biogenesis, and other cytoprotective
responses that promote DKD.
7. MULTIFACTORIAL PATHOGENESIS OF DKD
Only a third of people with type 1 diabetes develop overt nephropathy, while
almost all people with type 1 diabetes eventually develop some degree of
retinopathy. This suggests that risk factors other than hyperglycemia must be
involved in DKD [20]. Pathogenic pathways induced by high glucose levels and
maintained in the kidney can be enhanced by a number of different factors.
These include several metabolic factors, including fatty acid excess, carbonyl
and oxidative stress, and hemodynamic factors, including shear stress from
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
38
transmitted systemic hypertension, autoregulatory disturbances, hyper perfusion
and hypoperfusion, and activation of the renin-angiotensin-aldosterone system
(RAAS). These factors themselves do not cause DKD, but rather feed and
reinforce the common pathogenic mechanisms in diabetes, which are increased
levels of growth factors, vasoactive hormones, cytokines and chemokines in the
kidney.
8. IMPORTANT STRUCTURAL CHANGES IN THE GLOMERULI
Despite the importance of the vascular endothelium in microvascular
complications, many researchers suggest that early changes in the glomerulus
are critical for the later development of glomerulosclerosis and nephron loss. The
most important of these changes may be glomerular podocyte dysfunction [21-
23]. They are highly specialized terminally differentiated cells that line the urinary
side of the glomerular basement membrane (GBM). Along with glomerular
endothelial cells, podocytes are responsible for maintaining the shape and
integrity of the GBM, its charge barrier, and the glomerular capillary circuit; all
functions that are impaired in diabetic glomeruli. The diabetic environment
induces "pathoadaptive" changes in podocytes, including cytoskeletal
rearrangements, differentiation, apoptosis and autophagy, manifested by
morphological expansion, retraction and flattening (known as loss), decreased
motility, increased formation of intercellular tight junctions, glomerular
hypertrophy, detachment and disruption.
9. GLOMERULAR BASEMENT MEMBRANE THICKNENING
One of the earliest and most characteristic glomerular changes in diabetes is a
homogeneous thickening of the GBM [24,25]. GBM thickening occurs in most
patients with diabetes usually within a few years of diagnosis. It is unclear
whether GBM thickening is a marker of podocyte or endothelial dysfunction or a
mediator of progressive DKD. Certainly, thickening-related changes in GBM
composition, charge, or architecture may contribute to the development of
albuminuria. GBM stiffening may also reduce pericapillary wall stretch and
compromise the sub podocyte state, facilitating glomerular injury through
hemodynamic mechanisms.
10. RENAL TUBULAR DYSFUNCTION AND FIBROGENESIS
Diabetes also negatively affects the kidney tubules [26]. At the onset of diabetes,
an increased glucose load delivered to the proximal tubule results in cortical
tubular maladaptive hypertrophy and hyperplasia with increased glucose
transport, which may facilitate glucose reabsorption and reduce glucose waste.
However, the consequence of this is that sodium transport in the macula densa is
reduced and tubulo-glomerular feedback is activated, leading to increased
glomerular pressure and hyperfiltration [27]. Chronic hyperglycemia and other
metabolic abnormalities associated with diabetes can cause progressive atrophy
of tubular epithelial cells. Such tubular dysfunction results in deficient uptake,
transcytosis, and/or lysosomal processing of filtered protein, which also
contributes to albuminuria.
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
39
11. COMPLEX HISTOPATHOLOGY OF DKD
The same clinical presentation of DKD can be associated with a heterogeneous
set of pathological features, including nodular or diffuse glomerulosclerosis,
tubulointerstitial fibrosis, tubular atrophy and renal arteriolar hyalinosis, alone or
in combination [28-30]. A histopathologic staging system has been proposed for
glomerular lesions. However, its prognostic utility is yet to be determined.
Routine renal biopsy is not clinically appropriate in routine practice and DKD
remains the clinical diagnosis for most patients with diabetes.
12. NATURAL COURSE
The classification of clinical stages of diabetic nephropathy is as follows [31]:
Stage 1: Hyperfiltration: From the onset of diabetes, glomerular filtration and
renal blood flow increase, and leads to renal enlargement (renomegaly). Urinary
albumin excretion rate (UAE) is usually less than 30 mg in 24 hours and blood
pressure is also normal. The first pathologic finding is GBM thickening. If the
initial glomerular filtration rate (GFR) is more than 150 ml per minute, it increases
the possibility of diabetic nephropathy.
Stage II: Microalbuminuria: As kidney disease progresses, UAE also develops.
This is also called latent or subclinical nephropathy. A standard strip test is often
negative at this stage. Changes in the degree of albuminuria over time are
associated with different risks of renal decline, so that patients with
microalbuminuria with increased, persistent, or decreased albumin burden are at
very high, moderate, and low risk of GFR decline. In type 1 diabetes, when
albuminuria increases, the incidence of other microvascular complications, such
as retinopathy and neuropathy, also increases. Hyperlipidemia, age, duration of
diabetes are also risk factors for microalbuminuria. Diagnosis at this stage is a
very good chance to avoid progression to clinical nephropathy or even back to
normoalbuminuric. However, without treatment, the risk of developing clinical
nephropathy is high in type 1 diabetes after 10-15 years (more than 75%) and in
type 2 diabetes after 15-20 years (20-40%).
Stage III: Macroalbuminuria: This stage is also called clinical nephropathy,
which appears about 10-20 years after the onset of diabetes (about 5-10 years
after the onset of microalbuminuria). Coronary artery disease and
cerebrovascular events also clearly increase in this stage compared to the
previous stage. About 75 percent of patients at this stage have high blood
pressure. Controlling blood pressure in type 2 diabetics with a history of
hypertension is more difficult. At this stage, a standard dipstick is positive for
proteinuria, and urinary albumin excretion is greater than 300 mg in 24 hours.
After that, the GFR decreases, about 10-12 ml per year. When clinical
nephropathy appears and in the absence of therapeutic measures, kidney
function gradually deteriorates and proteinuria increases, which can reach the
nephrotic range. Diabetic retinopathy is very useful in confirming the diagnosis of
diabetic nephropathy. Hypertension and proteinuria independently lead to
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
40
decreased GFR and progression to ESRD. 20% of patients with type 2 diabetes
before the development of clinical nephropathy and 40% of patients with clinical
nephropathy have renal vascular disease. Tubulointerstitial fibrosis also begins at
this stage, and if its extent and severity are greater, kidney damage and
prognosis are worse.
Stage IV: End stage kidney disease Almost 20-30 years after the onset of
diabetes, about 10 years after the onset of clinical nephropathy, the patient
reaches this stage [32]. The risk with type 1 diabetes is higher than type 2
diabetes. However, the prevalence of type 2 diabetes is 9 times higher than type
1 diabetes, so most diabetics with ESRD have type 2 diabetes. Under these
conditions, the death rate of diabetic patients is higher than that of non-diabetic
patients [33-35]. In addition, cardiovascular diseases are higher in these patients.
At this stage, the incidence of diabetic foot ulcers is higher, sometimes up to
25% [36,37]. In the UKPDS report, the annual chance of diagnosis from diabetes
to microalbuminuria, from microalbuminuria to overt nephropathy, and from overt
nephropathy to elevated serum creatinine or conversion to renal replacement
therapy are 2%, 2.8%, and 2.3%, respectively.
13. RISK FACTORS FOR DKD
Several different factors contribute to the development of CKD in diabetics. Some
of these factors, including hyperglycemia, hypertension, weight gain, and
dyslipidemia, are potentially modifiable with optimized diabetes treatment [38]. In
addition, extensive clinical data show that intensive diabetes management
significantly reduces the incidence of albuminuria, renal failure as well as ESRD.
Indeed, the significant reduction in the incidence of CKD over the past 30 years
is largely due to improvements in diabetes management.
14. POOR GLYCEMIC CONTROL
The most important risk factor for CKD is hyperglycemia. Although there are
structural similarities to other kidney diseases, the DKD phenotype is essentially
only observed in the presence of elevated glucose levels [39, 40]. A definitive
prospective clinical study by Jean Pirart and colleagues in Belgium showed
unequivocally that the degree and duration of hyperglycemia were associated
with microvascular complications, including CKD. Subsequent randomized
controlled trials have confirmed this causal relationship in both type 1 and type 2
diabetes. However, although normal glucose levels, such as glycated
hemoglobin (HbA1c), are associated with microalbuminuria, it is also clear that
many patients with poor blood glucose levels do not develop kidney
complications, while others do despite intensive interventions and dedicated
control measures. Previous episodes of poor glucose balance, even before
diagnosis, can also have a long-term effect in the kidney, and therefore DKD risk
may not be represented by current or recent HbA1c levels. This phenomenon
was known as "metabolic memory", "metabolic karma" or the "inheritance effect"
[41]. The physiological mechanism or mechanisms responsible for metabolic
karma remain poorly defined, but may involve epigenetic programming,
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
41
remodeling, and permanent post-translational modifications such as enhanced
glucose end products. Understanding the molecular basis of the metabolic
inheritance of diabetes will certainly add new targets for intervention to reduce
the burden of CKD in diabetics.
15. HIGH BLOOD PRESSURE
Increased blood pressure is a major risk factor for CKD in both type 1 and type 2
diabetes. In people with type 1 diabetes, blood pressure levels are usually normal
at diagnosis, but increase near microalbuminuria [42]. In type 2 diabetes, other
factors influence the appearance and severity of hypertension, which can
precede or follow chronic kidney disease for several years. The importance of
hypertension in the pathogenesis of renal injury can be partially explained by the
loss of renal autoregulation in diabetes, in which systemic pressure is transmitted
directly to the vulnerable glomerular capillaries. Thus, there is no specific
threshold above which the specific risk of CKD can be expressed or below which
the therapeutic effect of blood pressure control on the development of
albuminuria can be ignored in diabetic patients.
16. LIPID ABNORMALITIES
Dyslipidemia is another important risk factor for the development of chronic
diseases of diabetes [43]. Specifically, high levels of triglycerides, low-density
lipoprotein cholesterol, apolipoprotein-B-100, or low high- density lipoprotein
(HDL) cholesterol are independently associated with the development of CKD in
both type 1 and type 2 diabetes. However, conventional lipid and lipoprotein
measurements do not fully account for the complex lipid and lipoprotein changes
associated with diabetes and/or CKD. For example, not only can HDL lose its
vascular protective, antioxidant, and anti-inflammatory properties in CKD, but
dysfunctional HDL can also be directly pathogenic. Lipidomics has been used to
create a "lipid fingerprint" linked to diabetes complications. However, it remains
unclear which lipids or lipoproteins are most important in the pathogenesis of
diabetic CKD.
17. INSULIN RESISTANCE
Insulin resistance is also independently associated with CKD, in addition to
indirect associations with glucose, blood pressure, body weight, and lipid
regulation [44]. Insulin-sensitizing measures (eg, thiazolidinedione therapy,
exercise, and weight loss) reduce albuminuria in addition to metabolic control.
Decreased insulin sensitivity also causes changes in glucose metabolism in
kidney cells. At the same time, increased insulin signaling due to compensatory
hyperinsulinemia in the setting of selective insulin resistance can contribute to
vasoreactivity, angiogenesis, fibrogenesis and other pathways involved in
progressive kidney disease and atherogenesis.
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
42
18. OBESITY
CKD occurs more frequently and progresses more rapidly in obese diabetics
than in their normal-weight counterparts [45]. This is one of the main reasons
why the cumulative incidence of CKD is higher in type 2 diabetes than in type 1
diabetes. Obesity adversely affects key risk factors associated with CKD,
including lipid, blood pressure, and glucose balance, and promotes insulin
resistance. Obesity also has direct effects on the kidney, including changes in
intraglomerular hemodynamics, increased sympathetic activity, hypertension,
systemic inflammation, endothelial dysfunction, altered expression of growth
factors, and renal compression associated with visceral obesity. Indeed, even
in the absence of diabetes, obesity can be associated with increased albuminuria
and its severity, and obesity-related glomerulopathy has been widely described.
19. HEPATITIS C VIRUS (HCV) INFECTION AND CKD
HCV infection is more common among CKD patients than in the general
population [46,47]. According to recent research, CKD patients with HCV
infection had a faster loss of renal function and a higher chance of developing
ESRD. The important question of whether treating to achieve a sustained
virologic response, defined as an undetectable viral load 12 weeks after
completion of treatment (SVR12), would slow the rate of decline in GFR was
raised by a study which demonstrated that HCV-infected patients with CKD had
an increased mortality and an accelerated rate of progression to ESRD [48].
Most patients with mixed cryoglobulinemia and many histological types of
glomerular damage, including membranoproliferative and membranous GN, have
been linked to HCV infection as the underlying cause of these conditions [49-55].
20. ESRD IN DIABETES
Globally, 80% of ESRD cases are caused by diabetes, hypertension or a
combination of these [56]. Compared to adults without diabetes, the incidence of
ESRD is up to 10 times higher in diabetics. However, only a limited number of
patients with CKD associated with diabetes ever receive renal replacement
therapy, as 78% of these individuals live in low- and middle-income countries
with limited resources, coverage, and access to dialysis and kidney
transplantation. The proportion of diabetes related ESRD varies greatly in
different parts of the world. In 2014, between 5% and 66% of new cases of
ESRD were mainly due to diabetes [57-59]. The highest proportion was in
Singapore, Malaysia and the Jalisco region of Mexico, and the lowest in Norway,
Romania and Iceland. In most countries at the upper end of this distribution, the
incidence of diabetes related ESRD has increased dramatically over the past
decade. Between 2001 and 2015, the highest growth was in Thailand at 1,448%,
Russia at 981% and the Philippines at 378% [60]. Because these figures are
reported for the entire state- based population, they primarily reflect the
increasing prevalence of diabetes in those populations, partly due to a shift in the
burden of disease from infections to chronic lifestyle diseases and increased life
expectancy. The prevalence of treated ESRD was highest, ranging from 1568 to
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
43
3219 per million inhabitants, in some of the Asian countries such as Japan,
Singapore, and in the United States [60]. Although the incidence of treated ESRD
has been fairly stable or declining in many countries in recent years, the
incidence has steadily increased in all 32 countries that reported data between
2001 and 2014. In the United States, 44% of new cases of ESRD are due to
diabetes (either type 1 or type 2), highest in non-Hispanic blacks and lowest in
non-Hispanic whites [61-63]. Detailed quantitative data on the trend of diabetes-
related ESRD and its association with other complications of diabetes in the
United States are presented in an analysis of several nationally representative
databases (NHIS, National Hospital Discharge Survey, US Renal Data System,
and National Vital Statistics System) in the years 1990-2010. Deaths from heart
attack, stroke, amputation, end-stage disease and hypoglycemia were reduced in
the diabetic population (primarily type 2 diabetes). The greatest reduction is for
cardiovascular events and the least for ESRD. In the 1990s, the incidence of
diabetes related ESRD was low because CVD morbidity and mortality were
important competing events. The decline in these competing causes over the
next decade allowed people with diabetes to live long enough to develop ESRD,
which explains why the decline in ESRD is smaller. However, the study found a
statistically significant decrease in diabetes related ESRD in all age groups after
2000, consistent with significant progress in the adoption of reno-protective
therapies nationwide.
21. SCREENING
Annual screening of all diabetic patients is recommended to detect abnormal
and/or variable levels of albuminuria and renal function (ie, eGFR) so that early
renoprotective therapy can be initiated [64]. Early morning spot urine is sufficient
for screening and monitoring and is convenient for the patient. To account for the
large intraday variability (30-40%), 2 of the 3 spot urine samples must be
collected within 3-6 months to confirm the diagnosis. A 24-hour urine collection
has been considered the gold standard for evaluating albuminuria and can
provide additional information on sodium and protein intake, but a complete
urine collection is often difficult for the patient, so this method is usually limited to
patients with established diabetic kidney disease. It should be noted that urinary
albumin excretion may increase independently of kidney disease due to factors
such as heavy exercise within 24 hours, severe UTI, menstruation, heart failure,
and marked hyperglycemia [65]. Another clinical variable assessed in diabetic
kidney disease screening is eGFR using creatinine-based formulas such as the
Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation. The
"natural" course of untreated diabetic nephropathy shows a continuous annual
decline in eGFR of 2-20 ml/min/1.73 m2 (average 12 ml/min/1.73 m2), but
effective treatment focuses on glycemic, blood pressure control, renin-
angiotensin system (RAS) inhibition, lowering blood cholesterol and improving
lifestyle factors can limit progression to 2-5 mL/min/1.73 m2 per year, indicating
the importance of screening and interventions.
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
44
22. MANAGEMENT
Treatment of hypertension: Observational studies in the general population
have shown a strong and linear association between blood pressure and
cardiovascular disease risk, with the risk of cardiovascular events doubling for
every 20/10 mmHg increase in systolic/diastolic blood pressure above 115/75
mmHg [66]. In addition to directly affecting CV risk, hypertension contributes to
kidney damage in diabetes and albuminuria; therefore, it is an important target
for management. According to the Kidney Disease Outcomes Initiative clinical
practice guidelines for blood pressure control in CKD, a goal of less than 140/90
mmHg was recommended in those diabetic patients without significant
albuminuria (30 mL/min/1.73 m2).
23. RAAS INHIBITION AND ALBUMINURIA
By opposing the renin-angiotensin-aldosterone system (RAAS); angiotensin-
converting enzyme inhibitors (ACEs) and angiotensin receptor blockers lower
blood pressure and albuminuria [67]. The Irbesartan Diabetic Nephropathy Trial
(IDNT) showed that irbesartan protects against the progression of nephropathy in
patients with type 2 diabetes regardless of the drop in blood pressure [68].
The Telmisartan Alone and in Combination with Ramipril Global Endpoint Trial
(ONTARGET) in diabetic and non-diabetic patients showed that although the
combination of ramipril and telmisartan reduced proteinuria compared with either
agent alone, the time to initiation of dialysis was not delayed [69,70]. There are
enough studies to compare the effectiveness of ACE inhibitors and angiotensin
receptor blockers in this situation. These results are already a cornerstone of the
treatment of diabetic nephropathy and can be used to support the routine use of
RAAS inhibitors in the treatment of patients with type 2 diabetes, regardless of
baseline albuminuria.
24. TREATMENT OF HYPERGLYCEMIA
Hyperglycemia, a hallmark of diabetes, is an important therapeutic goal in all
diabetic patients. As shown in important studies such as Epidemiology of
Diabetes Interventions and Complications (EDIC) and UKPDS, tight glycemic
control reduces the risk of microvascular complications, including nephropathy, in
patients with type 1 and type 2 diabetes [71]. Current evidence supports therapy
with a target HbA1c of 7.0% to slow the onset of microvascular complications,
including nephropathy. Of note, no prospective randomized clinical trial has
evaluated the effect of glycemic control on health outcomes in patients with CKD
stages 3-5. Because patients with CKD are more prone to hypoglycemia than
patients with preserved eGFR, it remains unclear whether the same HbA1c
target is optimal for this population.
25. METFORMIN
Metformin remains the first-line antihyperglycemic treatment for most patients
with T2D because of its low cost, high efficacy, and low risk of hypoglycemia [72].
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
45
In addition, it has weight- and lipid-lowering properties, as well as beneficial
effects on CV mortality. Metformin is mainly excreted by the kidneys. Despite
limited data, the increased risk of lactic acidosis in patients with lower eGFR has
limited its use to patients with eGFR >30 mL/min/1.73 m2.
26. INHIBITORS OF SODIUM-GLUCOSE COTRANSPORTER-2
(SGLT-2I)
SGLT2i is now a widely used antihyperglycemic drug for type 2 diabetes [73].
Several CV outcome studies, including the Empagliflozin Cardiovascular
Outcome Event (EMPA-REG OUTOME) study and the Canagliflozin
Cardiovascular Assessment Study (CANVAS), have shown that SGLT2i provides
significant renal benefits in addition to CV benefits [74]. The CANVAS Renal
study reported a corresponding 40% reduction in the renal composite outcome
(persistent decline in eGFR, need for renal replacement therapy, or death from
renal causes) [75,76]. SGLT2i reduces renal glucose reabsorption, resulting in
osmotic diuresis and plasma volume depletion. Approximately one-third of
SGLT2i-treated patients experience a reversible decline in eGFR of more than
10%. Although they are relatively weak glucose-lowering agents, they have the
added benefit of lowering blood pressure and weight and do not cause
hypoglycemia. Possible side effects include an increased risk of fluid depletion,
genital and urinary tract infections, perineal necrotizing fasciitis, and euglycemic
ketoacidosis.
27. GLUCAGON-LIKE PEPTIDE-1 RECEPTOR AGONISTS
In T2D, GLP-1 RA represents a family of injectable antihyperglycemic drugs [77].
In particular, liraglutide, semaglutide and dulaglutide have shown significant CV
and renal benefits in large CV outcome trials, particularly in patients with
established CVD or at high risk. In the LEADER (Liraglutide Effect and Action in
Diabetes: Evaluation of Cardiovascular Outcome Results) trial, liraglutide
reduced cardiovascular death by 22% and all-cause death by 15% compared to
placebo [78]. Long-term follow-up data in patients with DKD also showed a 22%
reduction in combined nephritis, mainly due to a lower incidence of severely
elevated albuminuria. Several GLP-1 receptor agonists, including liraglutide,
semaglutide have low renal clearance and hence are safer to use even in
patients with advanced diabetic kidney disease. Major guidelines, including those
of the American Diabetes Association (ADA) and the European Association for
the Study of Diabetes, now recommend the use of GLP-1 RAs to reduce
cardiovascular risk in T2D, regardless of glucose control. In clinical practice, the
most common side effects of GLP-1 RA are gastrointestinal symptoms, injection
site reactions, and increased heart rate. GLP-1 RAs should also be avoided in
patients at risk of developing medullary thyroid tumors or with a history of acute
pancreatitis.
28. TREATMENT OF DYSLIPIDEMIA
Dyslipidemia, a highly modifiable risk factor for CV events in the general
population, is common in patients with diabetes and CKD [79]. Data showing a
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
46
reduction in cardiovascular events due to low-density lipoprotein (LDL)
cholesterol lowering by in CKD patients was originally derived from multiple post
hoc analysis of large randomized controlled trials with enough CKD patients. It
used to be recommended for diabetics and people with CKD stages 1-4, with
statins as first-line agents, to achieve serum LDL cholesterol levels below 100
mg/dL or optionally below 70 mg/dL. However, some targets of LDL cholesterol
treatment were omitted from the recent recommendations for lipid management
in patients with CKD in the 2013 Clinical Practice Global Outcomes for the
Improvement of Kidney Disease. These recommendations are consistent with
recent prevention guidelines from the American Heart Association and the
American College of Cardiology, which recommended that the decision to initiate
cholesterol-lowering therapy (especially statin therapy) be based on the absolute
risk of coronary events [80]. Thus, statin therapy should be considered in almost
all patients with diabetes and chronic diseases due to their high CVD risk and the
pleiotropic effects of statins. It is not clear whether patients using statins should
stop using them when starting dialysis.
29. LIFESTYLE CHANGES
All patients with diabetic nephropathy should receive recommendations for
lifestyle changes that can reduce their risk of progression and development of
cardiovascular disease [81]. For patients who smoke, tobacco cessation is
essential, as it not only significantly and directly reduces the risk of
cardiovascular disease but can also slow the progression of early diabetic
nephropathy. In addition to following a diabetic diet, patients with nephropathy
have traditionally been advised to limit salt and protein intake. Reducing salt
intake by 8.5 g per day has been shown to lower blood pressure by 7/3 mmHg,
similar to monotherapy [82]. However, it should be noted that a recent
prospective observational study in patients with type 1 diabetes who did not have
ESRD showed that the relationship between salt intake and mortality as
assessed by urinary sodium excretion is non- linear, with decreased survival in
patients with the highest and lowest levels of excretion regardless of kidney
disease. Clinical trials are needed to assess the effect of dietary salt restriction
on mortality in this setting. Dietary protein restriction may improve eGFR in
patients with diabetic kidney disease, although the quality of the evidence is not
high. Finally, combining lifestyle changes with intensive treatment can
significantly reduce the risk of cardiovascular disease and prevent the
progression of microvascular disease.
30. MANAGEMENT OF CVD
Prevention and treatment of diabetic nephropathy and reduction of risk of
cardiovascular diseases Considering the high mortality rate of patients with
diabetic nephropathy, primary prevention of its development and efforts to
prevent the progression of the disease after detection are extremely important
[83]. Unfortunately, CKD is often unrecognized by patients and providers. In
addition, patients diagnosed with CKD are less likely to achieve an adequate
change in CVD risk factor than the general population. The key to improving
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
47
outcomes in this vulnerable patient population is simply to increase awareness of
these issues and intervene as early as possible.
31. CONCLUSION AND FUTURE RECOMMENDATIONS
Despite significant advances in the pharmacological management of patients
with diabetes, available DKD therapies can slow the decline in GFR, and a
significant CV risk remains. Research into potential new therapeutic targets for
diabetic kidney disease is currently active and brings a lot of expectation and
optimism to this field. New targets for therapeutic interventions include drugs that
inhibit the formation and function of Advanced Glycation End Products (AGEs) or
AGE receptors, drugs that target oxidative stress, inflammatory cytokines or
fibrosis. The role of microRNAs in the pathogenesis of DKD is an emerging field
and may also provide new therapeutic approaches. Cell therapies for
regenerating blood vessels within the kidney are in the early stages of clinical
trials. New insights into the molecular mechanisms underlying the origin and
progression of DKD are emerging from extensive genetic and molecular studies in
experimental models and humans.
COMPETING INTERESTS
Authors have declared that no competing interests exist.
REFERENCES
1. Digsu N Koye, Dianna J Magliano, Robert G Nelson, Meda E Pavkov. The
Global Epidemiology of Diabetes and Kidney Disease. Adv Chronic
Kidney Dis. 2018;25(2)121-132.
2. Palleti, Sujith Kumar, Sreekant Avula, and Sandesh Dewan. Kidney: A
Review on End Stage Renal Disease, Dialysis and Transplant. Int Clinc
Med Case Rep Jour. 2023;2(10):1-8.
3. Darin E Olson, Mary K Rhee, Kirsten Herrick, David C Ziemer, Jennifer G
Twombly, Lawrence S Phillips. Screening for Diabetes and Pre-Diabetes
with Proposed A1C-Based Diagnostic Criteria. Diabetes Care.
2010;33(10):2184-9.
4. Raza A Khan, Nidhi Patel, Atunde Folajimi, Bansari Raveena Bai, Vrushak
Patel, Praveen Kumar Komminni, et al. Comparison of the Efficacy and
Safety of Metformin-Based Combination Therapy Versus Metformin
Alone in Children and Adolescents With Type 2 Diabetes Mellitus: A Meta-
Analysis. Cureus. 2023;15(2):e35014.
5. Forouhi, Nita Gandhi, and Nicholas J. Wareham. Epidemiology of
Diabetes. Medicine (Abingdon). 2014;42(12):698-702.
6. Lena M Thorn, Daniel Gordin, Valma Harjutsalo, Stefanie Hägg, Robert
Masar, Markku Saraheimo, et al. The Presence and Consequence of
Nonalbuminuric Chronic Kidney Disease in Patients with Type 1 Diabetes.
Diabetes Care. Diabetes Care. 2015;38(11):2128-33.
7. Palleti, Sujith, et al. “Int Clinc Med Case Rep Jour (ICMCRJ) 2023 |
Volume 2 | Issue 11 Citation: Sujith Kumar Palleti, Sreekant Avula, Roopa
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
48
Naik. Chronic Kidney Disease.” International Clinical and Medical Case
Reports Journal Review Article. 2023;2(11):1–15,
8. Keith P McCullough, Hal Morgenstern, Rajiv Saran, William H
Herman, Bruce M Robinson. Projecting ESRD Incidence and Prevalence
in the United States through 2030. J Am Soc Nephrol. 2019;30(1):127-
135.
9. Rakesh Pilla, Sujith Kumar Palleti, Renuka Rayana, Satish Reddy Skss,
Aminah Abdul Razzack, Sruti Kalla. Glycated haemoglobin (HbA1c)
variations in nondiabetics with nutritional anemia. Cureus.
2020;12(11):e11479.
10. Paul Muntner, Barry R Davis, William C Cushman, Sripal Bangalore,
David A Calhoun, Sara L Pressel, et al. Treatment-Resistant
Hypertension and the Incidence of Cardiovascular Disease and End-
Stage Renal Disease. Hypertension. 2014;64(5):1012-21.
11. Muhammad Ali Sameer, Bilal Aziz Malik, Muhammad Obaid Ullah
Choudry, Muhammad Shoaib Anwar, Muhammad A Nadeem, Fizza
Mahmood, et al. Comparison of Coronary Artery Bypass Grafting
Combined With Mitral Valve Repair Versus Coronary Artery Bypass
Grafting Alone in Patients With Moderate Ischemic Mitral Regurgitation: A
Meta-Analysis. Cureus. 2023;15(4). e37238.
12. Hans-Joachim Anders, Tobias B Huber, Berend Isermann, Mario Schiffer.
CKD in Diabetes: Diabetic Kidney Disease versus Nondiabetic Kidney
Disease. Nat Rev Nephrol. 2018;14(6):361-377.
13. Duran-Salgado, Montserrat B, and Alberto F Rubio-Guerra.
Diabetic Nephropathy and Inflammation. World J Diabetes. 2014;5(3):
393-8.
14. Sungmi Park, Hyeon-Ji Kang, Jae-Han Jeon, Min-Ji Kim, In-Kyu Lee.
Recent Advances in the Pathogenesis of Microvascular Complications in
Diabetes. Arch Pharm Res. 2019;42(3):252-262.
15. Jithin Karedath, FNU Avanteeka, Muhammad Nouman Aslam, Ahmad
Nadeem, Rao Ahmed Yousaf, Sandesh Shah, et al. Comparison of
Effectiveness and Safety of Low-Dose Versus Standard-Dose Intravenous
Recombinant Tissue Plasminogen Activator in Patients With Acute
Ischemic Stroke: A Meta-Analysis. Cureus. 2023;15(2).
16. Dung V Nguyen, Lynn C Shaw, Maria B Grant. Inflammation in the
Pathogenesis of Microvascular Complications in Diabetes. Front
Endocrinol (Lausanne). 2012;3:170.
17. Mona Sedeek, Rania Nasrallah, Rhian M Touyz, Richard L Hébert.
NADPH Oxidases, Reactive Oxygen Species, and the Kidney: Friend and
Foe. J Am Soc Nephrol . 2013;24(10):1512-8.
18. Abshiro H Mayow, Fatima Ahmad, Muhammad Sohaib Afzal,
Muhammad Usama Khokhar, Daneyal Rafique, et al. A Systematic
Review and Meta-Analysis of Independent Predictors for Acute
Respiratory Distress Syndrome in Patients Presenting With Sepsis.
Cureus. 2023;15(4).
19. Matthew T James, Brenda R Hemmelgarn, Marcello Tonelli. Early
Recognition and Prevention of Chronic Kidney Disease. Lancet.
2010;375(9722):1296-309.
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
49
20. Toth-Manikowski, Stephanie, and Mohamed G. Atta. Diabetic Kidney
Disease: Pathophysiology and Therapeutic Targets. J Diabetes Res.
2015;2015:697010.
21. Luigi Gnudi, Richard J M Coward, David A Long. Diabetic Nephropathy:
Perspective on Novel Molecular Mechanisms. Trends Endocrinol Metab.
2016;27(11):820-830.
22. Sai Kiran Kondabolu, Yasitha Kakarlapudi, Haider Malik, Hamza Malik,
Saima Khan, Praveen Kumar Komminni, et al. Beneficial Impacts of
Fenoldopam on Patients With or at Risk for Acute Renal Failure and
Undergoing Surgery: A Meta-Analysis of Randomized Clinical Trials.
Cureus. 2023;15(2):e34584.
23. Mohsen Pourghasem, Hamid Shafi, Zahra Babazadeh. Histological
Changes of Kidney in Diabetic Nephropathy. Caspian J Intern Med.
2015;6(3):120-7.
24. Behzad Najafian, Charles E Alpers, Agnes B Fogo. Pathology of Human
Diabetic Nephropathy. Contrib Nephrol. 2011;170:36-47.
25. Marshall, Caroline B. Rethinking Glomerular Basement Membrane
Thickening in Diabetic Nephropathy: Adaptive or Pathogenic? Am J
Physiol Renal Physiol. 2016;311(5):F831-F843.
26. Suyan Duan, Fang Lu, Dandan Song, Chengning Zhang, Bo Zhang,
Changying Xing, et al. Current Challenges and Future Perspectives of
Renal Tubular Dysfunction in Diabetic Kidney Disease. Front Endocrinol
(Lausanne). 2021;12:661185.
27. M C Thomas, W C Burns, M E Cooper. Tubular Changes in
Early Diabetic Nephropathy. Adv Chronic Kidney Dis. 2005;12(2):177-
86.
28. Paola Fioretto, Michael Mauer. Histopathology of Diabetic Nephropathy.
Semin Nephrol. 2007;27(2):195-207.
29. Sujith K. Palleti, Hanna Larson, Sreekant Avula, Maria M. Picken,
Anuradha Wadhwa. Subarachnoid Hemorrhage in Autoimmune Vasculitis:
A Rare Presentation of Systemic Lupus Erythematosus- Antineutrophil
Cytoplasmic Autoantibody-Associated Vasculitis Overlap Syndrome.
Cureus. 2023;15(5):2023.
30. Sujith K. Palleti, Maria M. Picken, Anuradha Wadhwa. Sjögren’s
Syndrome Overlap with Lupus Nephritis: A Case Report and Literature
Review of a Rare Entity. Cureus. 2023;15(5).
31. C E Mogensen, C K Christensen, E Vittinghus. The Stages in Diabetic
Renal Disease: With Emphasis on the Stage of Incipient Diabetic
Nephropathy. Diabetes. 1983;32 Suppl 2:64-78.
32. F L Brancati, P K Whelton, B L Randall, J D Neaton, J Stamler, M J Klag.
Risk of End-Stage Renal Disease in Diabetes Mellitus: A Prospective
Cohort Study of Men Screened for MRFIT. Multiple Risk Factor
Intervention Trial. JAMA. 1997;278(23):2069-74.
33. Caroline S Fox, Kunihiro Matsushita, Mark Woodward, Henk J G Bilo,
John Chalmers, Hiddo J Lambers Heerspink,, et al. Associations of
Kidney Disease Measures with Mortality and End-Stage Renal Disease
in Individuals with and without Diabetes: A Meta-Analysis. Lancet.
2012;380(9854):1662-73.
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
50
34. Sujith K Palleti, Santhoshi R Bavi, Margaret Fitzpatrick, Anuradha
Wadhwa. First case report of Sphingobium lactosutens as a human
pathogen causing peritoneal dialysis-related peritonitis. Cureus.
2022;14(7):e27293.
35. Sujith K. Palleti, Santhoshi R. Bavi, Margaret Fitzpatrick, Anuradha
Wadhwa. Study about Sphingobium Lactosutens: A Human Pathogen
Causing Peritoneal Dialysis Related Peritonitis. Perspective of Recent
Advances in Medical Research. BP International. 2023:31-35.
36. Jeffcoate William J, Keith G Harding. Diabetic Foot Ulcers. Lancet
(London, England). 2003; 361(9368):1545-51.
37. Zaid Munir, Muhammad Akash, Fnu Jaiprada, Bilal Abu Tarboush, Osama
Ijaz, Anan Bseiso, et al. Evaluation of the Effects of Extracorporeal
Shockwave Therapy in Patients With Peripheral Arterial Disease:
A Meta-Analysis of Randomized Control Trials. Cureus. 2023;
15(2):e34729.
38. Kazancioğlu Rumeyza. Risk Factors for Chronic Kidney Disease: An
Update. Kidney Int Suppl (2011). 2013;3(4):368-371.
39. K Sharma, F N Ziyadeh. Hyperglycemia and Diabetic Kidney Disease: The
Case for Transforming Growth Factor–β as a Key Mediator. Diabetes.
1995;44(10):1139-46.
40. Benaicha, Karima, et al. Efficacy of Molnupiravir for the Treatment of
Mild or Moderate COVID-19 in Adults: A Meta-Analysis. Cureus.
2023;15(5).
41. Kato Mitsuo, Rama Natarajan. Epigenetics and Epigenomics in Diabetic
Kidney Disease and Metabolic Memory. Nat Rev Nephrol.
2019;15(6):327-345.
42. Bruce Horowitz, Dana Miskulin, Philip Zager. Epidemiology of
Hypertension in CKD. Adv Chronic Kidney Dis. 2015;22(2):88-95.
43. Kaysen GA. Hyperlipidemia in Chronic Kidney Disease. Int J Artif Organs.
2007;30(11):987-992.
44. Luigi Gnudi, Richard J M Coward, David A Long. Diabetic Nephropathy:
Perspective on Novel Molecular Mechanisms. Trends Endocrinol Metab.
2016;27(11):820-830.
45. Christine Maric, John E Hall. Obesity, Metabolic Syndrome and Diabetic
Nephropathy. Contrib Nephrol. 2011;170:28-35.
46. Marco Ladino, Fernando Pedraza, David Roth. Hepatitis c Virus Infection
in Chronic Kidney Disease. J Am Soc Nephrol. 2016;27(8):2238-2246.
47. Fareeha Naseer Syed, Nadia Kashif, Mubashir Razzaq Khan, Fayaz
Ahmed Memon, Muhammad Zubair Jabbar, Sujith Kumar Palleti. Hepatitis
C Virus Infection in Patients with Beta Thalassemia after Multiple
Transfusions at a Tertiary Care Hospital. Pakistan Journal of Medical &
Health Sciences 2023;17(1):363-363.
48. Marco Ladino, Fernando Pedraza, David Roth. Hepatitis c Virus Infection
in Chronic Kidney Disease. J Am Soc Nephrol. 2016;27(8):2238-46.
49. Ron T Gansevoort, Ricardo Correa-Rotter, Brenda R Hemmelgarn,
Tazeen H Jafar, Hiddo J Lambers Heerspink, Johannes F Mann, et al.
Chronic Kidney Disease and Cardiovascular Risk: Epidemiology,
Mechanisms, and Prevention. Lancet. 2013;382(9889):339-52.
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
51
50. Kashyap Kela, Chaitanya Rojulpote, Ajinkya Buradkar, Sreekant Avula,
Vijay Pratap Singh, Sujithraj Dommaraju, et al. AN UNUSUAL CAUSE of
ABNORMAL ECHODENSITY in the RIGHT ATRIUM. J Am Coll Cardiol.
2021;77(18_Supplement_1) 2698.
51. Daniel E Weiner, Sayed Tabatabai, Hocine Tighiouart, Essam Elsayed,
Nisha Bansal, John Griffith, et al. Cardiovascular Outcomes and All-Cause
Mortality: Exploring the Interaction between CKD and Cardiovascular
Disease. Am J Kidney Dis. 2006;48(3):392-401.
52. Karthik Gonuguntla, Komal Ejaz, Chaitanya Rojulpote, Ayesha Shaik,
Abhijit Bhattaru, Tapan Buch, et al. A nationwide cohort analysis to
determine the prevalence of sinus node dysfunction and rates of
pacemaker implantation in systemic lupus erythematosus. Am J
Cardiovasc Dis . 2021;11(4):478-483.
53. Collins Allan J. Cardiovascular Mortality in End-Stage Renal Disease. Am
J Med Sci. 2003;325(4):163-7.
54. Abdulmaleek Idanesimhe Sado, Muhammad S Afzal, Lavanya
Kannekanti, Hrushikesh Reddy Pamreddy, Jorge Pimentel Campillo,
Vaishnavi Kandukuri, et al. A Meta-Analysis on Predictors of Mortality
Among Patients Hospitalized for Acute Exacerbation of Asthma. Cureus.
2023;15(2):e35225.
55. John M Flack, Bemi Adekola. Blood Pressure and the New ACC/AHA
Hypertension Guidelines. Trends Cardiovasc Med. 2020;30(3):160-164.
56. Seyed Bahman Ghaderian, Fatemeh Hayati, Shokouh Shayanpour, Seyed
Seifollah Beladi Mousavi. Diabetes and End-Stage Renal Disease; a
Review Article on New Concepts. J Renal Inj Prev. 2015;4(2):28-33.
57. Digsu N Koye, Dianna J Magliano, Robert G Nelson, Meda E Pavkov. The
Global Epidemiology of Diabetes and Kidney Disease. Adv Chronic Kidney
Dis. 2018;25(2):121-132.
58. Sujith K Palleti, Anuradha Wadhwa, Abdul H Rasheed. The First Case of
Ceftazidime-Induced Antibiomania: A Case Report and Literature Review
Cureus. 2022;14(12):e32904.
59. Keith P McCullough, Hal Morgenstern, Rajiv Saran, William H
Herman, Bruce M Robinson. Projecting ESRD Incidence and Prevalence
in the United States through 2030. J Am Soc Nephrol. 2019;30(1):127-
135.
60. Maria Narres, Heiner Claessen, Sigrid Droste, Tatjana Kvitkina, Michael
Koch, Oliver Kuss, et al. The Incidence of End-Stage Renal Disease in the
Diabetic (Compared to the Non-Diabetic) Population: A Systematic
Review. PLoS One. 2016;11(1):e0147329.
61. Patrick Albertus, Hal Morgenstern, Bruce Robinson, Rajiv Saran. Risk of
ESRD in the United States. Am J Kidney Dis . 2016;68(6):862-872.
62. Palleti Sujith K, Anuradha Wadhwa, Abdul H. Rasheed. Ceftazidime-
induced Antibiomania: A Rare Case Study and Review of Literature.
Research Highlights in Disease and Health Research Vol. 2023:56-61.
63. Bethany J Foster, Mark M Mitsnefes, Mourad Dahhou, Xun Zhang,
Benjamin L Laskin. Changes in Excess Mortality from End Stage Renal
Disease in the United States from 1995 to 2013. Clin J Am Soc Nephrol.
2018;13(1):91-99.
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
52
64. Jorge L Gross, Mirela J de Azevedo, Sandra P Silveiro, Luís Henrique
Canani, Maria Luiza Caramori, Themis Zelmanovitz. Diabetic
Nephropathy: Diagnosis, Prevention, and Treatment. Diabetes Care.
2005;28(1):164-76.
65. Samsu Nur. Diabetic Nephropathy: Challenges in Pathogenesis,
Diagnosis, and Treatment. Biomed Res Int. 2021;2021:1497449.
66. Elaine Ku, Benjamin J Lee, Jenny Wei, Matthew R Weir. Hypertension in
CKD: Core Curriculum 2019. Am J Kidney Dis. 2019;74(1):120-131.
67. Hala Yamout, Ivana Lazich, George L Bakris. Blood Pressure,
Hypertension, RAAS Blockade, and Drug Therapy in Diabetic Kidney
Disease. Adv Chronic Kidney Dis. 2014;21(3):281-6.
68. Lawrence G Hunsicker, Robert C Atkins, Julia B Lewis, Gregory Braden,
Paul J Champion de Crespigny, Giacomo DeFerrari, et al. Impact of
Irbesartan, Blood Pressure Control, and Proteinuria on Renal Outcomes in
the Irbesartan Diabetic Nephropathy Trial. Kidney Int Suppl.
2004;(92):S99-101.
69. Peter Sleight, Josep Redon, Paolo Verdecchia, Giuseppe Mancia, Peggy
Gao, Robert Fagard, Helmut Schumacher, et al. Prognostic Value of
Blood Pressure in Patients with High Vascular Risk in the Ongoing
Telmisartan Alone and in Combination with Ramipril Global Endpoint Trial
Study. J Hypertens. 2009;27(7):1360-9.
70. Robin Rauniyar, Rahul Rauniyar, Ankita Agrawal, Shikha Yadav, Sreekant
Avula. Severe late‐onset neutropenia induced by ocrelizumab in a multiple
sclerosis patient: A case report. Clin Case Rep. 2022;10(1):e05299.
71. Trial, The Diabetes Control and Complications. Epidemiology of Diabetes
Interventions and Complications (DCCT/EDIC) Study Research Group
Intensive Diabetes Treatment and Cardiovascular Disease in Patients with
Type 1 Diabetes. N Engl J Med. 2005;353(25):2643-2653.
72. Flory James, Kasia Lipska. Metformin in 2019. JAMA. 2019;321(19):1926-
1927.
73. Brendon L Neuen, Tamara Young, Hiddo J L Heerspink, Bruce Neal,
Vlado Perkovic, Laurent Billot, et al. SGLT2 Inhibitors for the Prevention of
Kidney Failure in Patients with Type 2 Diabetes: A Systematic Review and
Meta-Analysis. Lancet Diabetes Endocrinol . 2019;7(11):845-854.
74. Marcel H A Muskiet, Daniël H van Raalte, Erik J M van Bommel, Mark M
Smits, Lennart Tonneijck. Understanding EMPA-REG OUTCOME Lancet
Diabetes Endocrinol. 2015;3(12):928-9.
75. Ashu Rastogi, Anil Bhansali. SGLT2 Inhibitors through the Windows of
EMPA-REG and CANVAS Trials: A Review. Diabetes Ther.
2017;8(6):1245-1251.
76. Wajeeha Aiman, Ayesha Anwar, Danish Iltaf Satti, Mohammad Hamza,
Alynna Mariz Perez, Arslan Ahmad Khan, et al. MM-082 Addition of
Daratumumab to Drug Regimens in the Treatment of Multiple Myeloma: A
Systematic Review and Meta-Analysis of Randomized Clinical Trials.
Clinical Lymphoma Myeloma and Leukemia. 2022;22:S405.
77. Dilip Sharma, Suril Verma, Shivani Vaidya, Kiran Kalia, Vinod Tiwari.
Recent Updates on GLP-1 Agonists: Current Advancements &
Challenges. Biomed Pharmacother. 2018;108:952-962.
Advances in Renal Diseases and Dialysis
Diabetic Kidney Disease
53
78. Arne Astrup, Stephan Rössner, Luc Van Gaal, Aila Rissanen, Leo
Niskanen, Mazin Al Hakim, et al. Effects of Liraglutide in the Treatment of
Obesity: A Randomised, Double-Blind, Placebo-Controlled Study. Lancet.
2009;374(9701):1606-16.
79. Chen, Szu-chi, and Chin-Hsiao Tseng. Dyslipidemia, Kidney Disease,
and Cardiovascular Disease in Diabetic Patients. Rev Diabet Stud. 2013
Summer-Fall;10(2-3):88-100.
80. Rosario Rey F, Sharma Prabhakar. Lipids and Diabetic Nephropathy. Curr
Diab Rep. 2006;6(6):455- 62.
81. Tziomalos, Konstantinos, and Vasilios G. Athyros. Diabetic Nephropathy:
New Risk Factors and Improvements in Diagnosis. Rev Diabet Stud.
2015;12(1-2):110-8.
82. Locatelli, F. The Importance of Diabetic Nephropathy in Current
Nephrological Practice. Nephrol Dial Transplant. 2003;18(9):1716-25.
83. Aso Yoshimasa. Cardiovascular Disease in Patients with Diabetic
Nephropathy. Current Molecular Medicine. 2008;8(6):533-543.
___________________________________________________________________________________
© Copyright (2023): Author(s). The licensee is the publisher (B P International).
DISCLAIMER
This chapter is an extended version of the article published by the same author(s) in the following journal.
Int Clinc Med Case Rep Jour., 2(11): 1-4, 2023. DOI: https://doi.org/10.5281/zenodo.7974808
________________________________________________________________________
a Department of Internal Medicine/Nephrology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre,
Pennsylvania, USA.
b Department of Internal Medicine, Lovelace Medical Center, Albuquerque, New Mexico, USA.
c Department of Internal Medicine, Geisinger Commonwealth School of Medicine, Scranton, PA,
Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA, USA.
d Department of Internal Medicine, MNR Medical College and Hospital, Telangana, India.
e Department of Internal Medicine/Gastroenterology, Brooklyn Hospital Center, Brooklyn, New York,
USA.
*Corresponding author: E-mail: drmaheshwarikata@gmail.com;
Chapter 4
Print ISBN: 978-81-19491-84-1, eBook ISBN: 978-81-19491-85-8
Continuous Renal Replacement Therapy
Atul Bali a, Harkesh Arora b, Roopa Naik c,
Maheshwari Kata d* and Rajeshwari Ramachandran e
DOI: 10.9734/bpi/mono/978-81-19491-84-1/CH4
ABSTRACT
Continuous renal replacement therapy (CRRT) is commonly used for acute
kidney injury (AKI) in intensive care units (ICU) for renal support, especially in
hemodynamically unstable patients in much of the developed world. However,
despite its widespread use, there is no formal evidence regarding the
improvement in patient related outcomes when CRRT is used instead of
intermittent hemodialysis (IHD). Various techniques can be used that differ in the
mode of solute removal, including continuous venous hemofiltration with primarily
convective solute removal, continuous venous hemodialysis with primarily
diffusional solute removal, and continuous venous hemodiafiltration, which is a
combination of both dialysis and hemofiltration methods. In this review, we
compare CRRT with other renal support modalities and examine the indications
for initiation of renal replacement therapy, as well as dosage and technical
considerations for administering CRRT. We also describe some of the
controversies and questions that remain to be answered regarding the use of
CRRT.
Keywords: Renal failure; hemodialysis; renal replacement therapy; acute kidney
injury.
1. INTRODUCTION
Acute kidney injury (AKI) is a common complication of critically ill patients and is
associated with significant morbidity and mortality [1,2]. About 5-10% of patients
with AKI require renal replacement therapy during their ICU stay, with a mortality
rate of 30-70%. Over the past two decades, the incidence of AKI requiring renal
replacement therapy has increased by approximately 10% per year [3]. Risk
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
55
factors for AKI requiring active treatment include older age, male sex, African-
American race, disease severity, sepsis, decompensated heart failure, cardiac
surgery, liver failure, and use of mechanical ventilation.
Continuous renal replacement therapy includes spectrum of dialysis techniques
developed in the 1980s that were developed specifically for the treatment of
critically ill patients with acute kidney injury who could not undergo traditional
intermittent hemodialysis due to hemodynamic instability or whose volume or
metabolic abnormalities could not be controlled with intermittent hemodialysis [4].
Slower solute clearance and fluid removal per unit time with continuous renal
replacement therapy compared with intermittent hemodialysis is thought to allow
for better hemodynamic tolerance. Although once considered an extraordinary
procedure, the provision of RRT care has become routine, even in the setting of
significant hemodynamic instability [5,6]. However, considerable uncertainty
remains regarding many key aspects of active care management, including the
optimal timing of initiation and termination and the choice of treatment modalities.
This article provides an overview of key issues in critical care management of
critically ill patients, with a primary focus on the use of continuous renal
replacement therapy (CRRT).
2. RRT MODALITIES
Several modalities of renal replacement therapy can be used in the treatment of
critically ill patients with renal failure [7]. These include CRRT, conventional
intermittent hemodialysis (IHD), and prolonged intermittent renal replacement
therapy (PIRRT), which is a combination of CRRT and IHD. All of them use
relatively similar extracorporeal circulation and differ mainly in the duration of
treatment and thus in net ultrafiltration rate and solute removal. IHD ensures
rapid removal and ultrafiltration of the solute during a relatively short (4 hour)
treatment; continuous therapy provides more gradual fluid removal and solute
clearance over a longer treatment period (optimally 24 hours per day, but often
interrupted by systemic coagulation or diagnostic or therapeutic procedures).
Different forms of PIRRT are characterized by treatments that typically last 8-16
hours, solute absorption and ultrafiltration are slower than IHD but faster than
CRRT. PIRRT is most often delivered by IHD-like devices, but with lower blood
and dialysate flow rates [8]. PIRRT can also be performed with equipment
designed for CRRT, but with higher rates of dialysate and/or ultrafiltration to
achieve a similar treatment response in shorted period of time. Peritoneal dialysis
offers an effective alternative to extracorporeal renal replacement therapies
[9,10].
3. CHOICE OF RRT MODALITY
Although CRRT and PIRRT are most commonly used in hemodynamically
unstable patients, there is considerable variation in practice. Some centers use
CRRT (or PIRRT) in most patients with renal failure, regardless of hemodynamic
status, while others may use intermittent hemodialysis even in some patients
who are vasopressor- dependent. Although the benefit of slow, continuous renal
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
56
support in hemodynamically unstable patients may seem self-evident,
randomized trials have failed to show any significant differences in terms of
important outcomes such as mortality or recovery of renal function comparing
CRRT with IHD or PIRRT [11,12]. However, it should be appreciated that to
provide IHD in hemodynamically unstable patients, the dialysis prescription may
require modifications, such as increasing the treatment time to allow more
gradual ultrafiltration, using higher sodium dialysate concentrations and lowering
dialysis temperatures. Although the Kidney Disease: Improving Global Outcomes
(KDIGO) Clinical Practice Guidelines for AKI recommend the use of CRRT in
hemodynamically unstable patients, the strength of this recommendation is low.
However, observational data indicate that CRRT is more effective than IHD in
achieving negative fluid balance. Furthermore, in brain-damaged patients with
acute liver failure or increased intracranial pressure, CRRT is associated with
better preservation of cerebral perfusion than IHD [13,14].
Although CRRT was originally developed as arteriovenous therapy, it is now
performed using pump-driven veno- venous circuits. Pump-driven venous
circulation provides greater and more consistent blood flow and eliminates the
hazards associated with long-term arterial cannulation with a large-bore catheter.
Several techniques have been developed to deliver CRRT. If the treatment is
used only for volume control, it is called slow continuous ultrafiltration (SCUF).
When CRRT is delivered as continuous venovenous hemofiltration (CVVH),
continuous venovenous hemodialysis (CVVHD), or continuous venovenous
hemodiafiltration (CVVHDF), it provides both solute clearance and volume
removal, and the differences between these methods relate to solute clearance.
In CVVH, the hydrostatic gradient across the semipermeable hemofilter
membrane creates a high ultrafiltration rate, and solute transport occurs by
convection. Dissolved substances are carried by the water flow through the
membrane, often referred to as "solvent drag" [15]. High ultrafiltration rates are
required to achieve sufficient solute uptake, and a volume of ultrafiltrate greater
than that required to remove the desired fluid is replaced with balanced IV
crystalloid solutions. These replacement solutions can be infused into the
extracorporeal circulation either before or after the hemofilter. Because the high
ultrafiltration rate hemoconcentrates the blood as it passes through the hemofilter
fibers, the risk of contamination and clogging of the fibers increases. An infusion
of prefilter replacement fluid dilutes the blood entering the hemofilter, thereby
reducing the hemoconcentration [16,17]. However, administration of a
replacement fluid prefilter dilutes the blood solute, reducing the effective solute
clearance at a fixed ultrafiltration rate. A post-filter infusion has no such effects.
In CVVHD, the dialysate is perfused through the outer surface of the dialysis
membrane and solutes are removed from the blood by diffusion of the dialysate
along its concentration gradient. Ultrafiltration rates are relatively low compared
to CVVH levels, allowing negative fluid balance without the need for IV
replacement fluids [18]. CVVHDF is a hybrid that combines CVVHD dialysis flow
with high ultrafiltration rates and the use of CVVH replacement fluids. The
different solute removal mechanisms provided by CVVH and CVVHD result in
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
57
different solute removal profiles for each method. Diffusion ensures effective
removal of low molecular weight solutes (< 500-1500 daltons); however, diffusion
clearance decreases rapidly as the molecular weight of the solute increases
[19,20]. In contrast, the solute movement in convection is largely limited by the
pore size of the hemofilter membrane. Removal of lower and higher molecular
weight solutes is similar until the molecular radius of the solute approaches the
membrane pore size. Thus, at equivalent outlet flow rates, CVVH provides
greater clearance than CVVHD for solutes ranging from 1000 to 20,000 daltons,
or even greater when larger pore membranes are used. Independent of
diffusion and convection, solute adsorption in the CRRT chain can also
contribute to complete solute uptake, depending on the saturation of membrane
binding sites. Thus, the choice of CRRT modality (CVVH, CVVHD, or CVVHDF)
depends primarily on provider preference rather than patient characteristics or
objective outcome data.
4. INITIATION OF RENAL REPLACEMENT THERAPY
Indications for starting renal replacement therapy Indications for initiation of
CRRT generally correspond to the general indications for the dialysis, including
volume overload, severe metabolic acidosis and electrolyte imbalance, and overt
uremic symptoms [21-24].
Volume overload: Volume overload in AKI occurs when the kidney's ability to
maintain fluid balance is compromised when IV fluids, blood products, and/or
other medications are administered to resuscitate and support a critically ill
patient. Renal replacement therapy is usually indicated when volume overload
impairs organ function and is resistant to diuretics.
Nawal et al. [25] Although observational data from both pediatric and adult
populations show a strong association between the severity of volume overload
at the start of intensive care and the risk of mortality, causality has not been
established.
Acid-base abnormalities: Progressive metabolic acidosis is an inevitable
consequence of renal failure that develops due to impaired renal acid secretion
[26-28]. Either intermittent or continuous RRT is effective in patients with severe
acidosis unresponsive to therapy, such as a fluid overloaded patient who cannot
tolerate alkali therapy. The generally recommended threshold values for starting
RRT are pH < 7.1 to 7.2 or serum bicarbonate < 12 to 15 mmol/L. Earlier
initiation of RRT may be necessary in patients with acute lung injury receiving
lung protective ventilation, as the combination of metabolic and respiratory
acidosis may lead to severe acidemia. Although RRT increases lactate
clearance, there is little evidence that initiation of active therapy to increase
lactate clearance changes clinical outcomes in patients with lactic acidosis
unrelated to drug toxicity (eg, metformin) [29-31].
Severe electrolyte imbalance: Severe hyperkalemia is the most life-threatening
and requires prompt treatment to prevent cardiotoxicity and arrhythmias [32-34].
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
58
Initiation of RRT is appropriate when hyperkalemia is refractory to medical
therapy or recurs after initial therapy. Although no rigid thresholds based on
serum potassium can be given, active treatment of hyperkalemia alone is rarely
appropriate when potassium levels are < 6 mmol/l. Conversely, RRT is generally
indicated if potassium level is > 6.5mmol/L despite treatment [35,36].
Other electrolyte disturbances, such as severe hyponatremia or hypernatremia
and severe hyperphosphatemia, may be associated with AKI and can lead to the
initiation of RRT [37,38]. In patients with severe hyponatremia associated with
AKI, CRRT may allow a slower and more controlled correction of sodium
concentration, which is necessary to prevent the neurological consequences of
osmotic demyelination, compared to IHD.
Uremia and progressive azotemia: The use of RRT in overt uremic symptoms
such as encephalopathy and pericarditis is well established. Although these are
relatively late complications of AKI, other manifestations of uremia such as
platelet dysfunction, nutritional disorders, increased susceptibility to infection
and sepsis, heart failure, and pulmonary edema may be difficult to distinguish in
the critically ill patients with multiple organ dysfunction [39-41]. In the absence of
specific indications for active therapy, it is much more common to initiate it
prophylactically in response to persistent or progressive azotemia before the
onset of uremic manifestations. The appropriate timing of such an initiation is still
a matter of debate.
Elimination of drugs and toxins: Many toxins and drugs, such as toxic
alcohols, lithium, salicylate, valproic acid and metformin, are dialyzable, and the
timely use of RRT for poisoning and drug poisoning can prevent serious
complications [42,43]. The ability of an RRT to remove a particular drug or toxin
from the bloodstream depends on its size, volume of distribution, and protein
binding [44,45]. Thus, RRT is effective in removing smaller non-protein bound
molecules with a volume of distribution < 1 L/kg body weight. The role of RRT in
the treatment of hyperammonemia is uncertain. As in the treatment of drug
intoxication, IHD also reduces the concentration of ammonia in the blood more
quickly. However, in small case series, high-dose CRRT has been shown to be
effective in the acute treatment of severe hyperammonemia (> 400 μmol/L) in
infants with congenital metabolic disorders [46,47]. The role of CRRT in adults
with hyperammonemia as a consequence of liver failure is still less certain.
Timing of RRT initiation: In the absence of specific indications, the optimal time
of initiation of RRT for AKI is uncertain [48,49]. Early initiation of RRT allows
optimization of volume status, early correction of acid-base and electrolyte
disturbances, and control of azotemia before the development of serious
metabolic disturbances, which are objective indications. However, these potential
benefits of early initiation must be balanced against the risks and burdens
associated with active treatment, including vascular access (eg: hemorrhage,
thrombosis, vascular injury, infection), intra-dialysis hypotension, and resource
use, as well as potential worries that active treatment may prevent the
subsequent recovery of renal function [50,51].
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
59
Several observational studies have shown improved survival associated with
earlier initiation of RRT. However, these studies only included patients who
ultimately received RRT and did not consider patients with AKI who did not
receive early RRT and who either recovered renal function or died without RRT.
Excluding these patients from the analysis introduces a potential bias, as the real
clinical question is not early versus late initiation of RRT, but rather early versus
non-early RRT in patients without an urgent indication.
5. CRRT DOSE
Solute Control: The CRRT dose is calculated from the sum of the total flow
rates of effluent, dialysate and ultrafiltrate. Although several studies published 15
to 20 years ago suggested that higher flow rates of effluent were associated with
improved survival, the results were inconsistent and this association was not
confirmed in two large multicenter randomized controlled trials [52]. Based on the
available data, the KDIGO clinical practice guidelines recommend a CRRT target
dose of 20 to 25 mL/kg per hour, noting that a higher prescribed dose may be
necessary to achieve this target dose.
Volume management: Another important aspect of RRT prescription is volume
control. Net ultrafiltration can be regulated independently of solute clearance
[53]. As previously noted, the severity of volume overload is strongly associated
with mortality risk in both children and adults with AKI who require RRT.
However, optimal volume management strategies are uncertain. Treatment must
be individualized for each patient, and ultrafiltration goals must be frequently
reevaluated. It should be noted that short-term blood pressure variability is not
usually related to volume status, and that transient hypotension during CRRT
should be carefully evaluated for non-volume-mediated factors and often requires
therapy regardless of changes in ultrafiltration goals.
The role of CRRT in sepsis: Although cytokine modulation with CVVH has been
proposed as an adjunctive therapy for sepsis, clinical trials have not shown any
significant benefit [54,55]. In a study of 80 patients randomized to isovolumic
hemofiltration or standard therapy, hemofiltration did not improve clinical
parameters or mortality. Current data do not support the use of CRRT as
adjunctive therapy for sepsis in addition to renal support.
Drug dosing during CRRT: Drug dosing during CRRT can be challenging
because drug dosing must consider many factors in addition to extracorporeal
drug elimination, including non-renal elimination, residual renal function, and
changes in volume of distribution and protein binding [56,57]. Medication dosing
errors can lead to both toxicity due to insufficient dose reduction and treatment
failure due to underdosing. The latter is particularly important for antibiotic dosing
in patients with sepsis associated with AKI.
Medications with a noticeable clinical effect, such as analgesics, sedatives and
vasopressors, should be titrated according to the desired clinical response
[58,59]. Drugs with high molecular weight, strong protein binding, or very large
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
60
volume of distribution are poorly eliminated by CRRT, and no dose adjustment is
necessary for active therapy. Extracorporeal elimination of nonprotein-bound
small molecule drugs approximates effluent flow. Estimated clearance of protein-
bound drugs must be adjusted for the percentage of unbound fraction. For all
drugs whose blood levels are easily measured, dosage adjustments should be
made based on pharmacokinetic monitoring. Finally, it should be recognized that
although published guidelines provide dosing estimates for many agents, they
only provide general parameters that may not correspond to the specific form and
dose of CRRT used.
Nutritional management: AKI patients undergoing CRRT usually have a
markedly negative nitrogen balance due to high protein catabolism [60,61]. In
addition, CRRT causes a loss of amino acids and water-soluble vitamins and
other micronutrients. Caloric intake should be around 35 kcal/kg per day, aim for
a protein intake of 1.5 g/kg and supplement with water soluble vitamins. Although
enteral feeding is preferred, parenteral support may be necessary.
Complications of CRRT: As with all medical procedures, CRRT is not without
risks. Initiation of CRRT requires placement of a large-bore central venous
catheter, which may require long-term maintenance. Known complications of
catheter placement include vascular or visceral injury causing hemorrhage,
pneumothorax, hemothorax, and arteriovenous fistula formation [62,63].
Prolonged use of the catheter is associated with venous blockage or stenosis.
Exposure to blood in the extracorporeal circulation can induce immediate allergic
or delayed immunological responses due to cytokine activation. Bradykinin-
mediated membrane reactions have been associated with certain synthetic
membranes when angiotensin-converting enzyme inhibitors are used. Air
embolism can occur during catheter insertion or removal and at any time during
therapy if air enters the circuit outside the return line air detector.
Circuit clotting and anticoagulation: The most common complication during
CRRT is clotting of dialysis circuit, and the most common cause is inadequate
catheter function, leading to flow restrictions and pressure towers that interrupt
blood flow [64]. If the blood flow cannot be maintained at 200-300 ml/min, a quick
catheter change may be necessary. An excessive filter fraction can cause
hemoconcentration in the hemofilter, which also contributes to filter coagulation
[65, 66]. If there is no catheter failure, blood flow is maximized and the filtration
fraction is < 20%, initiation or intensification of anticoagulant therapy should be
considered. Complications of heparin anticoagulant therapy may include bleeding
and heparin-induced thrombocytopenia. Citrate anticoagulation can cause citrate
toxicity due to citrate accumulation, overt hypocalcemia due to insufficient
calcium replacement, and both metabolic acidosis and metabolic alkalosis as a
result.
Electrolyte abnormalities: One of the most common electrolyte imbalances
observed during CRRT is hypophosphatemia [67]. Hypophosphatemia may
result from continued removal in the circulation and may potentially lead to delay
in weaning from mechanical ventilation. Hypophosphatemia can be prevented by
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
61
prophylactic enteral or parenteral phosphate supplementation or the use of
phosphate-containing dialysate or replacement fluids.
Hemodynamic instability: Dialysate and replacement fluids are not often
warmed up, in contrast to traditional hemodialysis. Thermal losses during CRRT
can mask the onset of fever, but they may trigger vasoconstriction and
supposedly increase hemodynamic stability. Significant hypothermia may
develop from more severe thermal loss, necessitating extensive external
rewarming. In certain studies, more than one third of patients have hypotension
during CRRT; nevertheless, this condition is typically unrelated to the actual
CRRT process [68,69]. Ultrafiltration, which exacerbates hemodynamic instability,
is the most common cause of hypotension [70]. Hypotension can also be
observed at the beginning of treatment, especially if the circulation is not
reinfused; this result was of particular concern in children and can be alleviated
by the use of albumin in priming the circuit [71,72]. If hypotension is associated
with fluid depletion, treat with fluid re-infusion and adjust ultrafiltration targets; in
other cases, alternative causes should be considered and hypotension treated
with vasopressors.
6. TERMINATION OF CRRT
There are no specific criteria for discontinuation of CRRT due to recovery of renal
function or switching to other forms of RRT [73]. The first manifestation of
recovery of renal function is usually increased urine output, although specific
criteria are rare. In a study titled, observational Beginning and Ending Supportive
Therapy for the Kidney (BEST Kidney), urine output > 400 mL/d without
concomitant diuretic therapy predicted successful discontinuation of CRRT [74].
In this observational cohort, patients who successfully discontinued CRRT
without restarting it were more likely to survive to hospital discharge compared
with patients who required restarting CRRT. Another study proposed urine output
> 500 mL/d as a criterion for discontinuation of renal replacement therapy in
patients with AKI. Although these strategies may aid in clinical decision-making,
there are no precise criteria for stopping RRT. Patients with improved
hemodynamic status but persistent AKI also have a very different transition to
other forms of RRT [75]. Based on the clinical context, PIRRT can be utilized as a
transitional therapy, or patients can be referred directly to IHD. Converting from
CRRT to PIRRT or IHD may make it simpler to begin physical therapy and bed
mobilization. In general, patients with persistent ICU-dependent AKI should be
transferred to IHD before ICU discharge.
7. CONCLUSIONS
CRRT remains the mainstay of therapy for AKI in critically ill patients. In practice,
the provision of CRRT in the ICU remains highly variable. In patients without
objective indications for initiation of renal therapy, the optimal time of RRT
remains controversial. Although the use of continuous regimens may facilitate the
management of hemodynamically unstable patients, the available data do not
show that the use of CRRT improves survival or recovery of renal function
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
62
compared with alternatives such as conventional IHD and PIRRT. Large, well-
designed clinical trials have shown that in most patients, increasing solute
clearance at effluent flow rates >20–25 mL/kg/h is not associated with improved
outcomes. however, optimal volume management strategies remain to be
defined. Other aspects of CRRT management also vary considerably in
practice, including anticoagulant strategies. Finally, the role of CRRT in setting
overall treatment goals and the use of other life-sustaining treatment strategies
must be considered.
COMPETING INTERESTS
Authors have declared that no competing interests exist.
REFERENCES
1. Eric A J Hoste, Sean M Bagshaw, Rinaldo Bellomo, Cynthia M Cely, Roos
Colman, Dinna N Cruz, et al. Epidemiology of Acute Kidney Injury in
Critically Ill Patients: The Multinational AKI-EPI Study. Intensive Care
Med. 2015;41(8):1411-23.
2. Palleti, Sujith Kumar, Sreekanth Avula, Sandesh Dewan. Kidney: A
Review on End Stage Renal Disease, Dialysis and Transplant. Int Clinc
Med Case Rep Jour. 2023;2(10):1-8.
3. Muhammad O Hanif, Atul Bali, Kamleshun Ramphul. Acute Renal Tubular
Necrosis. StatPearls. 2021.
4. Anne-Sophie Truche, Michael Darmon, Sébastien Bailly, Christophe
Clec'h, Claire Dupuis, Benoit Misset, et al. Continuous Renal Replacement
Therapy versus Intermittent Hemodialysis in Intensive Care Patients:
Impact on Mortality and Renal Recovery. Intensive Care Med.
2016;42(9):1408-17.
5. Max Bell, SWING, Fredrik Granath, Staffan Schön, Anders Ekbom, Claes-
Roland Martling. Continuous Renal Replacement Therapy Is Associated
with Less Chronic Renal Failure than Intermittent Haemodialysis after
Acute Renal Failure. Intensive Care Med . 2007;33(5):773-780.
6. Palleti, Sujith Kumar, Sreekant Avula, Roopa Naik. Chronic Kidney
Disease. Int Clinc Med Case Rep J. 2023;2(11):1-15.
7. Gianluca Villa, Zaccaria Ricci, Claudio Ronco. Renal Replacement
Therapy. Critical Care Clinics. 2015;31(4):839-848.
8. Fahad Edrees, Tingting Li, Anitha Vijayan. Prolonged Intermittent Renal
Replacement Therapy. Adv Chronic Kidney Dis. 2016;23(3):195-202.
9. Rajnish Mehrotra, Olivier Devuyst, Simon J Davies, David W Johnson.
The Current State of Peritoneal Dialysis. J Am Soc Nephrol.
2016;27(11):3238-3252.
10. Sujith K Palleti, Santhoshi R Bavi, Margaret Fitzpatrick, Anuradha
Wadhwa. First case report of Sphingobium lactosutens as a human
pathogen causing peritoneal dialysis-related peritonitis. Cureus.
2022;14(7):e27293.
11. Vladimir Gasparović, Ina Filipović-Grcić, Marijan Merkler, Zoran Pisl.
Continuous Renal Replacement Therapy (CRRT) or Intermittent
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
63
Hemodialysis (IHD)—What Is the Procedure of Choice in Critically Ill
Patients? Ren Fail. 2003;25(5):855-62.
12. Palleti Sujith K, et al. Study about Sphingobium Lactosutens: A Human
Pathogen Causing Peritoneal Dialysis Related Peritonitis. Perspective of
Recent Advances in Medical Research. BP International 9 (2023):31-35.
13. Davenport Andrew. Continuous Renal Replacement Therapies in Patients
with Liver Disease. Semin Dial . 2009;22(2):169-72.
14. Jithin Karedath, FNU Avanteeka, Muhammad Nouman Aslam, Ahmad
Nadeem, Rao Ahmed Yousaf, Sandesh Shah, et al. Comparison of
Effectiveness and Safety of Low-Dose Versus Standard-Dose
Intravenous Recombinant Tissue Plasminogen Activator in Patients With
Acute Ischemic Stroke: A Meta- Analysis. Cureus. 2023;15(2).
15. William R Clark, Joseph E Turk, Michael A Kraus, Dayong Gao. Dose
Determinants in Continuous Renal Replacement Therapy. Artif Organs.
2003;27(9):815-20.
16. Raghavan Murugan , Eric Hoste, Ravindra L Mehta, Sara Samoni,
Xiaoqiang Ding, Mitchell H Rosner, et al. Precision Fluid Management in
Continuous Renal Replacement Therapy. Blood Purif. 2016;42(3):266-78.
17. Zaid Munir, Muhammad Akash, Fnu Jaiprada, Bilal Abu Tarboush,
Osama Ijaz, Anan Bseiso, et al. Evaluation of the Effects of
Extracorporeal Shockwave Therapy in Patients with Peripheral Arterial
Disease: A Meta-Analysis of Randomized Control Trials. Cureus.
2023;15(2):e34729.
18. P Saudan, M Niederberger, S De Seigneux, J Romand, J Pugin, T
Perneger, et al. Adding a Dialysis Dose to Continuous Hemofiltration
Increases Survival in Patients with Acute Renal Failure. Kidney Int.
2006;70(7):1312-7.
19. Winchester James F, Patrick F Audia. UNRESOLVED ISSUES in
DIALYSIS: Extracorporeal Strategies for the Removal of Middle
Molecules. Seminars in Dialysis. 2006;19(2,3):110-114.
20. Atul Bali, John Kevin Hix, Peter Kouides. Safe and effective use of chronic
transdermal estradiol for life- threatening uremic bleeding in a patient with
coronary artery disease. Nephron Extra. 2014;4(2):134-7.
21. Noel Gibney, Eric Hoste, Emmanuel A Burdmann, Timothy Bunchman,
Vijay Kher, Ravindran Viswanathan, et al. Timing of Initiation and
Discontinuation of Renal Replacement Therapy in AKI: Unanswered Key
Questions. Clin J Am Soc Nephrol. 2008;3(3):876-80.
22. Sujith K Palleti, Anuradha Wadhwa, Abdul H Rasheed. The First Case of
Ceftazidime-Induced Antibiomania: A Case Report and Literature Review.
Cureus. 2022;14(12):e32904.
23. Bali Atul. Pseudo Metabolic Acidosis in the Setting of Hypertriglyceridemia:
A Case Report. 2020.
24. Palevsky Paul M. Renal Replacement Therapy I: Indications and Timing.
Crit Care Clin. 2005;21(2):347- 56.
25. Nawal Salahuddin, Mustafa Sammani, Ammar Hamdan, Mini Joseph,
Yasir Al-Nemary, Rawan Alquaiz, et al. Fluid Overload Is an Independent
Risk Factor for Acute Kidney Injury in Critically Ill Patients: Results of a
Cohort Study. BMC Nephrol. 2017;18(1):45.
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
64
26. S Lim. Metabolic Acidosis. Acta Medica Indonesiana. 2007;39(3):145-
50.
27. Sai Kiran Kondabolu, Yasitha Kakarlapudi, Haider Malik, Hamza Malik,
Saima Khan, Praveen Kumar Komminni, et al. Beneficial Impacts of
Fenoldopam on Patients with or at Risk for Acute Renal Failure and
Undergoing Surgery: A Meta-Analysis of Randomized Clinical Trials.
Cureus. 2023;15(2):e34584.
28. Jiachang Hu, Yimei Wang, Xuemei Geng, Rongyi Chen, Xialian Xu,
Xiaoyan Zhang, et al. Metabolic Acidosis as a Risk Factor for the
Development of Acute Kidney Injury and Hospital Mortality. Exp Ther Med.
2017;13(5):2362-2374.
29. Lalau Jean-Daniel. Lactic Acidosis Induced by Metformin. Drug Saf.
2010;33(9):727-40.
30. Raza A Khan, Nidhi Patel, Atunde Folajimi, Bansari Raveena Bai,
Vrushak Patel, Praveen Kumar Komminni, et al. Comparison of the
Efficacy and Safety of Metformin-Based Combination Therapy Versus
Metformin Alone in Children and Adolescents with Type 2 Diabetes
Mellitus: A Meta-Analysis. Cureus. 2023;15(2):e35014.
31. Rakesh Pilla, Sujith Kumar Palleti, Renuka Rayana, Satish Reddy Skss,
Aminah Abdul Razzack, Sruti Kalla. Glycated haemoglobin (HbA1c)
variations in nondiabetics with nutritional anemia. Cureus.
2020;12(11):e11479.
32. J Ahmed, L S Weisberg. Weisberg. Hyperkalemia in Dialysis Patients.
Semin Dial. 2001;14(5):348-56.
33. Gowrishankar K, Anurag Rawat, Pratibha Singh, Sujith Palleti. Study on
Recognition of Heart Arrhythmias by Deep Learning Approach of the ECG
Signals using Edge Devices. 2023.
34. Muhammad Ali Sameer, Bilal Aziz Malik, Muhammad Obaid Ullah
Choudry, Muhammad Shoaib Anwar, Muhammad A Nadeem, Fizza
Mahmood, et al. Comparison of Coronary Artery Bypass Grafting
Combined with Mitral Valve Repair Versus Coronary Artery Bypass
Grafting Alone in Patients With Moderate Ischemic Mitral Regurgitation: A
Meta-Analysis. Cureus. 2023;15(4):e37238.
35. Ho-Jung Kim, Sang-Woong Han. Therapeutic Approach to Hyperkalemia.
Nephron. 2002;92 Suppl 1:33- 40.
36. Karthik Gonuguntla, Komal Ejaz, Chaitanya Rojulpote, Ayesha Shaik,
Abhijit Bhattaru, Tapan Buch, et al. A nationwide cohort analysis to
determine the prevalence of sinus node dysfunction and rates of
pacemaker implantation in systemic lupus erythematosus. Am J
Cardiovasc Dis. 2021;11(4):478-483.
37. Lenar Yessayan, Jerry Yee, Stan Frinak, Balazs Szamosfalvi. Continuous
Renal Replacement Therapy for the Management of Acid-Base and
Electrolyte Imbalances in Acute Kidney Injury. Adv Chronic Kidney Dis.
2016;23(3):203-10.
38. Abdulmaleek Idanesimhe Sado, Muhammad S Afzal, Lavanya
Kannekanti, Hrushikesh Reddy Pamreddy, Jorge Pimentel Campillo,
Vaishnavi Kandukuri, et al. A Meta-Analysis on Predictors of Mortality
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
65
Among Patients Hospitalized for Acute Exacerbation of Asthma. Cureus.
2023;15(2):e35225.
39. Timothy W Meyer, Thomas H Hostetter. Uremia. New England Journal of
Medicine. 2007;357(13): 1316- 1325.
40. Abshiro H Mayow, Fatima Ahmad, Muhammad Sohaib Afzal, Muhammad
Usama Khokhar, Daneyal Rafique, Sai Krishna Vallamchetla, et al. A
Systematic Review and Meta-Analysis of Independent Predictors for
Acute Respiratory Distress Syndrome in Patients Presenting with Sepsis.
Cureus. 2023;15(4):e37055.
41. Palleti Sujith K, Anuradha Wadhwa, Abdul H. Rasheed. Ceftazidime-
induced Antibiomania: A Rare Case Study and Review of
Literature. Research Highlights in Disease and Health Research.
2023;6:56-61.
42. Winchester James F. Dialysis and Hemoperfusion in Poisoning. Adv Ren
Replace Ther. 2002;9(1):26-30.
43. Karima Benaicha, Raja Ram Khenhrani, Maha Veer, Sapna Devi, Usman
Shahbaz, Qais M. Salah, et al. Efficacy of Molnupiravir for the Treatment
of Mild or Moderate COVID-19 in Adults: A Meta-Analysis. Cureus.
2023;15(5).
44. Bert Bammens, Pieter Evenepoel, Kristin Verbeke, Yves Vanrenterghem.
Removal of Middle Molecules and Protein-Bound Solutes by Peritoneal
Dialysis and Relation with Uremic Symptoms. Kidney Int.
2003;64(6):2238-43.
45. Wajeeha Aiman, Ayesha Anwar, Danish Iltaf Satti, Mohammad Hamza,
Alynna Mariz Perez, Arslan Ahmad Khan, et al. MM-082 Addition of
Daratumumab to Drug Regimens in the Treatment of Multiple Myeloma: A
Systematic Review and Meta-Analysis of Randomized Clinical Trials.
Clinical Lymphoma Myeloma and Leukemia. 2022;22:S405.
46. Ari Auron, Patrick D Brophy. Brophy. Hyperammonemia in Review:
Pathophysiology, Diagnosis, and Treatment. Pediatr Nephrol.
2012;27(2):207-22.
47. Atul D Bali, Akiva Rosenzveig, William H Frishman, Wilbert S Aronow.
Nonalcoholic Fatty Liver Disease and Cardiovascular Disease: Causation
or Association. Cardiol Rev. 2023;e000537.
48. Susan P Y Wong, Elizabeth K Vig, Janelle S Taylor, Nilka R Burrows,
Chuan-Fen Liu, Desmond E Williams, et al. Timing of Initiation of
Maintenance Dialysis. JAMA Intern Med. 2016;176(2):228-35.
49. Kar Saikat, et al. A two-center, retrospective cohort trial of a tumor
necrosis factor inhibitor coupled with intravenous immunoglobulin and
heparin for the treatment of recurrent spontaneous abortion. 2023.
50. Maher JF, GE Schreiner. Hazards and Complications of Dialysis. N Engl J
Med. 1965;273(7):370-7.
51. Vaillant Angel A Justiz, Roopa Naik. HIV-1 associated opportunistic
infections. StatPearls [Internet]. 2021.
52. Robin Rauniyar, Rahul Rauniyar, Ankita Agrawal, Shikha Yadav, Sreekant
Avula. Dosing Patterns for Continuous Renal Replacement Therapy at a
Large Academic Medical Center in the United States. J Crit Care.
2002;17(4):246-50.
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
66
53. Daugirdas John T. Pathophysiology of Dialysis Hypotension: An Update.
American Journal of Kidney Diseases. 2001;38(4):S11-S17.
54. C Ronco, Z Ricci, R Bellomo. Importance of Increased Ultrafiltration
Volume and Impact on Mortality: Sepsis and Cytokine Story and the Role
for CVVH. EDTNA ERCA J. 2002;Suppl 2:13-8.
55. Syed, Fareeha Naseer, et al. Hepatitis C Virus Infection in Patients with
Beta Thalassemia after Multiple Transfusions at a Tertiary Care
Hospital. Pakistan Journal of Medical & Health Sciences. 2023;17(1):363-
363.
56. Gordon Choi, Charles D Gomersall, Qi Tian, Gavin M Joynt, Ross
Freebairn, Jeffrey Lipman, et al. Principles of Antibacterial Dosing in
Continuous Renal Replacement Therapy. Crit Care Med.
2009;37(7):2268-82.
57. Palleti Sujith K, Maria M. Picken, Anuradha Wadhwa. Sjögren’s Syndrome
Overlap with Lupus Nephritis: A Case Report and Literature Review of a
Rare Entity. Cureus. 2023;15(5).
58. Scoville Bridget A, Bruce A Mueller. Medication Dosing in Critically Ill
Patients with Acute Kidney Injury Treated with Renal Replacement
Therapy. Am J Kidney Dis. 2013;61(3):490-500.
59. Robin Rauniyar, Rahul Rauniyar, Ankita Agrawal, Shikha Yadav, Sreekant
Avula. Severe late‐onset neutropenia induced by ocrelizumab in a multiple
sclerosis patient: A case report. Clin Case Rep. 2022;10(1):e05299.
60. Marlies Ostermann, Nuttha Lumlertgul, Ravindra Mehta. Nutritional
Assessment and Support during Continuous Renal Replacement Therapy.
Semin Dial. 2021;34(6):449-456.
61. Bali Atul, Roopa Naik. The Impact of a Vegan Diet on Many Aspects of
Health: The Overlooked Side of Veganism. Cureus. 2023;15(2).
62. Sigwalt Florent, et al. Clinical Complications of Continuous Renal
Replacement Therapy. Contrib Nephrol. 2018;194:99-108.
63. Palleti SK, Larson H, Avula S, et al. Subarachnoid Hemorrhage in
Autoimmune Vasculitis: A Rare Presentation of Systemic Lupus
Erythematosus-Antineutrophil Cytoplasmic Autoantibody-Associated
Vasculitis Overlap Syndrome. Cureus. 2023;15(5).
64. Davies Hugh, Gavin Leslie. Maintaining the CRRT Circuit: Non-
Anticoagulant Alternatives. Aust Crit Care . 2006;19(4):133-8.
65. Joannidis Michael, Heleen M Oudemans-van Straaten. Clinical Review:
Patency of the Circuit in Continuous Renal Replacement Therapy. Crit
Care. 2007;11(4):218.
66. Rohit Sharma, Tristan Nguyen-Luu, Pragya Dhaubhadel, Amit Sharma,
Roopa Naik. A Rare Co- occurrence of Monkeypox Encephalitis and
Neurosyphilis. Cureus. 2023;15(3):e35945.
67. Young-Hye Song, Eun-Hye Seo, Yang-Sook Yoo, Young-Il Jo. Phosphate
Supplementation for Hypophosphatemia during Continuous Renal
Replacement Therapy in Adults. Ren Fail. 2019;41(1):72-79.
68. Adrianna Douvris, Khalid Zeid, Swapnil Hiremath, Sean M Bagshaw, Ron
Wald, William Beaubien- Souligny, et al. Mechanisms for Hemodynamic
Instability Related to Renal Replacement Therapy: A Narrative Review.
Intensive Care Med. 2019;45(10):1333-1346.
Advances in Renal Diseases and Dialysis
Continuous Renal Replacement Therapy
67
69. Eyth Emily, Roopa Naik. Hemoglobin A1c. StatPearls Publishing.
2022.
70. Sreekant Avula, Sandesh Dewan, Bushra Firdous Shaik, Atul Bali.
Diabetic Kidney Disease. International Clinical and Medical Case Reports
Journal. 2023;2(11).
71. Maria J Santiago, Jesús López-Herce, Javier Urbano, María José Solana,
Jimena del Castillo, Yolanda Ballestero, et al. Complications of
Continuous Renal Replacement Therapy in Critically Ill Children: A
Prospective Observational Evaluation Study. Crit Care. 2009;13(6):R184.
72. Zineb Barkhane, FNU Nimerta, Sualeha Zulfiqar, Saleha Dar, Muhammad
Sohaib Afzal, Amna Zaree, et al. Independent Predictors of Mortality in
Systemic Sclerosis-Associated Pulmonary Arterial Hypertension: A
Systematic Review and Meta-Analysis. Cureus. 2023;15(5).
73. S Katayama, S Uchino, M Uji, T Ohnuma, Y Namba, H Kawarazaki, et al.
Factors Predicting Successful Discontinuation of Continuous Renal
Replacement Therapy. Anaesth Intensive Care. 2016;44(4):453-7.
74. Shigehiko Uchino, Rinaldo Bellomo, Hiroshi Morimatsu, Stanislao
Morgera, Miet Schetz, Ian Tan, et al. Continuous Renal Replacement
Therapy: A Worldwide Practice Survey. Intensive Care Med.
2007;33(9):1563-70.
75. S Uchino, R Bellomo, J A Kellum, H Morimatsu, S Morgera, M R Schetz,
et al. Patient and Kidney Survival by Dialysis Modality in Critically Ill
Patients with Acute Kidney Injury. Int J Artif Organs. 2007;30(4):281-92.
___________________________________________________________________________________
© Copyright (2023): Author(s). The licensee is the publisher (B P International).
DISCLAIMER
This chapter is an extended version of the article published by the same author(s) in the following journal.
Int Clinc Med Case Rep Jour. 2(12):1-13, 2023. DOI: https://doi.org/10.5281/zenodo.8006885
London Kolkata Tarakeswar
India: Guest House Road, Street no - 1/6, Hooghly, West Bengal, India (Reg. Address),
Diamond Heritage Building, 16, Strand Road, Kolkata, 700001 West Bengal, India (Corporate Address),
Tele: +91 7439016438 | +91 9748770553, Email: director@bookpi.org,
(Headquarters)
UK: 27 Old Gloucester Street London WC1N 3AX, UK,
Fax: +44 20-3031-1429, Email: director@bookpi.org,
(Branch office)