You are on page 1of 62

Section 11

Section Editor: /BSJOEFS1 Singh

Nephrology
146. Management of Hypertension in 150. Dietary Salt and Hypertension
Chronic Kidney Disease PP Varma
Mritunjay Kumar Singh
151. Newer Avenues in Management of CKD Anemia
147. Prevention of Progression of CKD—What Is New? Manish Rathi, Rudreshwar Prabakaran
Krishnaswamy Sampathkumar
152. SGLT2 Inhibitors—Mechanisms of
148. Lupus Nephritis—Current Understanding Cardiorenal Protection
and Management Ashwini Balasaheb Gadde, Vijay Kher
Krishan Lal Gupta, Joyita Bharati
153. Optimizing Renal Care in Elderly
149. Diagnosis and Management of Narinder Pal Singh, Rahul Mishra, Shaurya Kaul
Nephrotic Syndrome 154. Understanding the Role of Vitamin D
Deepti Sharma, Sandeep S, Gopikrishnan R
in Diabetic Nephropathy
Srinath KM, Spoorthy Rajj, , Manjunath S Shetty

Section-11.indb 945 28-01-2021 12:25:18


Section-11.indb 946 28-01-2021 12:25:18
CHAPTER

146 Management of Hypertension in


Chronic Kidney Disease
Mritunjay Kumar Singh

Abstract 
The relationship between hypertension and chronic kidney disease is phenomenal hypertension (HTN) which is considered
as most important modifiable risk factor for chronic kidney disease (CKD). Personalized and individualized HTN therapy
using standardized office BP and home BP monitoring hold great promise. Angiotensin converting enzyme inhibitor (ACEI)
or angiotensin receptor blocker (ARB), Calcium channel blocker (CCB), appropriate diuretic therapy, dietary salt restriction,
and ensuring proper adherence to therapy make up the foundation for the treatment of HTN in CKD.

Introduction peripheral resistance which is due to autoregulatory


peripheral vascular bed vasoconstriction in response
Studies from India and abroad have provided concordant
to increase peripheral perfusion due to extracellular
data on high prevalence of chronic kidney disease (CKD)
volume expansion. So sodium retention can induce
worldwide. Up to 1 in 10 people world wide has CKD. It is
HTN in two ways—at first by volume dependent
a silent killer like other metabolic diseases and engages
mechanism (extracellular fluid expansion) and later
major human and economical resources. Association
on mainly through volume independent mechanism
of CKD and hypertension (HTN) is bidirectional. There
like increased peripheral vascular resistance.
is a cause and effect relationship between them. HTN
„„ Increased rennin angiotensin aldosterone activity
is considered as most important modifiable risk factor
(RAAS)—Hypersecretion of rennin in a sclerosed
for CKD. Managing HTN effectively not only retards the
kidney leads to increased angiotensin II and RAAS
progression of CKD, but also reduces the cardiovascular
activity leading to increased systemic vascular
risk significantly.
resistance, sodium retention, and high BP.
„„ Increased activity of systemic nervous system (SNS)—
Pathophysiology of HTN in CKD1-3 The kidney is richly innervated sensory organ. Several
Pathophysiology of HTN in CKD is multifactorial. It studies from animal and human subjects establish the
contains components of neural and hormonal pathways. kidney-brain axis theory. Sensory inputs from kidney
The important mechanisms include: transmitted to central nervous system in turn lead to
„„ Impaired sodium excretion—Guyton and his team increased activity of SNS leading to vasoconstriction,
have shown that HTN can be generated in partially rennin secretion, sodium retention, and HTN.
nephrectomized dog (renal mass reduced to 30%) with „„ Endothelin dysfunction—CKD is a chronic low-grade
salt loading.4 The elevated blood pressure (BP) in this inflammatory state and so endothelial dysfunctions are
setting is induced by increased cardiac output (volume commonly observed in CKD. Endothelial dysfunction
expansion) initially, and later on sustained by increased leads to impaired nitrous oxide production and

Section-11.indb 947 28-01-2021 12:25:18


948 SECTION 11 Nephrology

increased endothelin level leading to increased non-diabetics CKD, thus leads to progressive renal
peripheral vasoconstriction and HTN. damage with even moderate elevation of BP.
„„ Vascular stiffness—The distensibility of resistance
arteries is affected by certain functional and Measurement of BP in CKD3,7-9
m o r p h o l o g i ca l c ha ng e s. It i n c l u d e s i nt i ma e
The proper BP measurement is the first step toward the
hyperplasia, medial calcification, smooth muscle
effective BP management in CKD. BP measurement
cell hyperplasia, and endothelial dysfunction. These
can vary depending on the setting (e.g., casual office,
changes are largely contributed by disturbed calcium-
standardized office, or home) and type of device used (e.g.,
phosphorus balance, secondary hyperparathyroidism
aneroid or Oscillometric). Standardized office BP (SOBP) is
and other neurohumoral components discussed
5–10 mm Hg lower than casual office BP. Most of the recent
above. Vascular stiffness leads to increased systolic BP
trials on optimum BP goals have used SOBP as a method
and wide pulse pressure, which is a strong predictor of
of measurement. For standardized office BP measurement
cardiovascular mortality in CKD.
certain preparations are required—(e.g., 5 minutes of
„„ The erythropoietin induced HTN and obstructive sleep
quite rest/no nicotine, caffeine, and exercise 30 minutes
apnea (OSA) are important causes of HTN in CKD,
prior to measurement/well validated and periodically
especially in dialysis population. The erythropoietin or
calibrated device/correct cuff size/middle of the cuff
erythropoietin-stimulating agents used for treatment
should be placed at the level of right atrium/average of
of anemia in CKD can induce HTN or require change
more than two readings obtained on two occasions) as laid
in antihypertensive regimen in approximately 30%
down by 2017 ACC/AHA high BP guidelines.
of the cases. Besides rapid and over correction of
The out of office BP measurement (ambulatory BP and
anemia, HTN in this subset is thought to be caused
home BP monitoring) is required to diagnose white coat
by vasoconstrictor effect of erythropoietin, which
HTN (defined as elevated office BP with controlled out
is independent of hemoglobin.5 OSA is common in
of office BP, prevalence in CKD ranges from 2% to 41%)
advanced CKD population. Chronic intermittent
and masked HTN (defined as controlled office BP and
nocturnal hypoxemia provoke sympathetic nervous
elevated out of office BP, prevalence in CKD ranges from
system activity, RAAS activity, and increases nocturnal
6% to 51%). Additionally 24 hour ambulatory BP gives clue
BP.
about the nocturnal BP. Physiological nocturnal dipping
„„ Medications like over-the-counter nonsteroidal anti-
is absent in 14–75% of the CKD population. Thus in CKD
inflammatory drugs, herbal supplements, steroids,
out of office BP accurately define the clinical problem and
and decongestants can provoke HTN in the CKD
predicts more accurately about the cardiovascular and
population.
renal outcomes than office BP.
„„ The worsening of HTN has been reported in CKD
KDOQI (kidney disease outcome quality initiative)
patients with the use of potent anti-tuberculosis drug
US commentary on the 2017 ACC/AHA HTN guideline
rifampicin. Rifampicin is a potent enzyme inducer and
commented that home BP monitoring can accurately
decreases the level of commonly used antihypertensive
predict target organ damage and better placed than
drugs—amlodipine, metoprolol, and prazosin. So it is
ambulatory BP monitoring and office BP monitoring.
advisable to monitor HTN in CKD patients, once they
HBPM can also help to overcome the therapeutic inertia.
have been put on rifampicin.6
In our limited resource setting we should prefer
„„ The HTN is the most common complication of CKD
standardized office BP, because it requires nothing but
as well as major factor responsible for progression of
our little patience (pre-measurement preparation as
CKD. Renal autoregulatory mechanism protects renal
mentioned above).
vasculatures from elevated BP. Maintenance of afferent
arteriole tone in response to elevated systemic pressure
and increased sodium chloride delivery to macula Target BP in CKD2,3,10-14
densa are part of this autoregulatory mechanism. The management of HTN in CKD is a dynamic process,
However, it is impaired in patients with diabetes and as estimated glomerular filtration rate, comorbidities

Section-11.indb 948 28-01-2021 12:25:18


Management of Hypertension in Chronic Kidney Disease CHAPTER 146 949

and risk changes with time. Due to presence of vascular SBP is a big challenge. Additionally the benefit of lower
stiffness and wide pulse pressure in CKD, achieving a BP is less certain in diabetic CKD and advanced CKD
BP target rapidly may lead to postural hypotension. So population. The 2017 American College of Cardiology
a cautious and gradual approach is required to reach a (ACC) has adopted the result of SPRINT and suggested
desired BP target. more intensive BP control in CKD population. In light of
From 1994 till 2010, there were four landmark trials SPRINT and other newer studies KDIGO controversies
compared the standard versus intensive BP control in conference 2017 has also suggested for revision in BP
patients with CKD. These were: diagnosis threshold and BP treatment target.
„„ MDRD (modification of diet in renal disease), year
In our setting, we should target lower BP:
1994 „„ If SOBP/HBPM is the method used for measurement
„„ AASK (Africans American study of kidney disease and
of BP
hypertension), year 2002 „„ If patient is tolerating the target BP well without
„„ REIN-2 (Ramipril efficacy in nephropathy-2), year 2005
postural hypotension or other adverse effect
„„ ACCORD (Action to control cardiovascular risk in
„„ If patient is having non-diabetic CKD
diabetes), year 2010
Results of all these trials favored standard BP control
rather than intensive control in most of the CKD patients,
Secondary HTN in CKD3
exceptions are patients with proteinuria more than 1 g/ Secondary causes of HTN are potentially treatable and
day, which were benefited from intensive control of BP. On should be searched in following conditions:
„„ If the onset of elevated BP occurred before puberty and
the basis of these trials, KDIGO (kidney disease initiative
global outcome) 2012 suggested a lower BP for significant preceded the development of CKD.
„„ Severe or malignant HTN that is out of proportion to
proteinuria.
„„ Albuminuria (<30 mg/24 hr): ≤140/90 (1B)
the degree of CKD is present.
„„ Acute worsening of BP control occurs in a previously
„„ Albuminuria (>30 mg/24 hr): ≤130/80 (2D)

Till year 2015, it was thought that controlling BP to hypertensive patient with good BP control, or resistant
less than 140/90 mm Hg is likely to be beneficial in CKD HTN is present.
„„ Persistent hypokalemia off diuretic treatment (primary
population. But with the advent of SPRINT (systolic BP
intervention trial) study in 2015, intensive control of aldosteronism).
„„ Development of tremor and palpitation (pheochromo­
systolic BP (<120 mm Hg) has gained momentum. SPRINT
has included data from 9,361 adults age 50 or older with cytoma).
„„ Flash pulmonary edema (renal artery stenosis).
systolic BP of 130 mm Hg or higher and at least one
additional cardiovascular disease risk factor. Around 28%
of participants had CKD without significant proteinuria. Non-pharmacological Therapy1-3
The highlights of SPRINT study are: Early institution of non-pharmacological therapy in the
„„ intensive reduction in SBP reduces cardiovascular form of dietary modification and life style modification is
events the first step to the treatment of HTN in CKD.
„„ intensive reduction in SBP has mortality benefit in „„ Dietary salt restriction (sodium intake <1.5 g/day)—

CKD population Reduce BP and proteinuria; potentiate the action of


„„ the subgroup analysis of older patients with CKD in ACEI/ARB.
SPRINT has also favorable outcome „„ DASH (Dietary approach to stop HTN) Diet—Diet

„„ with adoption of SBP <120 mm Hg in all CKD rich in fruits and vegetables, low in saturated and
population, separate BP target for proteinuria is not unsaturated fat can lead to modest reduction in BP.
required But it is not appropriate to use DASH Diet in advanced
However, it should be noted that more than 50% of CKD because of the potential for hyperkalemia.
participants in intensive arm of SPRINT had not achieved „„ Modest physical activity of 150 minutes duration/

SBP target after 1 year. So targeting and achieving a lower week is useful for CKD population. It can be modified

Section-11.indb 949 28-01-2021 12:25:18


950 SECTION 11 Nephrology

depending on the cardiorespiratory fitness status and is associated with increase risk of hyperkalemia,
physical limitation of the individual. acute kidney injury, and hypotension.
„„ Other life style interventions include weight loss in „„ Calcium channel blockers (CCB)—dihydropyridine
obese people, limiting alcohol intake, and avoidance (DHP)-CCB (amlodipine) are frequently prescribed
of over the counter medications such as non-steroidal antihypertensives in CKD because:
anti-inflammatory drugs. —— they are the potent antihypertensive agents,

—— can work synergistically with ACEI/ARB,

Choosing Antihypertensive —— work well in volume expanded states, and

Drugs in CKD2,3,8,15 —— minimal side effect (pedal edema).

„„ Diuretics—
Most of the CKD patients require multiple —— Sodium retention and volume overload are
antihypertensives for adequate control of HTN. Certain
the main concerns of the majority of CKD
classes of antihypertensives are preferred because of their
patients. Diuretics act by natriuresis (promote
renoprotective, cardioprotective, and diuretics actions
sodium excretion through kidney), thus reduce
apart from antihypertensive effects. Before choosing
extracellular volume and has been shown to
antihypertensive agents in CKD we should look for
improve left ventricular mass index (LVMI)
comorbidities, risk benefit ratio of individual drug, volume
and arterial stiffness in CKD patients. Diuretics
status, and age of the patient.
„„ Angiotensin converting enzyme inhibitors (ACEIs)/
also enhance the BP lowering effect of ACEI/
ARB. Thiazide diuretics (hydrochlorothiazides
angiotensin receptor blockers (ARBs)—
—— ACEI/ARB are the agent of choice to treat HTN in
and chlorthalidone) are less effective at lower
glomerular filtration rate so switching to loop
diabetic CKD with severely increased proteinuria
diuretics (furosemide and torsemide) with frequent
(>300 mg/day). But the superiority of ACEI/ARB
over other agents (calcium channel blockers, dosing is done in advanced CKD. Some times loop
diuretics) is questionable in non-proteinuric CKD. diuretics can be combined with thiazide diuretics
However, as majority of CKD patients require to treat refractory edema in advanced CKD.
—— Diuretics should be avoided in end stage renal
multiple agents to achieve adequate control of
HTN, it is reasonable to include ACEI/ARB as add disease with no residual function and polycystic
on therapy in non-proteinuric CKD patients. kidney disease as they can lead to accelerated cyst
—— Th e c o m m o n s i d e e f f e c t o f AC E I / A R B i s growth and decreased renal function. The major
hyperkalemia particularly in advanced CKD hence adverse effects of diuretics are volume depletion
limiting their use despite their proven benefit. and electrolyte imbalance.
—— Mineralocorticoid receptor antagonists (MRA) are
Novel potassium binding agents (patiromer and
sodium zirconium cyclosilicate) in combination effective drugs for resistant HTN but may cause
w ith ACEI/ARB could change the way of hyperkalemia in patients with low-estimated
pharmacotherapy in HTN in CKD. But further glomerular filtration rate, so better to avoid at
research is required before their routine clinical estimated glomerular filtration rate below 45 mL/
use. min/m2.
—— Another potential problem with the use of ACEI/ „„ Beta blockers—
ARB is acute kidney injury. Recent guidelines —— Although beta blockers has lost credential in

suggest that up to 30% increase in serum creatinine treatment of primary HTN in general population,
in first few weeks and later on stabilization is an but still they are found to be useful adjunctive
acceptable physiological change which confers therapy in CKD population because they decrease
long-term renoprotection. sympathetic over activity and are cardioprotective.
—— Combination therapy (ACEI+ARB) has failed to A recent trial named HDPAL (HTN in hemodialysis
show any cardioprotection and renoprotection patients treated with atenolol or lisinopril) has
in major studies. Additionally this combination concluded that atenolol arm has less cardiovascular

Section-11.indb 950 28-01-2021 12:25:18


Management of Hypertension in Chronic Kidney Disease CHAPTER 146 951

events and better BP control than lisinopril arm in „„ Dialysate sodium restriction
hemodialysis patients. „„ Adequate time on dialysis
—— Other antihypertensive agents for patients with „„ Consideration of frequent dialysis
CKD include direct vasodilator (minoxidil and
hydralazine), centrally acting alpha adrenergic Managing HTN Following
agonist (clonidine), alpha blockers (prazosin), and
direct rennin inhibitors (aliskiren).
Kidney Transplantation1-3
„„ Resistant and refractory HTN in CKD1,3,16— HTN is common in post-transplant period. It has been
—— The prevalence is up to 40% as shown in CRIC reported that HTN is prevalent in more than 90% of
Study. It has got poor prognosis—increases the risk calcineurin inhibitors treated kidney transplant recipient.
of death by 30% and the risk of heart failure by 59%. The higher BP is associated with poor graft survival,
The selection of complementary medications and increased CVD risk, and most common cause of death
ensuring drug adherence often resolve treatment post-transplant. KDIGO and the ACC/AHA guidelines
resistance. Thiazide and thiazide-like diuretics in currently recommend a BP target <130/80 mm Hg. Some
combination with ACE/ARB or MRA can be very useful tips to manage HTN following kidney transplants
effective in treatment of CKD populations with are:
apparent drug resistant HTN. „„ Weight control

„„ Steroid and CNI dose optimization

Adherence2,9 „„ During 1st year after transplant, CCB (DHP) is the

preferred antihypertensive agent over ACEI/ARB


Adherence to therapy is poor due to frequent dosing of
„„ In patient with mild graft dysfunction with proteinuria,
pills, pill burden, drug interaction, and adverse effects.
ARB are the preferred drug
Strategies to improve adherence include:
„„ In case of post-transplant hyperuricemia losartan is
„„ Patients should be communicated regarding treatment
the preferred drug
and its importance.
„„ Use of combination pill and whenever possible long

acting, once a day medication to be used. Future Perspective21-25


In recent years, the underlying mechanism and unmet
2,17,18
Nocturnal Therapy therapeutic needs to halt the progression of CKD and CVD
have been better understood. Due to this few promising
Non-dipping is detected more frequently in later stages of
therapeutic measures have emerged. They are:
CKD and associated with significant CV death. Multiple
„„ Sodium glucose transport 2 inhibitor (SGLT-2I): Interest
clinical trials have shown an improvement in nocturnal
in molecules of this group has intensified following
dipping of BP by dosing at least one antihypertensive
the result of EMPA-REG OUTCOME (Empagliflozin
medication at bedtime.
Cardiovascular Outcome Event Trial in Type 2 Diabetes
Mellitus Patients) and CREDENCE (Canagliflozin and
Management of HTN in Renal Outcomes in Type 2 Diabetes and Nephropathy)
Patients on Dialysis2,3,19,20 trials, which demonstrated significant slowing of CKD
The relationship between BP and CVD in this group is progression and a reduction in the composite outcome
more complex. There is U-shaped relationship between of death from CVD. Combining ACEI/ARB with SGLT2I
24-hour ambulatory SBP and all cause mortality as noted may further reduce intraglomerular pressure by their
by Mayer et al. So BP treatment should be individualized synergistic effects.
in this group keeping in mind of comorbid conditions. „„ The DUET (Dual Endothelin Receptor and Angiotensin

Besides pharmacotherapy, these are the novel measures Receptor Blocker) trials with sparsentan and irbesartan
to treat HTN in dialysis patients: have shown promising results in control of HTN and
„„ Dry weight reduction (DRIP trial) proteinuria in IgA nephropathy and focal segmental
„„ Dietary sodium restriction glomerulosclerosis (FSGS).

Section-11.indb 951 28-01-2021 12:25:18


952 SECTION 11 Nephrology

„„ Renal denervation therapy (RDN): After initial hiccups 11. Wright JT Jr, Bakris G, Greene T, et al. Effect of blood pressure
this radio frequency energy based therapy for ablation lowering and antihypertensive drug class on progression of
hypertensive kidney disease: results from the AASK trial. JAMA.
of the networks of nerves around renal arteries has
2002;288(19):2421-31.
shown promising results in newer trials. However, this
12. Ruggenenti P, Perna A, Loriga G, et al. Blood-pressure control
strategy is still in experimental stage. for renoprotection in patients with non-diabetic chronic renal
disease (REIN-2): multicentre, randomised controlled trial. Lancet.
Conclusion 2005;365(9463):939-46.
13. Cushman WC, Evans GW, Byington RP, et al. Effects of intensive
The Prevalence of HTN in CKD is very high, but HTN control rate blood-pressure control in type 2 diabetes mellitus. N Engl J Med.
is far from optimal. When CKD and HTN coexist, CV morbidity 2010;362(17):1575-85.
and mortality is substantially increased. Personalized and 14. Wright JT Jr, Williamson JD, Whelton PK, et al. A randomized trial
individualized HTN therapy using standardized office BP and of intensive versus standard blood-pressure control. N Engl J Med.
home BP monitoring holds great promise. ACEI or ARB, CCB, 2015;373(22):2103-16.
appropriate diuretic therapy, dietary salt restriction, and 15. Sinha AD, Agarwal R. Clinical pharmacology of antihypertensive
ensuring proper adherence to therapy make up the foundation therapy for the treatment of hypertension in CKD. Clin J Am Soc
for the treatment of HTN in CKD. Nephrol. 2019;14(5)757-64.
16. Muntner P, Anderson A, Charleston J, et al. Hypertension awareness,
treatment, and control in adults with CKD: results from the

References Chronic Renal Insufficiency Cohort (CRIC) Study. Am J Kidney Dis.


2010;55(3):441-51.
1. Richard J, Feehally J, Floege J, et al. (Eds). Comprehensive Clinical 17. Judd E, Calhoun DA. Management of hypertension in CKD: beyond
Nephrology, 5th edition. Elsevier Saunders; 2014. p. 1320. the guidelines. Adv Chronic Kidney Dis. 2015;22(2):116-22.
2. Pugh D, Gallacher PJ, Dhaun N. Management of hypertension in 18. Hermida RC, Ayala DE, Mojon A, et al. Bedtime dosing of
chronic kidney disease. Drugs. 2019;79(4):365-79. antihypertensive medications reduces cardiovascular risk in CKD. J
3. Ku E, Lee BJ, Wei J, et al. Weir hypertension in CKD: core curriculum Am Soc Nephrol. 2011;22(12):2313-21.
2019. Am J Kidney Dis. 2019;74(1):120-31. 19. Mayer CC, Matschkal J, Sarafidis PA, et al. Association of ambulatory
4. Coleman TG, Guyton AC. Hypertension caused by salt loading in blood pressure with all-cause and cardiovascular mortality in
the dog. 3. Onset transients of cardiac output and other circulatory hemodialysis patients: effects of heart failure and atrial fibrillation. J
variables. Circ Res. 1969;25(2):153-60. Am Soc Nephrol. 2018;29(9):2409-17.
5. Krapf R, Hulter HN. Arterial hypertension induced by erythropoietin
20. Agarwal R, Alborzi P, Satyan S, et al. Dry-weight reduction
and erythropoiesis-stimulating agents (ESA). Clin J Am Soc Nephrol.
in hypertensive hemodialysis patients (DRIP): a randomized,
2009;4:470-80.
controlled trial. Hypertension. 2009;53(3):500-7.
6. Agrawal A, Agarwal SK, Kaleekal T, et al. Rifampicin and anti-
21. Wanner C, Lachin JM, Inzucchi SE, et al. Empagliflozin and
hypertensive drugs in chronic kidney disease: pharmacokinetic
clinical outcomes in patients with type 2 diabetes, established
interactions and their clinical impact. Indian J Nephrol.
cardiovascular disease and chronic kidney disease. Circulation.
2016;26(5):322-8.
2018;137(2):119-29.
7. Whelton PK, Gidding S, Carey RM, et al. 2017 ACC/AHA/AAPA/
22. Perkovic V, Jardine MJ, Neal B, et al. Canaglifloin and renal
ABC/ACPM/ AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the
Prevention, Detection, Evaluation, and Management of High Blood outcomes in type 2 diabetes and nephropathy. N Engl J Med.
Pressure in Adults A Report of the American College of Cardiology/ 2019,380(24);2295-306.
American Heart Association Task Force on Clinical Practice 23. Herrington WG, Preiss D, Haynes R, et al. The potential for improving
Guidelines. Hypertension. 2018;71(6):e13-e115. cardio-renal outcomes by sodium-glucose co-transporter-2
8. Cheung AK, Chang TL, Cushman WC, et al. Blood pressure in inhibition in people with chronic kidney disease: a rationale for the
chronic kidney disease: conclusions from a Kidney Disease: EMPA-KIDNEY study. Clin Kidney J. 2018;11(6):749-61.
Improving Global Outcomes (KDIGO) Controversies Conference. 24. Komers R, Gipson DS, Nelson P, et al. Efficacy and safety of
Kdigo Executive Conclusions. 2019;95(5):1027-36. sparsentan compared with irbesartan in patients with primary focal
9. Kramer HJ, Townsend RR, Griffin K, et al. KDOQI US Commentary segmental glomerulosclerosis: randomized, controlled trial design
on the 2017 ACC/AHA Hypertension Guideline. Am J Kidney Dis. (DUET). Kidney Int Rep. 2017;2(4):654-64.
2019;73(4):437-58. 25. Townsend RR, Mahfoud F, Kandzari DE, et al. Catheter-based renal
10. Klahr S, Levey AS, Beck GJ, et al. The effects of dietary protein denervation in patients with uncontrolled hypertension in the
restriction and blood pressure control on the progression of absence of antihypertensive medications (SPYRAL HTN-OFF MED):
chronic renal disease. Modification of Diet in Renal Disease Study a randomised, sham-controlled, proof-of-concept trial. Lancet.
Group. N Engl J Med. 1994;330(13):877-84. 2017;390(10108):2160-70.

Section-11.indb 952 28-01-2021 12:25:18


CHAPTER

147 Prevention of Progression of


CKD—What Is New?
Krishnaswamy Sampathkumar

Abstract 
Chronic kidney disease (CKD) afflicts approximately 7–10% of adult population of India. It is a silent killer due to its
far reaching ill effects on the cardiovascular system. The manifestations of CKD are varied and can mimic a variety of
medical and surgical conditions. Early recognition is of paramount importance since preventive strategies are successful
only if applied before irreversible renal fibrosis sets in. This article gives an overview of recent evidences. Dietary protein
restriction should be applied to meat based diets and not for vegetable proteins. Plant-based protein intake has been
shown to be renoprotective due to its alkaline nature and fiber content. RAS blocker therapy, and antihypertensive therapy
are the inseparable duo of prevention. A dose escalating strategy of RAS blockade aiming for proteinuria reduction has
been successfully applied in the Indian context. However, it requires close serum creatinine and potassium monitoring to
prevent harm. Based on well conducted large trials, SGLT2-inhibitors have provided substantial benefits in terms of renal,
cardiac, and all cause mortality. Their use is recommended for both diabetic and non-diabetic renal diseases. Sodium
bicarbonate and statin usage have shown benefits. However, uric acid lowering therapy has failed to provide beneficial
effects in recent trials.

Introduction Contributors to Accelerated


Chronic kidney disease (CKD) is a silent killer, which GFR Loss
afflicts approximately 7–10% of world’s adult population. It
The glomerular filtration rate (GFR) falls at a rate of
means that India which makes up 17% of world population
approximately 0.5 mL/min/year in the normal adult
carries a large burden of the disease. 1 It is commonly
population. Patients with progressive CKD the loss of
mistaken for a number of conditions such as nutritional
GFR may be as steep as 5–15 mL/min/year. In addition
anemia, nutritional rickets, peptic ulcer disease,
to non-alterable factors such as age, gender, and genetics
dysfunctional uterine bleeding, etc. In the absence of a
National Registry, the exact prevalence of the disease is a number of factors which are potentially treatable
open to conjecture. A population based study showed contribute to the risk of progression. A recent work
incidence of End Stage Renal Disease (ESRD) to be 159 showed genetic polymorphisms involving intracellular
per million population.2 Once a patient reaches ESRD the antioxidant systems can accelerate the progression and
treatment is not only prohibitively expensive but survival mortality dramatically.3 Dietary factors, lack of exercise,
rates are worse than advanced malignancy. Thus every albuminuria, hypertension, hyperglycemia, lipids, morbid
physician should update himself with current practice obesity, and metabolic acidosis, mini AKIs, and gut
guidelines for optimal prevention of progression of CKD. dysbiosis are some of the modifiable risk factors.

MU-147.indd 953 29-01-2021 15:02:04


954 SECTION 11 Nephrology

Lifestyle Modifications Albuminuria


Earlier view—Albuminuria is a marker of severity of renal
Diet
disease.
Earlier view—Restricting all forms of proteins in the diet Current view—It is both a marker and maker of
retards the progression of CKD. progression of renal disease.
Current view—Plant based proteins are renal friendly Albuminuria is both a marker and maker of glomerular
and should be promoted. injury. A direct quantitative relationship exists between
In a community-based study of adults without the higher proteinuric CKD and faster progression. Filtered
CKD, higher adherence to a healthy plant-based diet albumin is taken up by the proximal convoluted tubule
or a provegetarian diet was associated with lower CKD cells where it is toxic and inflammatory. Tubulointerstitial
risk. 4 Higher consumption of healthy plant foods fibrosis sets in faster when the rates of proteinuria are
(fruits, vegetables, whole grains, nuts, legumes, coffee, higher. RAS blockers have shown to reduce proteinuria
tea) was associated with a lower risk and slower eGFR and prevent progression of renal disease. Highest tolerable
decline in GFR. Dietary red meat contains cardiotoxic doses of angiotensin receptor blockers (ARBs) and
metabolites from lecithin and carnitine such as angiotensin converting enzyme (ACE) inhibitors are
trimethylamines. Plant-based proteins are recommended recommended to provide optimal renoprotection. Higher
for a variety of reasons such as higher fiber content, lesser doses are accompanied by hyperkalemia. An important
diet acid load, less phosphorus and uric acid generation, advance has been the clinical entry of New K binding agents
enhanced vitamin K (reduction of coronary calcium like zirconium and patiromer, which permit continued
scores), and promotion of healthy gut microbiome. and safe use of RAS blockade in the face of hyperkalemia.
Dals, lentils, and Soya beans can be substituted for In the OPAL-HK study involving CKD patients with
dietary animal proteins such as meat and dairy products. hyperkalemia, treatment with patiromer reduced the
Daily intake of locally available fruits and vegetables serum potassium levels and enabled significantly more
are recommended in CKD Stages 3–4. Compensatory patients to continue RAS blockers without interruption
potassium excretion from the colon increases when dietary than placebo.5 The drug was well tolerated.
fiber intake is liberalized. Vegetable diet also inhibits the
enzyme 11-beta-hydroxysteroid dehydrogenase type 2 Hypertension
in the distal convoluted tubule cells which increase the
Earlier target—BP reduction below 130/80.
cortisol induced K excretion in addition to aldosterone
Current view—More intensive BP reduction below
effect.
120–125/<80 in proteinuric CKD.
Long-term follow-up of 15 years in MDRD and AASK
Physical Exercise trials highlighted that intensive BP control reduced both
Physical deconditioning sets in during progressive overall mortality and progression to ESRD. The recently
CKD, which results in muscle energy dysmetabolism. released SPRINT trial data showed that in CKD subgroup
A reduction in mitochondrial activity develops even in the all cause mortality was lower in the intensive arm. 6
earlier stages of CKD, which is progressive. Oxidative However, the incidence of ESRD or a 50% decline in GFR
stress, inflammation, erythropoietin, vitamin D, and was not altered in the intensive arm. However, a note of
testosterone deficiencies are possible reasons. The caution is that diastolic BP reduction below 70 mm Hg is not
symptoms include easy fatigability and proximal muscle recommended in older adults with atheromatous central
weakness. Later, muscle atrophy sets in. As in general arteries, which can lead onto coronary insufficiency.
population, the cardiovascular benefits of exercise cannot ARBs and ACE inhibitors are the preferred first-line
be ignored in CKD patients. Regular moderate intensity agents. Maximally tolerated doses of these drugs should
exercises involving all the major core groups of muscles be given. As an example losartan is started on 50 mg OD
are recommended. Both endurance and weight bearing and titrated up depending on proteinuria reduction and
exercises are indicated in Stage 3 CKD. BP reduction to 50 mg BID. Supra pharmacological doses

MU-147.indd 954 29-01-2021 15:02:04


Prevention of Progression of CKD—What Is New? CHAPTER 147 955

can be prescribed with a close, watch on serum K and BP. tubule cells following SGLT2 inhibitor therapy. This
The dose required to optimize GFR reduction was 150±88 results in more sodium ions reaching the DCT and
mg/day in an Indian Study. 7 Though combinations of thereby producing afferent arteriolar vasoconstriction.
ACEi and ARBs are prohibited by most of the guidelines, This ultimately results in renal preservation since
individualization of therapy can result in better outcomes. intraglomerular pressure is lessened.
Since multiple drugs are required in CKD, calcium GLP1 analogs (liraglutide) have also shown both
channel blockers, diuretics (Thiazides in Stage 3 and loop renal and cardiovascular benefits in trials. In the LEADER
diuretics in Stage 4), are added in a stepwise fashion. trial involving liraglutide new onset of persistent
Dietary sodium restriction to below 2–3 gm/day is macroalbuminuria, occurred in significantly fewer
an important adjunct to improve BP control. Due to participants in the liraglutide group than in the placebo
concerns about hyperkalemia, there is reluctance to group (hazard ratio, 0.74).10 The reduction in GFR in the
start spironolactone therapy in those with resistant placebo arm was −5 mL/min/year, which reduced to −0.3
hypertension. Recently published AMBER study showed mL/min/year in the treatment group.
that when patiromer is added to spironolactone more In contrast, DPP4 inhibitors have not shown reno- or
patients could continue the latter drug without developing cardioprotective benefits in trials. But they are still useful
hyperkalemia.8 agents since the incidence of hypoglycemia is less, which
Canrenone and Finerenone are novel non-steroidal enhances the acceptability of the drug in CKD population.
aldosterone receptor antagonists with low risk of
The recommended target HbA1C level for the
hyperkalemia. Current prospective trials are testing their
prevention of progression of CKD is between 6.5–7%.
efficacy and safety in CKD patients.
However, in those with higher comorbidities who are
prone for complications of hypoglycemia, one can opt for
Glucose Control
a compromised target of 7.5–8%.
Earlier view—Insulin therapy for Stage 3 CKD onward.
Re c e nt v i e w —S G LT 2 i n h i b i t o r s a re s t ro ng l y Acidosis Correction
recommended to prevent progression of CKD and reduce
R e c e n t v i e w —S o d i u m b i c a r b o n a t e t h e r a p y i s
cardiovascular morbidity and mortality.
nephroprotective.
CREDENCE trial on canagliflozin versus placebo
The serum bicarbonate levels are ideally maintained
on CKD patients has shown remarkable renal and
above 22 meq/L in patients with CKD. Acidosis correction
cardiovascular benefits for patients with diabetic
nephropathy.9 All the patients had an estimated GFR of improves bone health and retards progression of renal
30 to less than 90 mL/minute and albuminuria and were disease. The latter is due to reduced ammonia generation,
treated with renin-angiotensin system blockade. The which is linked to inflammation by stimulation of
primary outcome was a composite of end-stage kidney alternate complement pathway. A condition called
disease, or a doubling of the serum creatinine level, or normobicarbonatemic acidosis develops in CKD, which
death from renal or cardiovascular causes. Over a 2.6-year is characterized by reduced urinary excretion of citrate.11
period, the relative risk of the primary outcome was 30% This can be addressed by providing fruits and vegetables,
lower in the canagliflozin group than in the placebo group which are a rich source of citrate.
(hazard ratio, 0.70). The relative risk of the renal-specific Sodium bicarbonate is commonly given by oral route
composite of end-stage kidney disease, a doubling of the in tablet or capsule forms. Adult dose is between 1.5–2.5
creatinine level, or death from renal causes was lower by gm/day depending on the degree of acidosis. Each gram
34% (hazard ratio, 0.66) and the relative risk of end-stage contains 12 mEq of base. The potential side effects include
kidney disease was lower by 32% (hazard ratio, 0.68). gastic distension and metabolic alkalosis. Aggravation of
Cardiovascular events were also significantly less in the hypertension is uncommon. It decreases the absorption
treatment arm. Risk of amputations or fractures were of ranitidine and increases the propensity of quinolones
not significantly elevated in the treatment arm. Sodium for crystalluria. It is contraindicated in hypocalcemia and
reabsorption is inhibited in the proximal convoluted hypokalemia.

MU-147.indd 955 29-01-2021 15:02:04


956 SECTION 11 Nephrology

Management of Hyperlipidemia has been shown to reduce the serum levels of products
Earlier view—Statin therapy is beneficial at the earlier of protein metabolism like p-cresol and indoxyl sulfate,
stages of CKD. No new data have emerged recently. which induce cardiovascular toxicity.
Observational studies among apparently healthy
individuals or in patients with preexisting cardiovascular Obesity
disease (CVD) have shown a roughly linear relationship Earlier view—Weight loss should be routinely advised in
between risk of cardiovascular-related death and serum CKD patients.
total and low-density lipoprotein (LDL) cholesterol. Recent view—Mildly overweight category of patients
Among patients with CKD, however, this relationship is show best survival.
much less obvious. The cardiovascular abnormalities The association between body mass index (BMI) and
in Stage 4–5 CKD are progressive medial vessel wall mortality has shown some surprising findings. In a large
calcification, left ventricular hypertrophy (LVH) and observational study from US veterans found that best
diastolic dysfunction. These are poorly responsive to statin survival of Stage 3–4 CKD was seen in those with BMI
therapy. There are no new evidences in the last decade in between 25 and 35 kg/m 2. In morbidly obese patients,
this field in the aftermath of SHARP trial, which showed a when bariatric surgery was undertaken, both proteinuria
significant, 17% reduction in major atherosclerosis events and GFR reduction are attenuated.12
and a 15% reduction in major cardiovascular events in
the entire cohort of 9,000 patients. The earlier the statin Uric Acid
therapy is initiated, the better were the cardiovascular Earlier view—Treatment of hyperuricemia is reno­
protection benefits. protective.
Recent view—Hyperuricemia correction does not
Prevention of AKI result in renal benefits.
Earlier view—Recovered AKI patients regain normal renal Uric acid produces inflammation and proliferation of
function. afferent arterioles in mouse models of CKD. Extracellular
Current view—Up to 30% of patients with comorbidities precipitation in the form of microscopic crystals and
who develop AKI may progress to CKD. macroscopic urate stones can cause structural damage.
In CKD patients multiple episodes of AKI are the cause Intracellular mechanisms show its propensity to precipitate
of accelerated decline in GFR. injury relating to its oxidative properties. Humans and
The downtrending of GFR slope from above 90 mL/min other primates are prone to hyperuricemia because
to 10 mL/min does not occur in a straight line if plotted enzyme uricase was lost in evolutionary mutation. To
against time. There are periods of flattening interspersed lower uric acid levels, xanthine oxidase inhibitors (XOIs)
with periods of acute reduction. These “Mini AKIs” due such as allopurinol and febuxostat are commonly used in
to volume depletion, cardiac failure, UTI, hypotension, clinical arena.
and NSAID drug toxicity result in incomplete recovery to
original levels. It is critically important that these events Does Reduction of Uric Acid
are aggressively managed. Produce Renal Benefits?
The short answer seems to be no. Based on many
Intestinal Microbial Flora observational studies which reported that higher serum
Recent view—It is beneficial to alter the dysbiosis, which is uric acid levels heightened the risk for new-onset CKD as
prevalent in CKD patients. well hastened its progression, a commonly held view was
Human gastrointestinal (GI) tract is home to trillions that a level above 7 mg% required intervention for renal
of bacteria, which undergo both a qualitative and benefits. However, two large, high-quality trials, which
quantitative alterations in CKD. Products of bacterial were published recently refuted a causal relationship and
metabolism are linked to inflammation, uremic syndrome, revealed that urate-lowering therapy neither prevents
and cardiovascular disease. Oral ingestion of probiotics CKD nor slows down its progression.

MU-147.indd 956 29-01-2021 15:02:04


Prevention of Progression of CKD—What Is New? CHAPTER 147 957

PERL trial randomized over 500 adults with type 1 References


diabetes and early-to-moderate diabetic kidney disease,
1. Varma PP. Prevalence of chronic kidney disease in India—where are
with allopurinol versus placebo. At 3 years treatment with we heading? Indian J Nephrol. 2015;25(3):133-5.
allopurinol had no effect on the change in measured GFR 2. Jha V. Current status of end-stage renal disease care in India and
compared with placebo; worryingly, allopurinol increased Pakistan. Kidney Int Suppl. 2013;3(2):157-60.
albuminuria, and nonsignificantly increased the rate of 3. Vasudevan V, Ramprasath T, Sampathkumar K, et al. GSTM1-null
allele predicts rapid disease progression in nondialysis patients and
fatal or nonfatal cardiovascular events. In the CKD-FIX
mortality among South Indian ESRD patients. Mol Cell Biochem.
study of over 350 adults with more advanced CKD, the rate 2020;469(1-2):21-8.
of decline of estimated GFR at 2 years was the same with 4. Kim H, Caulfield LE, Garcia-Larsen V, et al. Article plant-based diets
allopurinol and placebo. and Incident CKD and kidney function. Clin J Am Soc Nephrol.
Anemia correction and correction of calcium 2019;14(5):682-91.
phosphorus dysmetabolism have not been shown to have 5. Weir MR, Bakris GL, Bushinsky DA, et al. Patiromer in patients with
kidney disease and hyperkalemia receiving RAAS inhibitors. N Engl
any effect on the progression of CKD, and hence they are
J Med. 2015;372(3):211-21.
not discussed at length. But they have to be addressed 6. Wright JT, Williamson JD, Whelton PK, et al. A randomized trial of
all the same since cardiovascular and skeletal health are intensive versus standard blood-pressure control. N Engl J Med.
inseparable sufferers from renal dysfunction. 2015;373(22):2103-16.
Thus a multipronged strategy implemented with 7. Limesh M, Annigeri RA, Mani MK, et al. Retarding the progression
of chronic kidney disease with renin angiotensin system blockade.
attention to minutiae will be needed. Though the task
Indian J Nephrol. 2012;22(2):108-15.
looks daunting, it is achievable if pursued with dedication 8. Agarwal R, Rossignol P, Romero A, et al. Patiromer versus placebo to
as shown by Prof. M. K. Mani at Chennai. enable spironolactone use in patients with resistant hypertension
and chronic kidney disease (AMBER): a phase 2, randomised, double-
Conclusion blind, placebo-controlled trial. Lancet. 2019;394(10208):1540-50.
9. Perkovic V, Jardin MJ, Neal B, et al. Canagliflozin and renal outcomes
CKD can be termed as An Orphan Disease since awareness in type 2 diabetes and nephropathy. N Engl J Med. 2019;380:
levels are low even amongst the physicians. The four most 2295-306.
effective strategies for its prevention are RAS Blockade, SGLT2 10. Kalra S. Follow the LEADER—Liraglutide Effect and Action in
Diabetes: Evaluation of Cardiovascular Outcome Results Trial.
inhibitor therapy, Hypertension control, and Blood Glucose
Diabetes Ther. 2016;7(4):601-9.
control. In the years to come personalized medicine looking at
11. Maalouf NM, Moe OW, Gianella FG, et al. Spot urinary citrate-to-
the genetic makeup of the patient may become an added tool creatinine ratio is a marker for acid-base status in chronic kidney
to decide the therapeutic options. disease. Challenge parameter defense mechanisms excretion
results acid. Kidney Int. 2020.
12. Jun LL, Kamyar KZ, Jennie Z, et al. Association of body mass
index with outcomes in patients with CKD. J Am Soc Nephrol.
2014;25(9):2088-96.

MU-147.indd 957 29-01-2021 15:02:04


CHAPTER
Lupus Nephritis—
148 Current Understanding
and Management
Krishan Lal Gupta, Joyita Bharati

Abstract 
Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease, and lupus nephritis (LN) is associated with
increased morbidity and mortality. Kidney biopsy is essential and the “gold standard” for diagnosing LN. Extra-glomerular
involvement is seen in up to two-thirds of patients with LN and is associated with poor outcomes. The revised 2018
International Society of Nephrology/Renal Pathology Society classification for LN has changed parameters for activity index
and redefined few pathological parameters. Repeat kidney biopsy is done for resistant disease or during flare, usually when
atypical features are present. Protocol biopsy is a promising tool to monitor patients with LN and decide maintenance
therapy. Newer therapies like multi-targeted therapy and biological agents such as obinutuzumab have shown promise in
treating LN.

Introduction Pathology of Lupus Nephritis


Lupus nephritis (LN) is seen in 25–50% of patients at the LN is initiated by deposition of circulating immune
outset of systemic lupus erythematosus (SLE) and about complexes and by binding of autoantibodies to antigens in
60% of the patients are affected during the disease course, the glomerulus and vessel walls. This interaction activates
which varies with race and ethnicity.1-3 The probability complement mediated cytotoxicity, macrophage, and
of a new renal flare is 20–30% per patient-year of follow- natural killer cell-induced cell cytotoxicity along with
up.4 Patients with LN, particularly those with proliferative Fc receptor-based T-cell mediated cell damage. Anti-
histology, are at 2.28 times higher risk of mortality as phospholipid antibodies can result in thrombotic lesions
compared to those without LN and up to one-third of in the glomerulus and in the vessels. In 20–30% of patients
them progress to end stage renal disease (ESRD) within with LN, anti-neutrophil cytoplasmic antibody can
15 years. 5,6 The prevalence of SLE and the chances of be positive and may contribute to the development of
developing LN vary considerably between different vasculitic lesions.12
regions of the world, with higher rates in Africans and
Hispanics.7 Most often, LN occurs early (within 5 years) Value of Renal Biopsy
in the disease course of SLE.8 SLE is more prevalent in Treatment of LN is guided by the histological patterns
women than men across all age groups and populations; described in the classification system 2003 International
the female-to-male ratio is highest at reproductive age, society of Nephrology/Renal Pathology Society (ISN/RPS)
ranging between 8:1 and 15:1, and is lowest in prepubertal classification. Clinical severity does not always correlate
children at about 4:3.9-11 with histological severity and is shown in Table 1.

MU-148.indd 958 29-01-2021 15:01:50


Lupus Nephritis—Current Understanding and Management CHAPTER 148 959

TABLE 1 Clinicopathological correlation in lupus nephritis

Lupus nephritis class Proteinuria Active urine Increased serum Active lupus Hypertension
(ISN/RPS) sediment creatinine serology

Class I –* – – +/– –

Class II +/– * – – +/– –

Class III ++ (NS: 30%) + +/– +/– +/–

Class IV +++ (NS: 50%) ++ +/– ++/– ++/–

Class V +++ (NS: 60%) – +/– +/–– +/–

Class VI ++ +/– + +/–– +


(+) represents frequent occurrence of symptom/sign, (–) represents infrequent occurrence of symptom/sign, NS: Nephrotic syndrome; * NS is
seen in class I/II LN in presence of podocytopathy

Therefore, renal biopsy remains indispensable and the Extraglomerular Involvement


“gold standard” to correctly classify LN. The Kidney Disease Tubular atrophy and interstitial fibrosis (IFTA) are one of
Initiative Global Outcomes (KDIGO),13 American College the strongest predictors of renal failure. Various studies
of Rheumatology (ACR),14 and European League Against reported vascular involvement in LN to indicate increased
Rheumatism/European Renal Association-European risk for progression to renal failure. In a study from our
Dialysis and Transplantation Association (EULAR/ERA- center,9 of total 197 patients, thrombotic microangiopathy
EDTA)15 guidelines recommend that a renal biopsy should (TMA) was found in 25.4% with 10% having concomitant
be performed to confirm and classify LN in cases of SLE APS. Patients of LN with TMA had significantly higher
with clinical manifestation of proteinuria (>500 mg/day), rates of oliguria (p=0.035), advanced renal injury, that
urinary red blood cells (>5 per high power field) or cellular is, serum creatinine more than 3 mg/dL (p=0.002),
casts (any), or unexplained renal dysfunction. fibrocellular and fibrous crescents (p=0.01), and tubular
Classification of LN: The classification of LN (based on atrophy (0.001) compared to non-TMA patients. They also
histology) has been in place for over five decades with the had higher rates of treatment failure (p=0.02) compared
latest modification in 2018. to the group without TMA.9 Another study (unpublished
data) from our center included 241 patients with LN with
2003: ISN/RPS16 proposed a new classification system
60% having tubulointerstitial involvement, 32.3% having
that would provide better clinical correlation, uniformity
vascular involvement. Those with tubulointerstitial and
in reporting and reproducibility. The classification
vascular involvement had lower rates of remission.
schema is based on glomerular pathology, which
includes assessment by light microscopy (LM) and Role of Repeat Biopsy
immunofluorescence (IF). Class IV LN is the commonest
Repeat biopsy is done during renal flare for refractory
pattern seen worldwide. In a study from our center, of total
LN or as a protocol after induction and/or maintenance
232 patients, Class II was seen in 21, Class III in 36, Class IV
therapy for LN. Repeat biopsy is useful as histological
in 130, and Class V in 30 patients.17
transformations are common and may affect treatment
2018: Changes in the 2018 ISN/RPS18 revision were: the decisions. It has also been shown to be predictive of
distinction between class IV-S and IV-G was not evidence long-term outcomes. Protocol re-biopsies can guide
based and hence, has been removed; the threshold for therapeutic decision as 20–50% of patients in clinical
defining crescent is lowered and various types of crescents complete response after therapy were shown to have
are defined in addition; activity and chronicity scores are histological activity on the biopsy.19 Furthermore, protocol
modified and are required to be mentioned instead of A biopsy after prolonged clinical remission can help in
(activity) or C (chronicity). deciding safe withdrawal of maintenance therapy, as

MU-148.indd 959 29-01-2021 15:01:50


960 SECTION 11 Nephrology

was shown recently in a prospective study on clinically The NIH regimen includes monthly intravenous pulses of
quiescent LN patients. 20 Recently, the EULAR 2019 21 CYC at a dose of 500–1,000 mg/m2 for 6 months followed
guideline has recommended repeat biopsy in those with by maintenance with quarterly infusions at same dose for
incomplete renal response defined as 24-hour proteinuria the next 2 years.24 Using this protocol, remission could be
more than 0.8–1 g/day with stable/improved renal achieved in 61% of patients at the end of induction phase
function despite at least 1 year of immunosuppressive and there were no relapses in the 2-year follow-up period.
treatment. In a study from our center, which included A similar study at our center revealed overall response rate
62 patients who underwent repeat biopsy for clinical of 70% (Complete + Partial remission) and 30% failure rate
indications, we found histological transformations in (Failure 20% + Death 10%).25 Major side effects associated
61.3% of the patients.22 Class switch from proliferative to with CYC such as leukopenia, infections, gonadotoxicity,
non-proliferative occurred in 13.7% and 18.2% of patients hemorrhagic cystitis, and risk of malignancies in long-
with non-proliferative histology switched to proliferative term are associated with cumulative dose received. Fixed
classes. On repeat biopsy, endocapillary proliferation and low-dose intravenous CYC, that is, six doses of 500 mg
fibrinoid necrosis decreased whereas glomerulosclerosis every 2 weekly followed by azathioprine (2 mg/kg) as
and IFTA increased (P<0.001). Presence of IFTA more than maintenance agent was compared with NIH regimen in
30% and TMA on the second biopsy correlated with worse the European Lupus Nephritis Trial (ELNT).26 This trial,
long-term outcome.22 which included predominantly Caucasians and excluded
severe renal involvement, showed no difference in long-
Management of Lupus Nephritis term outcomes like patient survival, renal survival, or
doubling of serum creatinine in the two arms. Another
Treatment of LN involves use of aggressive immuno­
trial27 done at our center comparing CYC given as ELNT
suppressive therapy in the induction phase to achieve
protocol with mycophenolate mofetil (MMF) for induction
rapid control of inflammation in the kidneys followed by
in patients with LN showed similar renal outcomes in both
less intense immunosuppression to maintain remission.
the arms. A total of 100 patients were equally randomized
The initial phase for induction is for a duration of 3–6
to receive either CYC or MMF. At 24 weeks, 37 patients in
months followed by a maintenance phase of 3–5 years
each group achieved the primary end point. The complete
usually.
remission rate was 50% in CYC and 54% in MMF group.

Induction Agents Mycophenolate mofetil: MMF was first used in induction


phase in a pilot study conducted in Chinese patients
Corticosteroids: Steroids form the backbone of induction
with class IV LN and was shown to be non-inferior to
therapy of active LN. However, there is an increasing
oral CYC.28 Subsequently, a multiethnic study comparing
concern about the side-effects with high and prolonged
MMF with intravenous CYC, by Ginzler et al.29 showed
doses of steroids. Many newer trials have investigated this
non-inferiority of MMF as compared to CYC. One of the
by trying to compare regimens, which include introduction
largest multicentric randomized control trials by Appel et
of lesser toxic novel agents to allow reduction of steroid
al.,30 Aspreva Lupus Management Study (ALMS) involving
dose versus the standard induction therapy. Based on
370 patients comparing intravenous monthly CYC with
these concerns and the existing literature, the 2019
MMF showed similar clinical response rates (53% vs. 56%,
EULAR-ERA/EDTA guideline23 for LN has recommended
respectively) at the end of 6 months in both the arms.
the use of methylprednisolone pulse (at doses between
MMF had better response rates compared to CYC in Blacks
500–2,500 mg) followed by oral prednisolone of 0.3–0.5
and Hispanics (60% vs. 39%).
mg/kg/day and a rapid taper to reach less than 7.5 mg/day
Although guidelines for management of LN including
by 3–6 months.
ACR, EULAR/ERA-EDTA, and KDIGO recommend CYC or
Cyclophosphamide: Intravenous cyclophosphamide MMF as agents for induction in LN, high dose CYC (NIH
(CYC) along with corticosteroids became the standard regimen) is routinely reserved for severe renal disease
of care for treatment of LN based on randomized control (Sr Creatinine >3 mg/dL), crescentic glomerulonephritis,
trials conducted by the National institute of health (NIH). TMA, and in the presence of concomitant extraglomerular

MU-148.indd 960 29-01-2021 15:01:51


Lupus Nephritis—Current Understanding and Management CHAPTER 148 961

lesions like central nervous system (CNS) lupus or diffuse 0.6 mg/kg/day of prednisolone), called multitargeted
alveolar hemorrhage. therapy (MTT), as induction therapy was compared with
intravenous monthly CYC (0.75 g/m2) in 368 Chinese
Calcineurin inhibitors (CNIs): CNIs act by inhibiting T-cell
patients with active LN. MTT was superior to CYC, with
activation and, in turn, decreasing B-cell activation and
complete remission rates being 46% versus 26% at the
antibody production. Furthermore, it stabilizes the actin
end of 6 months. However, rates of serious infections were
cytoskeleton in podocytes, thus contributing to reduction
higher in the MTT group.35
in the degree of proteinuria. However, long-term uses of
CNIs are associated with nephrotoxicity and there is a
higher chance of relapse once the drug is withdrawn. Maintenance Therapy
After the induction phase, maintenance immuno­
Cyclosporine: Cyclofa-Lune study31 involved 40 patients
suppressive agent is necessary to reduce the risk of
of proliferative LN who were randomized to Cyclosporine
flares and further renal damage. NIH trials showed that
A and intravenous CYC. Clinical outcomes at the end of
maintenance with quarterly pulses of intravenous CYC
induction phase and maintenance phase were similar
for a period of 2 years had lesser episodes of relapses
between two groups; response rates were 52% versus 43%
in comparison to no maintenance. 24 Contreras et al. 36
in CYC and Cyclosporine arms, respectively at the end
showed that maintenance with MMF or azathioprine was
of induction, and they were 38% versus 58% for overall
better than quarterly pulses of CYC in terms of efficacy and
response at the end of maintenance phase. The extended
side effects.
follow-up (median: 7.7 years) of these patients showed
similar rates of renal survival, patient survival, and MAINTAIN trial:37 It included 105 predominantly white
proteinuria in the two arms.32 patients induced with intravenous CYC by ELNT regimen
and were followed up for 5 years. There was no statistical
Tacrolimus: Comparison of tacrolimus with MMF for
difference between rates of renal flare between MMF
induction in active LN patients showed similar rates of
(19%) and azathioprine (25%) maintenance, respectively.
complete remission at the end of 6 months.33 In those
with pure membranous LN, tacrolimus resulted in higher ALMS maintenance trial:38 It included 227 patients who
response rates of 100% as compared to 75% in MMF group. achieved remission with MMF induction and were
However, the rates of relapses by the end of 5 years were randomized to MMF and azathioprine maintenance. At
higher in those who received tacrolimus (62%) than those 36 months, composite outcome which included renal
who received MMF (42%).34 flare, ESRD, doubling of serum creatinine, death, or use of
rescue therapy was significantly lesser in the MMF group
Voclosporin: Voclosporin is a structural analogue (trans-
than the azathioprine group (16.4% vs. 32.4%). The ALMS
isomer) of cyclosporine which is fourfold more potent
maintenance trial, which included multiethnic patients,
than cyclosporine and is associated with lesser off-target
larger number of patients, and had compared composite
side effects such as cosmetic effects and nephrotoxicity. In
outcomes, favors use of MMF as maintenance agent
addition, it does not require drug level monitoring owing
over azathioprine. The ACR14 and KDIGO13 guidelines
to stable pharmacokinetics. In a recently completed phase
recommend use of either MMF or azathioprine as
2 trial on patients with LN, voclosporin, added to MMF
maintenance agent; however, 2019 EULAR/ERA-EDTA23
and low-dose steroid achieved complete renal remission
guideline recommends MMF maintenance specifically
in 32.6% patients with 23.7 mg twice a day dosage regimen
for patients induced with MMF. Duration of maintenance
as compared to 19.3% in placebo group. 34 The phase
immunosuppression is recommended to be at least 3
III AURA-LV trial evaluating the efficacy and safety of
years in patients who achieve complete remission at
voclosporin in patients with active LN is ongoing, with
the end of induction therapy.23 In those with high risk of
interim analysis showing promising results.
deterioration of renal function such as failure to achieve
Multitargeted therapy: Combination of tacrolimus complete remission after induction therapy, frequent
(4 mg/day) with MMF (1 g/day) and steroids (intravenous renal flares, and high disease activity index, prolonged
methylprednisolone 500 mg for 3 days followed by immunosuppression is given usually.

MU-148.indd 961 29-01-2021 15:01:51


962 SECTION 11 Nephrology

Newer Therapies „„ Anti-BAFF following anti-CD20 for LN did not improve


clinical outcome at week 24;
Biological Agents „„ Anti-BAFF delayed blood B-cell reconstitution
Despite the use of conventional immunosuppressive following B-cell depletion; and
agents, response rates of LN are only 60–70% at the end of „„ Anti-BAFF following anti-CD20 was not associated
induction therapy. High rates of relapses after achieving with hypogammaglobulinemia or an increase in
remission and reduction of drug toxicity have been unmet serious infections. Further analyses at 48 weeks and
needs in the management of LN. Hence, there is need for beyond are awaited.
newer drugs which are safer and more effective. Biological
agents with specific targets in the immune system have Belimumab: This humanized monoclonal antibody
been used as alternative options for conventional drugs for binds to BAFF leading to decreased activation of B-cells.
achieving higher rates of remission, managing refractory Belimumab is the only biological agent approved for
disease and as steroid sparing agents. treatment of active SLE. The BLISS-LN (Efficacy and safety
of Belimumab in patients with Active Lupus Nephritis)
Rituximab: This is a chimeric monoclonal antibody against study was recently announced to have favorable findings;
CD20 on the B-cell surface. Several nonrandomized although, the results are yet to be published (https://
studies have shown efficacy of rituximab in managing www.clinicaltrialsarena.com/news/gsk-benlysta-sle-
patients with LN. LUNAR (Lupus nephritis assessment phaseiii-data/). It met its primary endpoint demonstrating
with Rituximab) trial39 was a placebo-controlled phase that a statistically significant greater number of patients
3 RCT. Those in rituximab arm received 1,000 mg of achieved Primary Efficacy Renal Response over 2 years
rituximab 2 weeks apart at the start and it was repeated when treated with belimumab plus standard therapy
after 6 months. All the patients received three pulses of compared to placebo plus standard therapy in adults
intravenous methylprednisolone followed by oral steroids with active LN (43% vs. 32%, odds ratio (95% CI) 1.55
along with MMF at a dose of 2–3 g/day. Renal response (1.04, 2.32), p=0.0311). Based on the positive post hoc
at the end of 52 weeks was not different between both analyses from trials in non LN patients and the benefits of
the arms (57% vs. 46% in rituximab vs. placebo arms). belimumab in observational studies involving LN, it has
However, rituximab was shown to be effective in refractory been suggested to be used as add-on in treating extrarenal
LN. 40 Rituximab has also been proven to be effective flares/disease activity in LN patients by the 2019 EULAR/
as a steroid sparing agent in a pilot trial of 50 patients ERA-EDTA guideline. 23 It can also be used as steroid
of active LN who were given two doses of 1 gm/dose sparing agent.
rituximab 2 weeks apart along with MMF and single dose
of intravenous methyl prednisolone (500 mg). About 86% Obinutuzumab: NOBILITY trial 43 —In patients with
of patients achieved response (52% complete response proliferative LN, Obinutuzumab (intravenous) in addition
and 34% partial response) at the end of 52 weeks.41 Based to MMF (2–2.5 g/day) and steroids (0.5 mg/kg/day of
on the encouraging results, an open label, multicentric prednisone followed by taper) versus placebo in addition
RCT (RITUXILUP) was being carried out using rituximab to MMF and steroids at same doses as intervention arm—
for steroid sparing effect. Rituximab or any B-cell depleting overall renal response was higher than placebo arm at
agent causes B-cell activating factor/B lymphocyte 52 weeks. Obinutuzumab was as safe when compared
stimulating agent (BAFF/BLyS) to increase, which in to placebo. From this trial, it becomes apparent that
turn can lead to early repopulation of remnant B-cells. more effective B-cell depletion results in better clinical
So, combination of agents to block both would have the outcome, unlike the LUNAR trial. More stringent CD19
maximum effect. To test this hypothesis, CALIBRATE B-cell depletion could be achieved with Obinutuzumab
(ClinicalTrials.gov Identifier: NCT02260934) trial was as compared to Rituximab as Obinutuzumab is a type
conducted where in patients with resistant active LN were II monoclonal antibody known to enhance antibody
treated with CYC and rituximab followed by belimumab. dependent cytotoxicity of B-cells and therefore, better and
An interim analysis42 of data from CALIBRATE shows: deeper response.

MU-148.indd 962 29-01-2021 15:01:51


Lupus Nephritis—Current Understanding and Management CHAPTER 148 963

Other biological agents that were tested or being tested in patients with systemic lupus erythematosus. Arthritis Rheum.
in clinical trials for Lupus Nephritis are— 2005;52(4):1129-37.
5. Mok CC, Kwok RCL, Yip PSF, et al. Effect of renal disease on the
Trials which failed or were terminated: standardized mortality ratio and life expectancy of patients
„„ Abatacept (CTLA4-Ig)—(ACCESS trial)—failed to meet with systemic lupus erythematosus. Arthritis Rheum. 2013;65(8):
the endpoint 2154-60.
„„ Bortezomib (plasma cell proteasome inhibitor)—trial
6. Costenbader KH, Desai A, Alarcón GS, et al. Trends in the incidence,
demographics, and outcomes of end-stage renal disease due
terminated to lupus nephritis in the US from 1995 to 2006. Arthritis Rheum.
„„ Rituximab (anti-CD20)—failed to meet endpoint
2011;63(6):1681-8.
„„ Ocrelizumab (anti-CD20)—failed to meet endpoint 7. Ortega LM, Schultz DR, Lenz O, et al. Review: lupus nephritis:
„„ Sirukumab (anti-IL6)—failed to meet endpoint pathologic features, epidemiology and a guide to therapeutic
„„ Tabalumab (anti-BLyS)—failed to meet endpoint decisions. Lupus. 2010;19(5):557-74.
8. Delfino J, dos Santos TAFG, Skare TL, et al. Comparison of lupus
Trials which showed encouraging results or are ongoing: patients with early and late onset nephritis: a study in 71 patients
„„ Laquinimod—encouraging phase 2 trial results from a single referral center. Adv Rheumatol. 2020;60(1):5.
„„ Obinutuzumab: humanized anti-CD20 antibody: 9. Rathi M, Gupta KL, Joshi K, et al. Histopathological indicators of
disease outcome in class IV lupus nephritis: a revisit of various
NOBILITY—phase 2 clinical trial—encouraging
indices. Rheumatol Int. 2015;35(9):1511-7.
results—show better overall renal response than 10. Pattanashetti N, Anakutti H, Ramachandran R, et al. Effect of
placebo arm thrombotic microangiopathy on clinical outcomes in Indian
„„ Belimumab—anti-BLyS/BAFF antibody: BLISS-LN— patients with lupus nephritis. Kidney Int Rep. 2017;2(5):844-9.
phase 3 clinical trial-met the primary endpoint 11. Gupta KL. Lupus nephritis in children. Indian J Pediatr.
„„ Rituximab and cyclophosphamide followed by 1999;66(2):215-23.
12. Nasr SH, D’Agati VD, Park HR, et al. Necrotizing and crescentic lupus
belimumab: CALIBRATE—phase 2 clinical trial
nephritis with antineutrophil cytoplasmic antibody seropositivity.
„„ Anifrolumab—anti-IFN alpha-R antibody: phase 3
Clin J Am Soc Nephrol. 2008;3(3):682-90.
clinical trial—TULIP-LN1-ongoing 13. K idne y Disease: I mproving Global Outcomes (KDIGO)
„„ Voclosporin—calcineurin inhibitor: AURA-LV-phase Glomerulonephritis Work Group. KDIGO Clinical Practice Guideline
2 trial—met its primary endpoint; AURORA phase for Glomerulonephritis. Kidney Int Suppl. 2012;2(2):139-274.
3 clinical trial (https://www.clinicaltrialsarena. 14. Hahn BH, McMahon MA, Wilkinson A, et al. American College
of Rheumatology guidelines for screening, treatment, and
com/news/aurora-trial-lupus-kidney-disease-drug-
management of lupus nephritis. Arthritis Care Res (Hoboken).
therapy/): met the primary endpoint 2012;64(6):797-808.
15. Bertsias GK, Tektonidou M, Amoura Z, et al. Joint European
Conclusion League Against Rheumatism and European Renal Association–
European Dialysis and Transplant Association (EULAR/ERA-EDTA)
LN is characterized by flares and about 30–40% of patients do recommendations for the management of adult and paediatric
not respond to conventional induction therapy. Protocol repeat lupus nephritis. Ann Rheum Dis. 2012;71(11):1771-82.
biopsy is a promising tool to monitor LN. Less toxic and more 16. Weening J, D’Agati VD, Schwartz MM, et al. International Society
effective agents are the unmet needs in the management of LN. of Nephrology/Renal Pathology Society Working Group on the
Classification of Lupus Nephritis. J Am Soc Nephrol. 2004;15(2):
241-50.
References 17. Gupta KL, Sharma BK. Lupus nephritis—clinicopathological
1. Seligman VA, Lum RF, Olson JL, et al. Demographic differences in correlations and management. In: Bansal BC (Ed). Postgraduate
the development of lupus nephritis: a retrospective analysis. Am J Medicine of API; 1997. pp. 366-77.
Med. 2002;112(9):726-9. 18. Bajema I, Wilhelmus S, Alpers C, et al. Revision of the International
2. Al Arfaj AS, Khalil N. Clinical and immunological manifestations in Society of Nephrology/Renal Pathology Society classification for
624 SLE patients in Saudi Arabia. Lupus. 2009;18:465-73. lupus nephritis: clarification of definitions, and modified National
3. Bastian HM, Roseman JM, McGwin G, et al. Systemic lupus Institutes of Health activity and chronicity indices. Kidney Int.
erythematosus in three ethnic groups. XII. Risk factors for lupus 2018;93(4):789-96.
nephritis after diagnosis. Lupus. 2002;11(3):152-60. 19. Malvar A, Pirruccio P, Alberton V, et al. Histologic versus clinical
4. Linnik MD, Hu JZ, Heilbrunn KR, et al. Relationship between anti- remission in proliferative lupus nephritis. Nephrol Dial Transplant.
double-stranded DNA antibodies and exacerbation of renal disease 2017;32(8):1338-44.

MU-148.indd 963 29-01-2021 15:01:51


964 SECTION 11 Nephrology

20. De Rosa M, Azzato F, Tobili JE, et al. A prospective observational maintenance treatment regimens for proliferative lupus nephritis
cohort study highlights kidney biopsy findings of lupus based either on cyclophosphamide or on cyclosporine A. Lupus.
nephritis patients in remission who flare following withdrawal of 2014;23(1):69-74.
maintenance therapy. Kidney Int. 2018;94(4):788-94. 33. Mok CC, Yong KY, Yim CW, et al. Tacrolimus versus mycophenolate
21. Fanouriakis A, Kostopoulou M, Alunno A, et al. 2019 update of the mofetil for induction therapy of lupus nephritis: a randomized
EULAR recommendations for the management of systemic lupus controlled trial and long-term follow up. Ann Rheum Dis.
erythematosus. Ann Rheum Dis. 2019;78(6):736-45. 2016;75(1):30-6.
22. Gupta KL, Bharati J, Anakutti H, et al. Contribution of clinically 34. Rovin BH, Solomons N, Pendergraft WF 3rd, et al. A randomized,
indicated repeat renal biopsy in Indian patients with lupus controlled double-blind study comparing the efficacy and safety of
nephritis. Indian J Nephrol. 2020. dose-ranging voclosporin with placebo in achieving remission in
23. Fanouriakis A, Kostopoulou M, Cheema K, et al. 2019 Update of the patients with active lupus nephritis. Kidney Int. 2019;95(1):219-31.
Joint European League Against Rheumatism and European Renal 35. Bao H, Liu ZH, Xie HL, et al. Successful treatment of class V+IV
Association-European Dialysis and Transplant Association (EULAR/ lupus nephritis with multitarget therapy. J Am Soc Nephrol.
ERA-EDTA) recommendations for the management of lupus 2008;19(10):2001-10.
nephritis. Ann Rheum Dis. 2020. 36. Contreras G, Pardo V, Leclercq B, et al. Sequential therapies for
24. Boumpas D, Austin H, Vaughn E, et al. Controlled trial proliferative lupus nephritis. N Engl J Med. 2004;350(10):971-80.
of pulse methylprednisolone versus t wo regimens of 37. Houssiau FA, D’Cruz D, Sangle S, et al. Azathioprine versus
pulse cyclophosphamide in severe lupus nephritis. Lancet. mycophenolate mofetil for long-term immunosuppression in
1992;340(8822):741-5. lupus nephritis: results from the MAINTAIN Nephritis Trial. Ann
25. Gupta KL. Treatment of Lupus Nephritis. J Int Med Sci Acad. Rheum Dis. 2010;69(12):2083-9.
1998;11:19-25. 38. Dooley MA, Jayne D, Ginzler EM, et al. Mycophenolate versus
26. Houssiau FA, Vasconcelos C, D’Cruz D, et al. Immunosuppressive azathioprine as maintenance therapy for lupus nephritis. N Engl J
therapy in lupus nephritis: the Euro-Lupus Nephritis Trial, a Med. 2011;365(20):1886-95.
randomized trial of low-dose versus high-dose intravenous 39. Rovin BH, Furie R, Latinis K, et al. Efficacy and safety of rituximab in
cyclophosphamide. Arthritis Rheum. 2002;46(8):2121-31. patients with active proliferative lupus nephritis: the lupus nephritis
27. Rathi M, Goyal A, Jaryal A, et al. Comparison of low-dose assessment with rituximab study. Arthritis Rheum. 2012;64(4):
intravenous cyclophosphamide with oral mycophenolate mofetil 1215-26.
in the treatment of lupus nephritis: Kidney Int. 2016;89(1):235-42. 40. Weidenbusch M, Römmele C, Schröttle A, et al. Beyond the LUNAR
28. Chan TM, Li FK, Tang CS, et al. Efficacy of mycophenolate mofetil trial. Efficacy of rituximab in refractory lupus nephritis. Nephrol Dial
in patients with diffuse proliferative lupus nephritis. Hong Transplant. 2013;28(1):106-11.
Kong-Guangzhou Nephrology Study Group. N Engl J Med. 41. Condon MB, Ashby D, Pepper RJ, et al. Prospective observational
2000;343(16):1156-62. single-centre cohort study to evaluate the effectiveness of treating
29. Ginzler EM, Dooley MA, Aranow C, et al. Mycophenolate or lupus nephritis with rituximab and mycophenolate mofetil but no
intravenous cyclophosphamide for lupus nephritis. N Engl J Med. oral steroids. Ann Rheum Dis. 2013;72(8):1280-6.
2005;353(21):2219-28. 42. Aranow C, Dall’Era M, Byron M, et al. FRI0305 Phase 2 trial of
30. Appel GB, Contreras G, Dooley MA, et al. Mycophenolate mofetil induction therapy with anti-cd20 (RITUXIMAB) followed by
versus cyclophosphamide for induction treatment of lupus maintenance therapy with anti-baff (BELIMUMAB) in patients with
nephritis. J Am Soc Nephrol. 2009;20(5):1103-12. active lupus nephritis. Ann Rheum Dis. 2018;77:690.
31. Zavada J, Pesickova S, Rysava R, et al. Cyclosporine A or intravenous 43. Furie R, Aroca G, Alvarez A, et al. A Phase II Randomized, Double-
cyclophosphamide for lupus nephritis: the Cyclofa-Lune study. Blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety
Lupus. 2010;19(11):1281-9. of Obinutuzumab or Placebo in Combination with Mycophenolate
32. Závada J, Sinikka Pesicková S, Rysavá R, et al. Extended follow-up of Mofetil in Patients with Active Class III or IV. Lupus Nephritis
the CYCLOFA-LUNE trial comparing two sequential induction and [abstract no. 939]. Arthritis Rheumatol. 2019;71(10).

MU-148.indd 964 29-01-2021 15:01:51


CHAPTER

149 Diagnosis and Management of


Nephrotic Syndrome
Deepti Sharma, Sandeep S, Gopikrishnan R

Abstract 
Nephrotic syndrome is an important presentation of glomerular disease featured by heavy proteinuria, hypoalbuminemia,
and edema. The etiology of nephrotic syndrome is more complex and heterogenous in adults compared to children. The
renal function is often normal but can progress to chronic renal failure which depends on the duration and amount of
proteinuria. Acute renal failure is rare and mostly caused by a precipitating event. Early recognition is possible through
urine protein estimation, but a renal biopsy is often recommended. Current treatment recommendations are mostly based
on randomized control trials (RCTs) in children, and there are only small RCTs and a few case series studies in adults.
Diuretics are required in large doses due to the low-serum albumin levels. Immunosuppressive treatment is often used with
little evidence. Prophylactic treatment to prevent infection or thrombosis is not recommended routinely unless indicated.

Introduction may modulate the passage of macromolecules across


the glomerular capillary wall.2 The podocyte appears
Nephrotic syndrome is a glomerular disease, defined
to be the major target of injury in idiopathic nephrotic
as a pentad of proteinuria more than 3.5 g/day,
syndrome. Adult-onset primary MN and focal segmental
hypoalbuminemia less than 3.5 g/dL, edema, hyper­
glomerulosclerosis (FSGS) may be due to autoantibodies
cholesterolemia, and lipiduria.1 Patients present with
to podocyte or circulating factor that affect the podocyte.
edema, typically periorbital or at dependant sites or as a
complication of nephrotic state such as thromboembolism.
Hypoalbuminemia
Etiology Most of albumin loss is due to urinary excretion leading
to reduction in total protein and serum albumin with
Ne p h ro t i c s y n d ro m e may b e d u e t o a p r i ma r y
increased α2-globulin and reduced γ-globulin fraction.
glomerulopathy, secondary to systemic diseases or as a
Hepatic albumin synthesis is increased in response to the
result of genetic mutations. The primary disorders are
albumin loss mediated by an increase in gene expression
minimal change disease, focal segmental glomerulo­
and the release of an unidentified circulating factor.3
sclerosis, and membranous nephropathy (MN). Secondary
causes are elaborated in Table 1.
Edema
Pathophysiology and Clinical Manifestation Two major mechanisms have been thought to be
responsible for the development of edema. One is
Proteinuria “underfill hypothesis” where secondary sodium retention
Protein loss is due to glomerular proteinuria. Electrical due to low plasma oncotic pressure causes shifting of
potential differences generated by transglomerular flow fluid into the interstitium leading to underfilling of the

MU-149.indd 965 29-01-2021 15:01:46


966 SECTION 11 Nephrology

TABLE 1 Secondary causes of nephrotic syndrome

MCD FSGS MN Others


Drugs Virus associated Autoimmune diseases zz Diabetes
zz NSAID zz HIV-1 zz Class V lupus nephritis zz Plasma cell disorder
zz Interferon-a zz Parvovirus B19 zz Rheumatoid arthritis zz Paraprotein deposit disease

zz Lithium zz Simian virus 40 (SV40) CMV zz Sarcoidosis zz AA amyloidosis

zz Gold Drug induced zz Sjogren zz Pre-eclampsia

Allergy zz Heroin Infections


zz Pollens zz Interferon zz Hepatitis B

zz House dust zz Lithium zz Hepatitis C

zz Insect stings zz Pamidronate zz HIV

zz Immunizations zz Sirolimus zz Syphilis

zz Poison oak zz Anabolic steroids zz Malaria

Malignancy zz Tyrosine kinase inhibitors zz Schistosomiasis

zz Hodgkin disease Hyperfiltration injury Malignancy


zz Mycosis fungoides zz Renal agenesis zz Solid tumors (colon, stomach, lung, prostate)

zz CLL

vasculature and activation of the renin–angiotensin– Enhanced hepatic synthesis of lipoproteins containing
aldosterone system (RAAS) causing further sodium apolipoprotein B and cholesterol and diminished
and water retention. Second mechanism is “Overfill catabolism caused by inhibition of lipoprotein lipase
hypothesis” where primary sodium retention occurs due due to elevated levels of sialylated angiopoietin like-4
abnormalities in tubular function like increased activity (especially triglycerides) is thought to be the mechanism
of the Na-K-ATPase pump, epithelial sodium channel behind this. Lipoprotein glomerulopathy is a rare form of
(ENaC),4 and relative resistance to ANP and urodilatin due glomerular disease that is associated with dyslipidemia
to increased phosphodiesterase activity. and lipoprotein deposits in the glomeruli.

Hypercoagulability AKI and CKD in Nephrotic Syndrome


Patients with the nephrotic syndrome have an increased Acute kidney injury (AKI) is less common in nephrotic
incidence of arterial and venous thrombosis, particularly syndrome. Most cases of nephrotic syndrome have a risk
deep vein thrombosis (DVT) and renal vein thrombosis. of progression to chronic kidney disease (CKD) except
Thrombotic state occurs mainly due to urinary loss of minimal change disease (MCD). Greatest risk factor is the
antithrombin III, plasminogen, protein C and S. Increased degree of proteinuria (>5 g/day). Table 2 shows various
platelet activation, thrombocytosis, hyperfibrinogenemia, causes for AKI in nephrotic syndrome.
and inhibition of plasminogen activation also contributes.
Renal vein is a common site of thrombosis in nephrotic Diagnosis
syndrome, possibly due to stimulation of thrombin
production in the glomerular efferent vessels as a result of Urine Analysis
glomerular injury. „„ Twenty-four hour urine protein estimation: More than
3.5 g/day is considered nephrotic-range proteinuria.
Hyperlipidemia „„ Protein-to-creatinine ratio: It is only an alternative
Hyperlipidemia is triggered by the reduction in plasma to the 24-hour urine protein estimation. The total
oncotic pressure. The severity of the hyperlipidemia protein-to-creatinine ratio (mg/mmol) on early
is inversely related to the fall in oncotic pressure and morning sample of urine is calculated. This ratio
resolves with the remission of nephrotic syndrome. correlates closely with daily protein excretion in

MU-149.indd 966 29-01-2021 15:01:46


Diagnosis and Management of Nephrotic Syndrome CHAPTER 149 967

TABLE 2 Various causes for AKI in nephrotic syndrome TABLE 3 Serologic studies (marker and diagnosis)

zz AIN secondary to drugs (including diuretics) Serological marker Diagnosis


zz ATN due to volume depletion or sepsis Anti-PLA2R Primary MN (70–80%)
zz Hemodynamic response to NSAIDS, ACEI, ARBs Anti-THSD7A Primary MN (5%)
zz Nephrosarca ANA, anti-dsDNA, anti-Ro/La Lupus nephritis (MN), Sjögren (MN)
zz Pre-renal failure due to volume depletion HBSAg, Anti-HCV, HIV, Secondary MN
zz Renal vein thrombosis VDRL/FTA-Ab

zz Transformation of underlying glomerular disease PSA Ca prostates (MN)


CEA Ca colon (MN)
HIV antibody FSGS >> MN
g/1.73 m 2 of body surface area. A value more than Low C3.C4 levels MPGN Type 1
300 mg/mmol is considered nephrotic range. But this Plasma-free light chains Multiple myeloma (MN)
method has limitations and 24-hour urine protein
RA factor, Anti-CCP MN
estimation is more preferred.
RBS, HbA1c Diabetic nephropathy

Serologic Studies
A number of serologic studies often are obtained in the Histological Patterns
evaluation of patients with the nephrotic syndrome
See Table 4.
depending upon clinical setting (Table 3).

Renal Biopsy Management


In adult nephrotic syndrome, biopsy is indicated when Management of nephrotic syndrome consists of general
the etiology of persistent nephrotic-range proteinuria is in supportive measures, disease-specific therapy for the
doubt in order to determine management decisions. underlying cause in secondary nephrotic syndrome,
and immunosuppression in case of primary nephrotic
Indication for biopsy:
syndrome.
„„ Adult with nephrotic syndrome

„„ Children with:

—— age <1 year or >12 years


General Supportive Treatment
—— Gross hematuria It includes measures to reduce proteinuria, edema,
—— Marked hypertension control blood pressure, and to address other metabolic
—— Renal failure without severe hypovolemia consequences of nephrotic syndrome.
—— Low C3 level

Treating Edema
Imaging All patients with nephrotic edema are initially treated with
„„ Ultrasound: It must be done routinely before planning diuretics and dietary sodium and water restriction. It is
renal biopsy to rule out small kidneys or kidneys with recommended to restrict salt intake to less than 3 g per/
severe cortical thinning, as they indicate chronic day and water intake less than 1.5 liter/day.5 Most patients
irreversible disease. These findings can limit renal respond well to loop diuretics. They are highly protein-
biopsy and aggressive immunosuppressive therapy. bound but this bonding is reduced with hypoalbuminemia
„„ 2D Echocardiography: When infective endocarditis is resulting in a slower rate of delivery to the kidneys. Loop
suspected. of Henle may be relatively resistant to loop diuretics. Gut
„„ Renal Doppler: Indicated in patients with flank pain, edema also limits the oral absorption of diuretics. Thus,
hematuria, and rapid worsening of renal function to the effective diuretic dose is usually higher in patients
rule out renal vein thrombosis. with nephrotic syndrome. Intravenous infusion can also

MU-149.indd 967 29-01-2021 15:01:47


968 SECTION 11 Nephrology

TABLE 4 Histopathological patterns

MCD FGGS MN
LM Normal Sclerosis in parts (segmental) of Homogenus thickening of capillary wall (H&E)
some (focal) glomeruli
Spikes in GBM (Silver stain)
EM Diffuse foot process Diffuse foot process effacement Sub-epithelial deposit on surface of GBM
effacement
Wrinkling and retraction of GBM Foot process effacement
Hyaline accumulation
IF No deposits IgM, C3, C1q (Classical) Diffuse global finely granular deposits of IgG along outer
surface of all capillary walls
Pathology Circulating Podocyte injury Immune complex and complement mediated podocyte injury
permeability factor (Secondary MN)
Abnormal regulation Nephron loss Autoantibody mediated podocyte injury
of T-cell

be tried as it increases the diuretic efficacy. Furosemide of urinary protein loss to dietary protein intake. As a last
40 mg orally twice a day or bumetanide 1 mg twice daily resort, nephrectomy and renal artery embolization can be
is a reasonable starting dose and may be increased up done if protein loss cannot be managed medically.
to a dose of 600 mg/day according to need. Patients who
do not respond adequately to loop diuretic require the Hypertension
addition of a thiazide diuretic. Alternatively, amiloride 6 Hypertension in nephrotic syndrome requires strict
or acetazolamide7 can be combined with loop diuretics control to delay the progression of renal disease and
in patients with refractory edema. Prior administration of prevent cardiovascular complications. Initially life style
albumin increases the efficacy of diuretics.8 modification can be adopted. High dose diuretics and
dietary salt restriction are necessary. The KDIGO guideline
Proteinuria
recommends a target BP <130/80 mm Hg,9 but recent
Progressive loss of renal function can be reduced if SPRINT trial suggests a target of 120/80 mm Hg. ACEIs and
proteinuria can be reduced below 0.5 g/day. It can ARBs are the first choice. DHP-CCBs are not preferred as
be controlled by either blocking efferent arteriole they increase GFR causing worsening of proteinuria and
vasoconstriction or decreasing the preglomerular pressure also add to peripheral edema.
(antihypertensives). Angiotensin converting enzyme
inhibitors (ACEIs) and angiotensin-receptor blockers
Hyperlipidemia
(ARBs) are the agents of choice. They reduce proteinuria
independent of blood pressure and can be safely used in Control of hyperlipidemia is important in preventing
normotensive patients. If proteinuria persists in spite of cardiovascular complications. Dietary modification alone
ACEI/ARB, aldosterone antagonists can be added. They has only a moderate effect on hyperlipidemia. Statin alone
can raise the serum creatinine by 30–40% as glomerular is preferred in patients more than 50 years of age with
filtration rate (GFR) is reduced, but should not be stopped early stage CKD and in late stages, addition of ezetimibe is
unless serum creatinine levels are continuously increasing. recommended. In young adults, consider statin if having
significant comorbidity.
Hypoproteinemia
Adequate dietary protein intake of 0.8–1 g/kg/day with Prophylaxis for Thrombosis
high carbohydrate to maximize the use of that protein The decision to prescribe prophylactic anticoagulation
is recommended. In heavy proteinuria, add the amount must be balanced against the risk of bleeding. If the

MU-149.indd 968 29-01-2021 15:01:47


Diagnosis and Management of Nephrotic Syndrome CHAPTER 149 969

bleeding risk is unclear, ATRIA Risk score or HAS-BLED Steroid Dependent (Table 5),
risk can be used to take a decision. Prophylactic low-dose Frequently Relapsing and Resistant MCD
heparin is indicated in cases with high risk for thrombosis,
These patients should be started on second-line therapy.
like in pregnancy or an immobilized patient, with a
This includes:
serum albumin less than 2.5 g/dL. If albumin is less than
„„ Cyclophosphamide: 2–2.5 mg/kg/day orally for 12
2 g/dL, full anticoagulation with low-molecular-weight
weeks is given. Due to risk of infertility, banking of
heparin (LMWH) or warfarin should be considered. 10
sperm and ova is to be considered before starting
Patients with MN are at high risk for thrombosis, and if the
therapy.
albumin level is very low and even if the risk of bleeding
„„ Calcineurin inhibitors:
is intermediate, they may benefit from anticoagulation.
—— Cyclosporine: 4–6 mg/kg/day for at least 12
Warfarin is preferred over LMWH because of low levels of
months is given. Nephrologists prefer calcineurin
antithrombin III impeding efficacy.
inhibitors over cyclophosphamide in young adults
to prevent infertility.
Management and Prevention of Infection —— Tacrolimus: A dose of 2–4 mg twice daily, adjusted
R i s k o f i n f e c t i o n i s e x a c e r b a t e d b y n e p h ro t i c to maintain a target level of 5–10 ng/mL is given. It
immunodeficiency caused by T-cell transformation may be considered as a first line or second line in
dysfunction and urinary loss of immunoglobulins patient with contraindication for or intolerance to
including IgG and alternate pathway complement factors. high dose corticosteroid.12
Patients may develop recurrent respiratory and urinary „„ MMF: 750–1,000 mg twice daily for 6 months may be
tract infections, peritonitis, and sepsis; particularly with used in cyclosporine dependant cases but did not
encapsulated bacteria. Antibiotic with pneumococcal appear as effective as cyclosporine in studies.
coverage is the mainstay in infections and prophlylactic „„ Rituximab: It is a B-Lymphocyte depleting agent (anti-
pneumococcal vaccination is recommended. In case of CD20) and is used when disease is unresponsive to all
repeated infection, IgG level should be estimated and if
low IVIgG should be given to maintain a level above 600
Terminologies regarding treatment outcomes of
mg/dL.11 TABLE 5
nephrotic syndrome

Treating MCD Complete Reduction of proteinuria to ≤0.20 g/day and serum


remission albumin >3.5 g/dL
Initial Treatment Partial Reduction of proteinuria to between 0.21 g/day and
remission 3.4 g/day ± decrease in proteinuria of ≥50% from
Low dose oral prednisolone (1 mg/kg/day up to maximum baseline
80 mg/day) should be started according to KDIGO No remission Failure to reduce urine protein excretion by 50%
guideline and recommends it to be continued up to 6 from baseline or persistent excretion uPCR >2000
months. Initial dose should be maintained for a minimum mg/g (>200 mg/mmol)
of 4 weeks if full remission achieved or 16 weeks if Steroid Persistence of proteinuria despite prednisolone
full remission not achieved. Once remission has been resistance therapy, 1 mg/kg/day for 16 weeks
achieved, dose should be tapered at least within 4 weeks Relapse Proteinuria ≥3.5 g/day occurring after complete
of response. Induction with methylprednisolone pulse remission obtained for >1 month
therapy may lead to rapid response and fewer relapses. Infrequent One relapse within 6 month of initial response, or
relapse one to three relapses in any 12-month period
If the patient has not achieved remission even after
12–16 weeks of treatment, consider repeating biopsy, Frequent Two or more relapses within 6 month of initial
relapse response, or four or more relapses in any 12-month
as it can be FSGS which might have been missed due to period
limited number of nephrons in earlier specimen. If MCD Steroid Two consecutive relapses during corticosteroid
presents with non-nephrotic range proteinuria, it can dependence therapy, or within 14 days of completing
be managed with ACEIs/ARBs and there is no need for corticosteroid therapy
starting steroids. Reference: Comprehensive Clinical nephrology - 6th edition

MU-149.indd 969 29-01-2021 15:01:47


970 SECTION 11 Nephrology

other treatments, but RCT have not been performed. Membranous Nephropathy
Non RCT studies have shown benefit and reduction It is very difficult to treat MN due to its chronic nature, the
in relapse in corticosteroid dependant and frequent tendency for spontaneous remission and relapse and the
relapse cases. 13 A dose of 375 mg/m 2 weekly for 4 variability in clinical severity. Specific immunosuppressive
weeks. therapy should not be considered unless the patient
has a persistent nephrotic-range proteinuria (>4 g/day)
FSGS and it has not declined more than 50% from baseline,
It is very difficult to distinguish between primary and over a minimum period of 6 months, despite maximum
secondary FSGS and most of the times they are treated antihypertensive and antiproteinuric therapy.16 Treatment
as primary FSGS. All patients will benefit from good of secondary MN focuses on cessation of the offending
drug or effective treatment of the underlying disease.
supportive treatment and immunosuppression.

Initial Treatment Immunosuppression


It consist of 6 months of continuous corticosteroid (1 mg/ „„ Corticosteroids: RCTs showed no significant long-term
kg/day), later tapered over 4–8 weeks. benefit on proteinuria or rate of disease progression.
Therefore, use of oral corticosteroids as a single agent
Steroid Resistant Cases is not recommended.
„„ Cytotoxic agents with corticosteroids (PONTICELLI
„„ Cyclosporine: Low dose of 3–6 mg/kg/day is given for
REGIMEN): Benefitted in patients at moderate risk
2–6 months. Recent studies show that combination
for progression. It starts with methylprednisolone
of cyclosporine and corticosteroids has a higher
pulses 1 g IV for 3 days at the beginning of months
remission rate.14
1, 3, and 5 followed by oral methylprednisolone 0.4
„„ Tacrolimus: Used in cases resistant or intolerant to
mg/kg/day for 27 days, and each cycle followed by
cyclosporine.
1 month of cytotoxic agent (cyclophosphamide or
„„ MMF and Dexamethasone: As per KDIGO guidelines,
chlorambucil).17 Cyclophosphamide-based regimen is
they can be given in corticosteroid resistant cases that
preferred because of a better safety profile.
are intolerant to calcineurin inhibitors. „„ Calcineurin inhibitors : Given in patients with
„„ Rituximab: Used when all other treatments fail and corticosteroid-resistant MN but has a higher relapse
this drug has shown promising results in steroid- rate.
dependent cases. —— Cyclosporine: It is used along with low-dose
„„ Abatacept (CTL A-4-Ig): An inhibitor of T-cell corticosteroids. It may reduce proteinuria not only
costimulator y molecule B7-1(CD-80) showed through its immunosuppressive effects but also by
remission in some patients and further studies are in direct effects on the podocyte.
progress.15 —— Tacrolimus: It can be used as monotherapy, but

relapse rate was found to be high.


Secondary FSGS „„ MMF: Along with steroids, it showed good initial
response, but relapse rate was high.18
Patients with primary FSGS typically present with an acute
„„ Rituximab: Used in steroid resistant cases. Decline in
onset nephrotic syndrome, whereas slowly increasing
anti-PLA2R antibody and proteinuria was achieved,
asymptomatic proteinuria and renal insufficiency over
subsequently resulting in a partial or complete
a period of time without profound hypoalbuminemia
remission.
or edema are characteristic of secondary FSGS. Unlike
primary FSGS, these patients are less responsive to
immunosuppression and the mainstay of the treatment Plasma Exchange
is to correct the primary cause and provide supportive It may be useful in resistant MN, severe nephrotic
treatment. syndrome, and high anti-PLA2R antibody titer cases. A

MU-149.indd 970 29-01-2021 15:01:47


Diagnosis and Management of Nephrotic Syndrome CHAPTER 149 971

study in such type of patients, treated with a “rescue” 6. Hoorn EJ, Ellison DH. Diuretic resistance. Am J Kidney Dis.
regimen consisting of four plasma exchanges, 20 g of 2017;69(1):136-42.
7. Fallahzadeh MA, Dormanesh B, Fallahzadeh MK, et al. Acetazolamide
intravenous (IV) immunoglobulin, and 375 mg/m2 of and hydrochlorothiazide followed by furosemide versus furosemide
rituximab showed shorter remission time compared to and hydrochlorothiazide followed by furosemide for the treatment
previous regimens.19 of adults with nephrotic edema: a randomized trial. Am J Kidney
Dis. 2017;69(3):420-27.
8. Fliser D, Zurbrüggen I, Mutschler E, et al. Coadministration of
Adrenocorticotropic Hormone (ACTH) albumin and furosemide in patients with the nephrotic syndrome.
ACTH acts via melanocortin 1 receptor expressed on Kidney Int. 1999;55(2):629-34.
podocytes. Synthetic form of ACTH, 1–2 mg weekly 9. Kidney Disease: Improving Global Outcomes. Clinical practice
guideline for the evaluation and management of chronic kidney
intramuscularly for 1 year showed prolonged remission in
disease. Kidney Int. 2013;3(Suppl 1):1-150.
the majority of patients with MN.20 10. Lee T, Biddle AK, Lionaki S, et al. Personalized prophylactic
anticoagulation decision analysis in patients with membranous
Conclusion nephropathy. Kidney Int. 2014;85(6):1412-20.
11. Ogi M, Yokoyama H, Tomosugi N, et al. Risk factors for infection
Nephrotic syndrome includes a wide spectrum of primary and immunoglobulin replacement therapy in adult nephrotic
syndrome. Am J Kidney Dis. 1994;24(3):427-36.
glomerular disorders and secondar y diseases. Early
12. Li X, Liu Z, Wang L, et al. Tacrolimus monotherapy after intravenous
recognition through urine protein estimation and bringing methylprednisolone in adults with minimal change nephrotic
out the histological variant through renal biopsy aids in syndrome. J Am Soc Nephrol. 2017;28(4):1286-95.
planning treatment properly in order to prevent progression 13. Ruggenenti P, Ruggiero B, Cravedi P, et al. Rituximab in
into renal failure. At present, reducing proteinuria and steroiddependent or frequently relapsing idiopathic nephrotic
immunosuppression remains the mainstay in most of the syndrome. J Am Soc Nephrol. 2014;25(4):850-63.
cases and larger RCTs are lacking. Many new treatment targets 14. Laurin LP, Gasim AM, Poulton CJ, et al. treatment with glucocorticoids
or calcineurin inhibitors in primary FSGS. Clin J Am Soc Nephrol.
have been identified, which needs further studies. A proper
2016;11(3):386-94.
recommendation for management of nephrotic syndrome
15. Hogan J, Bomback AS, Mehta K, et al. Treatment of idiopathic focal
needs to be postulated. segmental glomerulosclerosis with adrenocorticotropic hormone
gel. Clin J Am Soc Nephrol. 2013;8(12):2072-81.
16. Johnson R, Feehally J, Floege J, et al. Comprehensive Clinical
References Nephrology, 6th edition. Philadelphia: Elsevier; 2019.
17. Ponticelli C, Zucchelli P, Passerini P, et al. A 10-year follow-up of a
1. Or th SR, Ritz E. The nephrotic syndrome. N Engl J Med.
randomized study with methylprednisolone and chlorambucil in
1998;338(17):1202-11.
membranous nephropathy. Kidney Int. 1995;48(5):1600-4.
2. Hausmann R, Kuppe C, Egger H, et al. Electrical forces determine
18. Dussol B, Morange S, Burtey S, et al. Mycophenolate mofetil
glomerular permeability. J Am Soc Nephrol. 2010;21(12):2053-8. monotherapy in membranous nephropathy: a 1-year randomized
3. Sun X, Martin V, Weiss RH, et al. Selective transcriptional controlled trial. Am J Kidney Dis. 2008;52(4):699-705.
augmentation of hepatic gene expression in the rat with Heymann 19. Müller-Deile J, Schiffer L, Hiss M, et al. A new rescue regimen
nephritis. Am J Physiol. 1993;264(3 Pt 2):F441-7. with plasma exchange and rituximab in high-risk membranous
4. Svenningsen P, Bistrup C, Friis UG, et al. Plasmin in nephrotic glomerulonephritis. Eur J Clin Invest. 2015;45(12):1260-9.
urine activates the epithelial sodium channel. J Am Soc Nephrol. 20. van de Logt AE, Beerenhout CH, Brink HS, et al. Synthetic
2009;20(2):299-310. AC TH in high risk patients with idiopathic membranous
5. Hull RP, Goldsmith DJ. Nephrotic syndrome in adults. BMJ. nephropathy: a prospective, open label cohort study. PLoS ONE.
2008;336(7654):1185-9. 2015;10(11):0142033.

MU-149.indd 971 29-01-2021 15:01:47


CHAPTER

150 Dietary Salt and Hypertension


PP Varma

Abstract 
Epidemiological data suggests that dietary salt is a modifiable risk factor for hypertension; especially in those with salt
consumption in the highest tertile. WHO recommends 5 gm of salt intake per day but we in India are consuming 9–12 gm
of salt per day. Besides hypertension, salt is also incriminated for CV mortality, stroke, and obesity. Hence, there is need
to restrict salt consumption to check the menace of hypertension. Here we bring the update on association of salt and
hypertension.

Introduction (NaCl) crystals are white in color and readily dissolve in


water. The molar mass is 58.5 g/mol.
Today over 30% of world’s adult population has
With agricultural revolution our food habits changed,
hypertension (HT). Over 60% of the cerebrovascular
and from 100% animal food, now plant based food formed
diseases and about half of ischemic heart diseases are
50% of our diet; however, salt intake remained less than
attributable to HT. Dietary salt is one of the modifiable
a gram per day. Earliest comment relating to dietary salt
culprits in causation of HT. Present commentary is about
and HT came from Chinese doctor, Huang Ti Nei Ching
data on dietary salt and its association with HT.
Su Wen, who wrote in The Yellow Emperor’s Classic of
Internal Medicine—“therefore, if large amounts of salt are
Historical Perspective taken, the pulse will stiffen or harden.” However, over the
During paleolithic times, hunter gathering man was years with civilization and changing food habits our dietary
dependent on raw meat and his dietary salt intake was salt intake has increased and this is being correlated with
less than 1 gm a day. Around 5,000 years back, Chinese increasing prevalence of HT. Today average Indian salt
learnt that food can be preserved with the help of salt intake is 9–12 gm/day against WHO recommendation
and Egyptian used this knowledge for preservation of of 5 gm.4 In this article we shall review the available data
mummies. Civilizations started developing along the salt showing correlation between salt intake and HT.
routes, which was considered a precious entity. In Libya,
salt was exchanged for equivalent amount of gold. French
salt tax “gabellee” probably incited French revolution. In Consumption and Its Source
India, Britishers did not allow Indians to make their own A study reported by George Institute from India found
salt and wanted them to buy expensive salt brought from that average Indian is taking 10.98 gm of salt per day.
England. This was the genesis of famous Dandi March, Salt consumption is higher in south Indians than north
an important event in the freedom struggle of India. Salt Indians. If one looks at the source of consumed salt, there

MU-150.indd 972 29-01-2021 15:01:42


Dietary Salt and Hypertension CHAPTER 150 973

are three sources: our natural food (cereals, vegetables, „„ The first double-blind controlled study of moderate
fruits, non-vegetables) contains less than a gm of salt salt restriction on human was from MacGregor et al.3
and makes 10% of daily intake. Another 2–3 gm (20–30%) They recruited 19 patients with mild to moderate HT
is added as cooking salt in our food and vegetables. (average supine BP of 156/98 mm Hg). Patients were
However, 70% of our consumed salt comes from processed advised not to take sodium laden food. After 2 weeks
food—snacks, bread and bread products, pizza, French of dietary salt restriction, patients entered an 8-week
fries, pasta, pickles, sauces, etc. It may be interesting to double-blind randomized crossover study. One group
know that each large slice of bread has 500 mg of salt in it. was given “Slow Sodium Ciba” (Ciba-10 mmol of
Twenty-four hour urinary sodium estimation is the best sodium per tablet) and other group received “Slow
marker of salt consumption. Sodium Placebo.” In fourth week, the mean supine BP
was 7.1 mm Hg (6.1%) lower in slow sodium placebo
Data on Salt Studies and Hypertension group (p<0.001). Urinary sodium excretion in the
„„ Epidemiologically: It is known that areas with low salt fourth week of slow sodium ciba was 162±9 mmol/24
consumption have lower incidence of HT. Solomon hours and that in the fourth week of slow sodium
islanders consume less than 2 gm of salt per day and placebo was 86±9 mmol/24 hours (p<0.001). They
have an HT prevalence of 1%. Among Yanomamo suggested that moderate sodium restriction should
Indians from Brazilian amazon, 84% have urinary become part of the management of essential HT and
sodium excretion of 1 mmol/24 hours. Their mean BP one should avoid sodium-laden foods.
is 96/60.0 mm Hg (78/37–128/86 mm Hg) and there „„ First large international study on salt and HT was
is no change in BP with age. In tribes with salt intake “Intersalt study.” 2 The study was conducted in 52
of 3 gm/day, prevalence of HT is 3%. Interestingly in centers from 22 countries; each with sample size
Newfoundland average salt intake is of 6.7–7.3 gm of 200 with a total of 10,079 participants. Four of
and HT prevalence is 15%. Its coastal area where salt these 52 centers, were tribal belts with very low
consumption is of 8.4–8.8 gm/day, HT prevalence is salt consumption. Data from 48 centers showed an
27%. This data suggests that with increasing salt intake insignificant trend but pooled data from 52 centers
prevalence of BP increases. showed that higher salt consumption was associated
„„ The effect of life style change and migration is with age-related rise in BP. This study prompted lot
exemplified by Yi community living in southwestern of debate whether to reduce salt consumption or not.
China. Those still living in mountainous environment A revisit of study inferred that 100 mmol of extra salt
and eating sodium-poor diet had yearly rise of systolic intake (70 mmol vs. 170 mmol) was associated with
and diastolic BP by 0.13 and 0.23 mm Hg, respectively. higher BP by 5–7/2–4 mm, stressing the need to reduce
In contrast, Yi community who had migrated in urban salt intake.
areas and consumed sodium-rich diet had a yearly „„ A community trial was done to study the effect of salt on
rise of 0.33 mm Hg for both systolic and diastolic BP, HT in Portugal. Two communities with 800 persons each
stressing the importance of life-style change on BP. were selected. Both had salt intake of 21 gm/day and HT
„„ Fi r s t g o o d a n i m a l s t u d y a b o u t s a l t a n d i t s prevalence of 30%. In one of the two communities with
correlation with HT came from chimpanzees that extensive health education, salt intake could be reduced
are phylogenetically similar to humans. Normally to 12 gm/day. By the end of second year BP dropped
chimpanzees eat diet rich in fruits/vegetables with in intervention community by 13/6 mm Hg. BP drop
dietary salt content of 0.5 gm/day. Study was done on was across all age groups and in both normotensives
26 chimpanzees.1 To their normal diet ~15 gm of salt and hypertensives alike. Those with greatest fall in salt
was added. BP started rising and after 84 weeks BP rose excretion had the largest fall in BP.
by 33/10 mm Hg. Again chimpanzees were reverted „„ Trials of hypertension prevention (TOHP I & II): TOHP
back to their original diet, without added salt. BP I was conducted over 18 months’ duration and TOHP
reverted back to normal 6 months later. Study shows II for over 36 months. Lower salt intake (lower by 44
the effect of salt on BP in chimpanzees. mmol and 33 mmol/day) in intervention group was

MU-150.indd 973 29-01-2021 15:01:42


974 SECTION 11 Nephrology

associated with 25% lower CV events. So, besides —— high salt intake—150,
lowering BP, lower salt intake is associated with lower —— intermediate salt intake—100, and
CV events also. 6 A metanalysis of 13 studies, with —— low salt intake—50 mmol/day.

177 025 participants (FU 3.5–19 years) showed that In control group, reduction of sodium intake from the
higher salt intake was associated with greater risk of “high to the intermediate level” over 30 days, resulted in
stroke (RR1.23) and cardiovascular disease (RR 1.14). reduction of systolic BP by 2.1 mm Hg (p<0.001) and in
A Finnish study on impact of salt and CV events, the “from intermediate to the low level” by 4.6 mm Hg
comprising of 1,173 men and 1,263 women found that (p<0.001). In DASH group salt reduction resulted in drop
increased sodium intake by 100 mmol/day increased of BP by 1.3 mm Hg (p=0.03) and 1.7 mm Hg (p<0.01)
the hazard ratio for coronary artery disease, CV “from high to intermediate” and “intermediate to low
disease, and all-cause mortality by 50% (HR of 1.51). salt” group respectively. Benefit of salt reduction was
„„ INTERMAP study: A recent study (Hypertension observed in both sexes, all races and in normotensives
2018;71:631-37)9 included 4,680 persons from China, also. The study inferred that participants in DASH diet
Japan, UK, and the USA. This study addressed the had a significantly lower systolic BP at each sodium level.
effect of dietary salt intake on BP and its possible If one compares control diet group with high salt intake
modulation with other dietary factors. Study found and DASH diet group with low salt intake, there is a BP
that group with two SD higher sodium excretion was difference of 7.1 mm Hg in normotensives and 11.5 mm Hg
associated with rise in BP by 3.5/1.7 mm Hg (p<0.001) in hypertensives.
and most other 26 micro- or macronutrients in diet
Few important points emerge from these studies:
had only a modest countervailing effects on Na-BP „„ Epidemiological data clearly shows low prevalence of
relationship. The study again emphasized the need for
HT in areas with low salt intake.
salt restriction as other micro- or macronutrients did „„ Animal and human trials show drop in BP with
not make much difference.
reducing salt intake and increase in BP with increased
„„ Prospective urban rural epidemiology (PURE) study
salt intake.
(Mente et al., 2018)10 was conducted in 18 countries „„ Cardiovascular mortality also increases with increase
on a population of 1,68,000, in the age group of 35–70 in salt intake.
years. The 664 communities from low, middle, and An important inference is that prevalence of HT and
high income countries were selected. A 24-hour risk of CV mortality is prominent and significant in group
sodium excretion was divided into three tertiles: with highest tertile of salt intake.
low—4.04, middle—4.70, and high tertile—5.75 gm. „„ Salt intake and obesity: It has been observed that
The study found that mean systolic BP increased by excessive salt intake is associated with obesity. As small
2.86 mm Hg per 1 gm increase in salt intake. Sodium as 1 gm of extra salt consumption per day by children
intake was also associated with cardiovascular disease and adolescents has been found to be associated
and strokes. This association was however significant with consumption of 27 gm of sugar-sweetened soft
only in those falling in highest tertile of salt intake drink. There has been controversy, if obesity is due
(p<0.0001). Study suggested that those in highest to consumption of extra sugar or processed food
tertile of salt consumption had significant detrimental or due to extra salt. However, increasing evidence
effect. suggests that there is direct link between salt intake
„„ Dietary approaches to stop hypertension (DASH): In this
and obesity independent of total energy intake. Even
study 412 normotensive and hypertensive participants
after adjusting variables like ethnic group, social status,
were assigned to eat either typical American diet
(control diet) or DASH diet. DASH diet is rich in fruits, energy intake, educational status, smoking, alcohol
vegetables, and has low-fat dairy products. Aim of the consumption, etc., 1 gram a day of extra salt increases
study was to test if DASH diet reduced BP and if salt the risk of obesity by 28% (p=0.0002) in children and
reduction in patients on DASH diet had an additional 26% (p<0.0001) in adults.
advantage. Study population was divided in three „„ Is very low salt intake detrimental: In the “Framingham

groups: Offspring study”—a 16-year follow-up data, of 2,632

MU-150.indd 974 29-01-2021 15:01:42


Dietary Salt and Hypertension CHAPTER 150 975

men and women between 30–64 years, showed that


very low intake of salt (<2.5 gm) is associated with
increased BP. This is in line with many workers who feel
that there is J-shaped curve between salt consumption
and hypertension (Fig. 1).

Salt Sensitive Hypertension


An important and debatable issue is that should salt
restriction advise be universal or only for those having
inappropriately high consumption/salt sensitive
population. Luft et al studied 14 subjects (7 white and 7
black) and gave them increasing doses of salt, 10-1500
meq/day. He found mean rise in blood pressure in both
groups but rise in blood pressure was higher in blacks
(Fig. 2); showing the racial difference. Salt sensitivity can
be studied by giving high and then low salt—1 week of 200
mmol and another week of 30 mmol of salt and then seeing
the BP response. A 24-hour urinary sodium is collected on
last day of the week to see compliance. However, long 29
days compliance for this test is an issue, so an abbreviated
version (7 day test) has been suggested. Rise and drop Fig. 1: Graph shows J-shaped relationship between salt and
in BP by 5–10% is taken as positive test.7 Castiglioni et hypertension

Fig. 2: Percentage change in mean arterial pressure in normotensive subjects receiving incremental increases in sodium. Blood pressure at the
end of 7 days of low (10 mmol/d) salt intake was taken as baseline. All subjects demonstrated an increase in blood pressure with salt loading

MU-150.indd 975 29-01-2021 15:01:43


976 SECTION 11 Nephrology

al.8 suggested that ambulatory BP monitoring (ABPM) 2. Intersalt Cooperative Research Group. Intersalt: an international
is a better option and more practical. It is assumed that study of electrolyte excretion and blood pressure: results for 24
hour urinary sodium and potassium excretion. BMJ. 1988;297(6644):
patients who are salt sensitive retain salt and water with
319-28.
resultant loss of circadian nocturnal dip of BP and higher 3. MacGregor GA, Markandu ND, Sagnella GA, et al. Double-blind
mean heart rate. No salt restriction is required in this test. study of three sodium intakes and long-term effects of sodium
Based on these two parameters Indices have been devised restriction in essential hypertension. Lancet. 1989;2(8674):1244-7.
and patients have been divided into mild, moderate, or 4. World Health Organization. Guideline: sodium intake for adults
highly salt sensitive. Most workers believe that 25% of and children. Available from https://www.who.int/publications/i/
item/9789241504836
general population and 50% of hypertensive population is 5. Weinberger MH. Salt sensitivity of blood pressure in humans.
salt sensitive. Salt sensitivity is largely genetic. Hypertension. 1996;27(3 Pt 2):481-90.
6. Strazzullo P, D Elia L, Kandala NB, et al. Salt intake, stroke, and
Conclusion cardiovascular disease: meta-analysis of prospective studies. BMJ.
2009;339:b4567.
In India, average salt intake is between 9–12 gm/day, against 7. Galletti F, Ferrara I, Stinga F, et al. Evaluation of a rapid protocol
recommended 5 gm by WHO. Most of the epidemiological for the assessment of salt sensitivity against the blood pressure
animal and human data show that high sodium intake in both response to dietary sodium chloride restriction. Am J Hypertens.
normotensive and hypertensive individuals is associated with 1997;10(4):462-6.
age related HT. Excessive salt intake is also associated with 8. Castiglioni P, Parati G, Brambilla L, et al. Detecting sodium-sensitivity
increased cardiovascular mortality and obesity. Reduction of in hypertensive patients: information from 24-hour ambulatory
salt intake reduces BP and CV mortality. Some limited data also blood pressure monitoring. Hypertension. 2011;57(2):180-5.
suggests that very low salt intake (<3 gm) may be detrimental. 9. Stamler J, Chan Q, Daviglus ML, et al. Relation of dietary sodium to
It is recommended to consume salt in moderation (approx. blood pressure and its possible modulation by other dietary factors
5 gm per day). Maximum benefit of salt restriction occurs in The INTERMAP Study. Hypertension. 2018;71(4):631-7.
population consuming high/very high amount of salt. 10. Mente A, O’Donnell M, Rangarajan S, et al. Urinary sodium
excretion, blood pressure, cardiovascular disease, and mortality:
a community-level prospective epidemiological cohort study.
References Lancet. 2018;392(10146):496-506.
1. Denton D, Weisinger R, Mundy NI, Wickings EJ, Dixson A, Moisson 11. Luft FC, Rankin LI, Bloch R, Weyman AE, Willis LR, Murray RH,
P, Pingard AM, Shade R, Carey D, Ardaillou R, Paillard F, Chapman Grim CE, Weinberger MH. Cardiovascular and humoral responses
J, Thillet J, Michel JB. The effect of increased salt intake on blood to extremes of sodium intake in normal black and white men.
pressure in chimpanzees. Nat Med. 1995; 1: 1009–1016. Circulation. 1979; 60:697-706.

MU-150.indd 976 29-01-2021 15:01:43


CHAPTER

151 Newer Avenues in Management


of CKD Anemia
Manish Rathi, Rudreshwar Prabakaran

Abstract 
Anemia remains one of the challenging aspects in the management of chronic kidney disease (CKD) till date. Oral/
intravenous iron and erythropoietin stimulating agents (ESAs) remain the key modalities in the management of anemia in
CKD. Traditional forms of oral iron therapy are limited by poor oral tolerance and insufficient absorption from the gut in
dialysis population. Newer forms of oral iron such as sucrosomial iron and ferric citrate offer advantages such as alternate
mechanism of absorption from the gut, relatively better oral tolerance, and additional properties such as phosphorous
binding. Newer forms of intravenous iron preparations have the advantage of higher stability, lesser risk of infusion
reactions, and bolus dosing. ESAs are being used for the treatment of anemia in dialysis patients since the 1980s. Although
they are effective, higher doses, and higher hemoglobin targets are associated with significant risk of adverse cardiovascular
events such as myocardial infarction and stroke. Studies have shown that though anemia predisposes to a poor quality of
life and high cardiovascular risk in CKD, correction of anemia does not reduce the cardiovascular risk in these patients.
This could be because of the limited options of therapeutic agents available at present and higher doses of both intravenous
iron and ESAs have been shown to predispose to higher cardiovascular risk. Thus, there is a need for agents which can not
only correct anemia but also not contribute to the pre-existing cardiovascular risk in CKD patients. Hypoxia inducible factor
(HIF) stabilizers are one of the newer agents being studied at present in various trials. Studies have shown that these agents
can not only reduce the dosages of intravenous iron and ESAs needed to maintain hemoglobin levels in dialysis patients but
can also reduce cardiovascular risk. They also have beneficial effect on iron profile such as reduction of hepcidin levels,
thus enabling better iron absorption. Thus, research for newer modalities of anemia management in CKD aims to address
not only the hemoglobin levels but also improving the quality of life and longevity of CKD patients.

Introduction dialysis dependent patients.2 Intravenous iron although


rapidly restores iron stores is limited by infusion reactions,
Anemia in Chronic Kidney Disease (CKD) predisposes to
infections, oxidative stress, iron overload, and increased
low quality of life, increased mortality, and cardiovascular
cardiovascular risk.3 ESAs, though they reduce the need
disease risk. 1 The incidence of iron deficiency anemia for blood transfusions, are associated with increased
increases with the progression of CKD. Cornerstone of cardiovascular risk when hemoglobin levels exceed
anemia management in CKD so far has been iron therapy 13 g/dL.4 Thus, at present, search is ongoing for agents
and use of erythropoietin stimulating agents (ESAs). which not only increase hemoglobin levels, but also reduces
Traditional oral iron therapy is limited by poor tolerability, the risk of adverse cardiovascular events seen with the
gastrointestinal adverse event, and poor absorption in present agents.

MU-151.indd 977 29-01-2021 15:01:38


978 SECTION 11 Nephrology

Erythropoietin-stimulating Agents recommend iron therapy if TSAT less than or equal to 30%
and serum ferritin is less than or equal to 500 ng/mL.7
Traditionally used ESAs include epoetin alfa (half-live, t1/2
However, TSAT is not an ideal marker of iron status.
6.8–19.4 hours, 50–100 u/kg/week dosing), darbepoetin
Levels can increase in the setting of inflammation and
alfa (t1/2 25.3–48.8 hours, 0.45 µg/kg per week to every
decrease in the setting of malnutrition predisposing to
2–4 weeks dosing), and methoxypolyethylene glycol-
low and high TSAT respectively if the circulating iron
epoetin beta (t1/2 130 hours, 0.6 µg/kg every 2–4 weeks).
levels are constant. Transferrin levels also exhibit diurnal
Longer half-live are achieved through subcutaneous
variation.11 Serum ferritin being an acute phase reactant
route for epoetin and darbepoetin preparations. Dose
can be increased in later stages of CKD because of
escalations beyond double the initial weight based
systemic inflammation and by itself may not reflect true
dose are discouraged by KDIGO due to risk of adverse
iron status at high levels.12 As a result of these variations
cardiovascular events as observed from the TREAT
NICE guidelines in 201510 recommend not to use TSAT
study. Recently the MIRCERA PASS trial,5 a multicentre
or ferritin levels alone to assess iron deficiency status in
randomized non-inferiority trial, randomized 2818 CKD
CKD. The guidelines recommend the use of percentage of
patients to methoxypolyethylene glycol-epoetin beta
hypochromic red cells (<6%) and reticulocyte hemoglobin
(MIRCERA) and reference erythropoiesis stimulating
content (CHr, <29 pg), if possible, in the place of TSAT and
agents. The primary outcome of the study was composite
ferritin levels.
of time to occurrence of death, non-fatal myocardial
infarction, or nonfatal stroke. This occurred in 45.4% in Newer Forms of Oral Iron Therapy
the MIRCERA group and 45.7% in the reference group.
KDIGO 2012 guidelines recommend that 1–3 months trial
Higher dose of ESAs was associated with higher risk of
of oral iron therapy can be considered in non-dialysis
primary outcome.
dependent CKD population with anemia and TSAT less
than or equal to 30% and serum ferritin less than or equal
Iron Deficiency Anemia in to 500 ng/mL.7 Elemental iron (200 mg) per day is the
Chronic Kidney Disease recommended daily dosage for these patients. In a normal
Iron deficiency is one of the important causes of anemia individual 1–2 mg of dietary iron is absorbed per day and
in CKD. The NHANES (National Health and Nutrition with oral iron supplementation. The maximum absorption
Examination Survey) 1988 to 2004 data of non-dialysis of iron per day during oral iron supplementation is around
dependent CKD showed that low iron stores defined as 25–30 mg/day.13 This is impaired in patients with uremia14
TSAT less than 20% or serum ferritin less than 100 ng/ due to rising hepcidin levels,15 hence oral iron therapy is
mL were present in 57.8–58.8% of men and 69.9–72.8% not very effective in dialysis dependent CKD population.
of women. 6 Causes of iron deficiency in CKD include Oral iron supplementation comes in ferric and ferrous
gastrointestinal bleeding, retention of blood in dialyzers forms. The bioavailability of ferrous form is 10–15%
and blood lines, repeated sampling, surgical procedures whereas that of ferric forms of iron is three to four times
such as arteriovenous fistula creation, drugs such as lower due to reduced solubility of ferric iron in the alkaline
proton pump inhibitors and phosphate binders and media of the gut.16 Several forms of oral iron are available
reduced absorption.7 with wide variations in their extent of absorption and
Transferrin saturation (TSAT) and serum ferritin are adverse effects (Table 1).
widely used as indicators of iron status in CKD population. The most common forms of side effects with oral iron
KDOQI 2006 guidelines recommend maintaining serum therapy are gastrointestinal like nausea, heartburn, pain,
ferritin more than 200 ng/mL with TSAT more than 20% constipation, and diarrhea. This is seen in 30–70% of the
in dialysis dependent CKD population and serum ferritin cases. Among all the oral iron formulations, this risk is
more than 100 ng/mL and TSAT more than 20% in non- highest with ferrous fumarate.10
dialysis dependent CKD population.8 ERBP (European
renal best practice)9 and NICE guidelines10 recommend Ferric Citrate
iron therapy when serum ferritin is less than 100 ng/ Ferric citrate is ferric iron preparation, which was
mL and TSAT less than 20%. KDIGO 2012 guidelines approved as a phosphate binder by US FDA in 2014 for

MU-151.indd 978 29-01-2021 15:01:38


Newer Avenues in Management of CKD Anemia CHAPTER 151 979

TABLE 1 Different forms of available oral iron preparations17


acetate). The trial showed ferric citrate significantly raised
hemoglobin levels, TSAT, and serum ferritin and had a
Agent Elemental iron Salt content comparable phosphate binding ability when compared
per tablet per tablet with active control. Those in ferric citrate arm received
Ferric sulfate 65 mg 325 mg less intravenous iron and dose of erythropoietin also
Ferrous fumarate 106 mg 325 mg significantly reduced with similar adverse events between
Ferrous gluconate 37.5 mg 325 mg the two groups. 22 Median daily dose was 1680 mg of
Ferric citrate 210 mg 1g elemental iron per day. Similar results were shown in 2019
Ferric citrate hydrate 45 mg 250 mg by the ASTRIO study which compared ferric citrate with
Liposomal iron 30 mg 30 mg
non-iron based phosphate binders in 93 hemodialysis
dependent patients.23

dialysis dependent CKD patients. The formation of ferric Ferric Maltol


citrate coordination inhibits the precipitation of ferric iron Ferric maltol is novel preparation consisting of ferric iron
and enables better absorption. Formation of oligomeric complexed with maltol (3-hydroxy-2-methyl-4-pyrone).
complexes in acidic pH enables phosphate binding Its hydrophilic and lipophilic properties enable higher
and monomeric complex formation in alkaline pH of bioavailability and better absorption of ferric iron. Since
duodenum enables ferric iron absorption.17 Ferric citrate it is not a salt-based formulation, iron is directly absorbed
is always administered with meals. from the complex, and the adverse effects due to free iron
seen in salt-based formulations are reduced. Although
Non-Dialysis Dependent CKD (ND-CKD) this compound was described as early as 1980s, it was
Population approved by US FDA for the treatment of iron deficiency
anemia in CKD in 2019.
Phase 3 multicenter double-blind randomized placebo-
AEGIS-CKD study24 is a placebo controlled double blind
controlled trial with primary end point as change in
randomized trial conducted in non-dialysis dependent
serum phosphate showed significant reduction of serum
CKD patients, which showed that the compound was well
phosphate and FGF23 level and significant increase in
tolerated and produced a significant rise in hemoglobin at
serum iron, ferritin, and TSAT compared to placebo. 18
week 16. The dose used was 30 mg twice daily.
Placebo controlled phase 2 (n=149) 19 and phase 3
(n=233)20 trials with primary end point as mean change Sucrosomial Iron
in TSAT/phosphorous and more than or equal to 1 g/
Sucrosomial iron consists of a ferric pyrophosphate
dL hemoglobin rise showed positive results for patients
core surrounded by a phospholipid bilayer consisting of
in ferric citrate arm with similar rate of adverse events
lecithin and sucrester matrix. Ingredients such as starch
between the two groups. Mean drug doses used in the two
and tricalcium phosphate further the coat the structure
studies were 5.4 g and 5.7 g/day. Pooled analysis of both
forming the sucrosome. The phospholipids allow the iron
the trials showed that the percentage of adverse events
to be absorbed in a vesicular form through transcellular
were not different from that seen with older conventional
and paracellular routes. The absorption is mediated by ‘M’
forms of oral iron with gastrointestinal events being the
cells of the Peyer patches.25 Thus, bioavailability of iron is
commonest.21 To our knowledge trials with head to head
high with less free iron mediated gastrointestinal adverse
comparison with other oral forms of iron do not exist.
effects. Cell culture studies using the Caco-2 cell lines
Ferric citrate was approved for the treatment of iron
showed threefold higher absorption rates for sucrosomial
deficiency anemia in ND-CKD patients in 2017.
iron when compared to ferrous sulfate.26 Animal studies
with iron deficient mice have shown that increase in
Dialysis Dependent-CKD (DD-CKD) hepcidin that is seen with other oral iron formulations
Population is not seen with sucrosomial iron. 27 An open label
In a randomized trial of 441 patients, ferric citrate was randomized control of 99 ND-CKD patients28 compared
compared with active control (sevelamer and calcium sucrosomial iron (30 mg/day for 3 months) and IV ferrous

MU-151.indd 979 29-01-2021 15:01:38


980 SECTION 11 Nephrology

gluconate (1 g) in a 2:1 ratio. The study found that at the dialysate iron significantly reduced the need for IV iron
end of 1 month, greater number of patients in the IV iron and increase in ESA dose. Phase 3 CRUISE 1 and 2 studies31
group had increase in hemoglobin, but this difference showed that dialysate iron significantly raised hemoglobin
was absent at the end of 3 months. On discontinuation, when compared to placebo. The study had three stages:
hemoglobin levels were stable in IV iron group whereas Stage 1—run in period, Stage 2—randomization without
it fell to baseline in sucrosomial iron group. Adverse change in ESA dose (no IV iron), and Stage 3—open
events such as hypotension, headache, and infusion label. Hypotension, headache, and muscle spasms were
reaction were more common in IV iron group. Only 5% commonly reported side effects.
of patients experienced gastrointestinal side effects in the
sucrosomial group. Thus, although sucrosomial iron is Intravenous Iron
a safer formulation and high bioavailability, iron stores Intravenous iron preparations contain an iron hydroxide
repletion may be slower when compared to conventional core surrounded by a carbohydrate shell.10 The stability
iron formulations. of this determines how much iron is released into the
circulation at a time. In older preparations such as iron
Dialysate Iron (Ferric Pyrophosphate Citrate) sucrose, adverse effects like infusion reactions and
This water-soluble preparation consists of ferric iron oxidative stress frequent due to low stability of the core
tightly complexed to citrate and pyrophosphate to reduce and higher release of free iron. Newer preparations
the amount of free iron released into the circulation.29 (Table 2) have a more stable core and thus relatively fewer
This form of iron is administered via the bicarbonate adverse effects.10 Table 2 shows the different intravenous
component of the dialysate. On entering the circulation, iron preparations used over the years.
the iron component is directly transferred thus raising KDIGO guidelines 2012 suggests that a trial of
TSAT levels. The advantage of this preparation is that it intravenous iron may be considered in adult CKD patients
reduces risk of iron overload. For an individual patient with anemia and TSAT less than or equal to 30% and
5 mL (5.44 mg/mL) is added to 9.46 liters of bicarbonate serum ferritin less than or equal to 500 ng/mL, who are
concentrate, which gives a concentration 110 µg/L of not on ESAs or in those who are on ESA and increase in
iron in the dialysate. The drug is administered in each hemoglobin or reduction in ESA dose is desired. 6 The
HD session with TSAT and ferritin levels being done guidelines were based on short-term studies with small
every 3 months. Doses are held if TSAT more than 50% number of patients and there were very few trials which
or serum ferritin more than 1000 ng/mL. PRIME, 30 a looked at the safety of giving IV iron in patients with TSAT
phase 2 prospective randomized double blind trial with more than 30% and ferritin more than 500 ng/mL. In 2007,
primary end point as change in ESA dose showed that DRIVE 33 study randomized 134 hemodialysis patients

TABLE 2 Properties of different intravenous iron preparations32

Iron gluconate Iron sucrose Low molecular Iron Iron Ferumoxytol


weight dextran isomaltoside carboxymaltose
Carbohydrate shell Gluconate Sucrose Dextran Isomaltoside Carboxymaltose Polyglucose sorbitol
polysaccharide carboxymethylether
Stability of complex Low Medium High High High High
Labile iron release High High Medium Low Low Low
Plasma half-life (hrs) 1 6 5–20 20 7–12 15
Maximum single 125 mg 200 mg 20 mg/kg 20 mg/kg 1000 mg 510 mg
dose
Minimum infusion 30–60 60 60 15 15 15
time (min)

MU-151.indd 980 29-01-2021 15:01:38


MU-151.indd 981
TABLE 3 HIF alpha stabilizers and their related trials

Drug Dosage used in Trials Primary end point Results Other observations
studies
Roxadustat 20–250 mg zz Phase 341 (n=154) multicentre, Mean change in 1.9±1.2 g/dL raise in zz Significant reduction in hepcidin
double blind trial comparing hemoglobin over week hemoglobin in roxadustat and cholesterol levels in roxadustat
roxadustat with placebo in 7 through 9. group (p<0.001) group. Hyperkalemia and metabolic
non-dialysis dependent CKD Mean change in Greater mean change in acidosis seen more in roxadustat
42
zz Phase 3 (n=305) study hemoglobin level from hemoglobin in roxadustat group
comparing roxadustat baseline during weeks group zz Significant reduction in mean

and epoetin alfa in dialysis 23–27 hepcidin and cholesterol levels in


dependent CKD roxadustat group. Hyperkalemia and
upper respiratory tract infection seen
more in roxadustat group. Greater
rise in Hemoglobin in those with
higher CRP
43
Molidustat 25–200 mg zz Phase 2b trial (16 weeks) Change in hemoglobin DIALOGUE 1: Significant Estimated difference in mean change in
DIALOGUE 1: 25, zz DIALOGUE 1(n=121): level between baseline number of patients hemoglobin between molidustat and
50, or 75 mg once randomized double-blind and the mean value achieved the estimated darbepoetin was 0.6 g/dL
daily/25 or 50 mg control trial comparing from the last 4 weeks of mean hemoglobin in the Lower starting doses were associated
twice daily molidustat and placebo for the treatment period molidustat group with a fall below target range in
DIALOGUE 2: 25, 50, patients not in dialysis Change in hemoglobin DIALOGUE 2: Mean hemoglobin in the first week. Small
or 75 mg once daily zz DIALOGUE 2(n=124): open from baseline to each hemoglobin was increase in hemoglobin was seen with
(plus additional 15, label molidustat in previously post-baseline visit maintained in the target starting dose of 150 mg/day
100, or 150 mg) darbepoetin treated patients range for each dose in Side effects: Adverse effects were
DIALOGUE 3: 25, 50, versus continuing darbepoetin molidustat group comparable between both the groups
75, or 150 mg once for patients not on dialysis DIALOGUE 3: Mean and were mild to moderate in intensity
daily (plus additional zz DIALOGUE 3 (n=199): open hemoglobin levels were in all three trials. Numerically more
15, 100, and 200 mg label molidustat in previously maintained for 75 mg– number of patients had hypertension
once daily) epoetin alfa or beta treated 150 mg daily as a starting and nasopharyngitis in molidustat
patients versus continuing dose group
epoetin for patients on dialysis Mean hemoglobin Mean hemoglobin levels were
zz DIALOGUE EXTENSION concentration during maintained from baseline and
STUDIES: study were 11.10±0.508 throughout the study period in both the
44
—— DIALOGUE 4 (<36 g/dL in molidustat group groups. Similar percentage of adverse
months): all patients from and 10.98±0.571 g/dL in events in both the group (85.6% vs.
DIALOGUE 1 and 2 who darbepoetin group 85.7%). 21% discontinued drug in
achieved their mean Hb Mean hemoglobin molidustat arm when compared to 10%
targets were made to concentration during in darbepoetin arm
continue their respective study were 10.37±0.56 g/ Mean hemoglobin levels were
treatment for 36 months dL in molidustat group maintained from baseline and
Newer Avenues in Management of CKD Anemia

44
—— DIALOGUE 5: patients and 10.52±0.47 g/dL in throughout the study period in both the
from DAILOGUE 4 who epoetin group groups. Similar percentage of adverse
achieved mean Hb targets events in both the groups (91.25%
were continued on their vs. 92.2%). More number of patients
treatments for 36 months experienced severe adverse events in
molidustat arm (51% vs. 37%). More
number of patients discontinued in
CHAPTER 151

molidustat arm (23% vs. 7%)


Contd...
981

29-01-2021 15:01:39
982

MU-151.indd 982
Contd...
SECTION 11

Drug Dosage used in Trials Primary end point Results Other observations
studies
Vadadustat 450–600 mg 1) Phase 2b study (non-dialysis Percentage of patients 54.9% patients in More number of patients had serious
300 mg OD, 450 dependent CKD, n=210)45 who in the last 2 weeksvadadustat arm achieved adverse events in the vadadustat arm
mg OD and 450 mg 2) Open label phase 2 trial achieved Hb ≥11 g/ primary endpoint compared to placebo (23.9% vs. 15.3%).
thrice weekly (n=94)46 comparing vadadustat dL or an increase in compared to 10.3% in Three deaths occurred in vadadustat
Nephrology

and epoetin alfa Hb ≥1.2 g/dL over the placebo arm


predose average No statistically significant Nausea, diarrhea, and vomiting were the
Mean change in hemoglobin from common adverse effects noted in the
in hemoglobin pre-baseline average was vadadustat group. No serious adverse
concentration observed for all three event was noted in the same
doses
Daprodustat 10, 25, 50, and 100 1) Multicenter, single blind, Increase and response Both non dialysis and Treatment was discontinued in 33% in
mg placebo-controlled study (n=70, rates in achieving target dialysis dependent the ND-CKD group and 22% in DD-CKD
0.5, 2, and 5 mg NDD-CKD and 37, DD-CKD)47 hemoglobin, plasma population showed a group due to high rate of hemoglobin
2) Phase 2a placebo-controlled Erythropoietin level and dose dependent increase rise (≥1 g/dL in any 2 week period)
study (n=73, NDD-CKD and n=83, reticulocyte count in hemoglobin, plasma or high absolute Hb value (≥13 g/
DD-CKD)48 Change in hemoglobin EPO concentrations dL). Hepcidin level decreased and
over 4 weeks of and reticulocyte count TIBC and unsaturated iron binding
treatment compared to placebo capacity increased significantly in the
NDD-CKD group: Dose daprodustat group
dependent increase in Hemoglobin rise occurred with
hemoglobin concurrent rise in endogenous
DD-CKD group: 5 mg once erythropoietin levels in daprodustat
daily dose maintained group. The drug was well tolerated
mean hemoglobin
levels after switch from
erythropoietin
Enarodustat 2 ,4, and 6 mg 1) Phase 2 study placebo Rate of rise in ESA naïve group: Dose Ferritin and hepcidin levels decreased
controlled randomized trial in hemoglobin per week dependent increase TIBC increased in enarodustat group
nondialysis dependent CKD in ESA naïve group and in hemoglobin in
(n=94, ESA naïve group and proportion of patients enarodustat group
n=103, who previously received who maintained Previously ESA treated
ESA)49 change in hemoglobin group: 70% of subjects
in previously ESA in enarodustat arm
treated group maintained their
hemoglobin levels over
24 weeks

29-01-2021 15:01:39
Newer Avenues in Management of CKD Anemia CHAPTER 151 983

with ferritin 500–1200 ng/mL and TSAT less than or equal nature (Table 3). Phase 3 studies are ongoing. So far, phase
to 25% to intravenous ferric gluconate and no iron. The 2 studies have shown that these drugs are well tolerated
study found that hemoglobin increased significantly in the in both dialysis dependent and dialysis independent
IV iron group with similar side effects between both the CKD population. Apart from correction of anemia, other
groups. FIND-CKD study in 2014 found that hemoglobin effects such as reduction in cholesterol levels have been
rise was significant in ND-CKD patients randomized to documented in studies.38 Preliminary data from phase 3
high ferritin targets (400–600 µg/L) when compared to trials (DOLOMITES study, NCT02021318) of Roxadustat
lower targets. REVOKE34 trial in 2015 which randomized has shown that it reduces major adverse cardiac events by
patients to oral ferrous sulfate and IV iron sucrose found 30%. Despite their beneficial effects shown in the studies,
similar results but with increased serious adverse events other proposed harmful effects such as promotion of
in the Intravenous group. PIVOTAL, 35 an open label tumor growth39 by increasing VEGF levels are yet to be
multicenter trial in 2019 randomized 2141 patients on studied. Long-term studies with large numbers of patients
maintenance hemodialysis less than 1 year to IV iron are required to enable safe introduction of these drugs into
sucrose in a proactive fashion (400 mg monthly, unless clinical practice.
TSAT ≥40% or ferritin ≥700 µg/L) or reactive fashion (400
mg if ferritin ≤200µg/L or TSAT <20%). The primary end Other Upcoming Therapeutic Strategies
point was composite of nonfatal MI, stroke, death, and
Lexaptepid pegol is a pegylated L-oligo-ribonucleotide,
hospitalization for heart failure. The study found that
which inactivated hepcidin. Phase 1 studies 40 of the
hemoglobin rise was rapid, blood transfusion, and ESA
compound have shown good safety profile and dose
dosage was reduced in the proactive group. Adverse effects
dependent reduction of hepcidin levels in healthy
were similar between the groups and the most common
volunteers and in hemodialysis patients. Activins are
adverse event was infection. Thus, higher ferritin targets
dimmers, which belong to transforming growth factor beta
than that proposed by 2012 KDIGO guidelines, may reduce
(TGF-β) family which influence erythropoiesis. Sotatercept
the need for blood transfusion and higher ESA exposure.
is a fusion protein of Fc domain of human IgG1 and activin
receptor IIa. Phase 2 studies in hemodialysis patients
HIF Stabilizers have shown acceptable safety profiles, stable hemoglobin
Hypoxia inducible factors (HIFs) are transcription levels, and lower rates of rescue with ESAs.
factors made of α (1α, 2α, and 3α) and β subunits. 36
HIF 1α is widely expressed across all normal tissues Conclusion
whereas HIF 2α expression is restricted to endothelium,
selected cells in the kidney, gut, lung, liver, and carotid Newer modalities of management of anemia in CKD patients
are aimed at development of agents, which can reduce the
body. During normoxia, the enzyme prolyl hydroxylase
need for blood transfusion, need for escalation of dose of ESAs
(PHD1, PHD2, and PHD3) hydroxylate prolyl residues
and maintain stable hemoglobin levels for prolonged periods
in the alpha subunit of HIF. Hydroxylation leads to
of time without increasing the risk for adverse cardiovascular
recognition by von Hippel Lindau (VHL) ubiquitin E3 events associated with the current available agents.
ligase and subsequent proteosomal degradation of HIF.
During hypoxic conditions, the PHDs are inactive and
this leads to dimerization of α and β subunits of HIF and References
target gene expression. HIF 2α regulates erythropoiesis. 1. Iimori S, Naito S, Noda Y, et al. Anaemia management and mortality
HIF 2 binds to hypoxia response elements of genes risk in newly visiting patients with chronic kidney disease in Japan:
encoding proteins such as duodenal cytochrome B and The CKD-ROUTE study. Nephrology (Carlton). 2015;20(9):601-8.
ferroportin and enables iron absorption. 37 HIF 2α also 2. Hörl WH. Iron therapy for renal anemia: how much needed, how
much harmful? Pediatr Nephrol. 2007;22(4):480-9.
suppresses hepcidin levels which are elevated in chronic
3. Macdougall IC, Bircher AJ, Eckardt KU, et al. Iron management
inflammatory states such as ESRD.38 HIF alpha stabilizers in chronic kidney disease: conclusions from a “Kidney Disease:
which are currently under study are mentioned in Table 3. Improving Global Outcomes” (KDIGO) Controversies Conference.
Most of the trials related to HIF stabilizers are phase 2 in Kidney Int. 2016;89(1):28-39.

MU-151.indd 983 29-01-2021 15:01:39


984 SECTION 11 Nephrology

4. Pfeffer MA, Burdmann EA, Chen CY, et al. TREAT Investigators: a trial 21. Chertow GM, Block GA, Neylan JF, et al. Safety and efficacy of ferric
of darbepoetin alfa in type 2 diabetes and chronic kidney disease. citrate in patients with nondialysis-dependent chronic kidney
N Engl J Med. 2009;361(21):2019-32. disease. PLoS One. 2017;12(11):0188712.
5. Locatelli F, Hannedouche T, Fishbane S, et al. Cardiovascular safety 22. Umanath K, Jalal DI, Greco BA, et al. Ferric citrate reduces
and all-cause mortality of methoxy polyethylene glycol-epoetin intravenous iron and erythropoiesis-stimulating agent use in ESRD.
beta and other erythropoiesis-stimulating agents in anemia of J Am Soc Nephrol. 2015;26(10):2578-87.
CKD: a randomized noninferiority trial. Clin J Am Soc Nephrol. 23. Yokoyama K, Fukagawa M, Akiba T, et al. Randomised clinical trial
2019;14(12):1701-10. of ferric citrate hydrate on anaemia management in haemodialysis
6. Fishbane S, Pollack S, Feldman HI, et al. Iron indices in chronic patients with hyperphosphataemia: ASTRIO study. Sci Rep.
kidney disease in the National Health and Nutritional Examination 2019;9:8877.
Survey 1988-2004. Clin J Am Soc Nephrol. 2009;4(1):57-61. 24. Kopyt NP. AEGIS-CKD Study Group. Efficacy and safety of oral ferric
7. Kidney Disease: Improving Global Outcomes (KDIGO) Anemia Work maltol (FM) in treating iron-deficiency anemia (IDA) in patients with
Group. KDIGO clinical practice guideline for anemia in chronic CKD: randomized controlled trial [FR-OR120]. J Am Soc Nephrol.
kidney disease. Kidney Int Suppl. 2012;2(4):279-335. 2018;29:70-1.
8. KDOQI; National Kidney Foundation. KDOQI clinical practice 25. Gómez-Ramírez S, Brilli E, Tarantino G, et al. Sucrosomial® Iron:
guidelines and clinical practice recommendations for anemia in a new generation iron for improving oral supplementation.
chronic kidney disease. Am J Kidney Dis. 2006;47(5.3):11-145. Pharmaceuticals (Basel). 2018;11(4):97.
9. Locatelli F, Covic A, Eckardt K, et al. Anaemia management in 26. Tarantino G, Brilli E, Zambito Y, et al. Sucrosomial® Iron: a new highly
patients with chronic kidney disease: a position statement by the bioavailable oral iron supplement. Blood. 2015;126(23):45-61.
Anaemia Working Group of European Renal Best Practice (ERBP). 27. Asperti M, Gryzik M, Brilli E, et al. Sucrosomial® Iron supplementation
Nephrol Dial Transplant. 2009;24(2):348-54. in mice: effects on blood parameters, hepcidin, and inflammation.
10. Ratcliffe LE, Thomas W, Glen J, et al. Diagnosis and management
Nutrients. 2018;10(10):1349.
of iron deficiency in CKD: a summary of the NICE guideline
28. Pisani A, Riccio E, Sabbatini M, et al. Effect of oral liposomal iron
recommendations and their rationale. Am J Kidney Dis.
versus intravenous iron for treatment of iron deficiency anaemia
2016;67(4):548-58.
in CKD patients: a randomized trial. Nephrol Dial Transplant.
11. Wish JB. Assessing iron status: beyond serum ferritin and transferrin
2015;30(4):645-52.
saturation. Clin J Am Soc Nephrol. 2006;1(1):4-8.
29. Fishbane S, Shah HH. Ferric pyrophosphate citrate as an iron
12. Kalantar-Zadeh K, Kalantar-Zadeh K, Lee GH, et al. The fascinating
replacement agent for patients receiving hemodialysis. Hemodial
but deceptive ferritin: to measure it or not to measure it in chronic
Int. 2017;21(1):104-9.
kidney disease? Clin J Am Soc Nephrol. 2006;1(1):9-18.
30. Gupta A, Lin V, Guss C, et al. Ferric pyrophosphate citrate
13. Girelli D, Ugolini S, Busti F, et al. Modern iron replacement
administered via dialysate reduces erythropoiesis-stimulating
therapy: clinical and pathophysiological insights. Int J Hematol.
agent use and maintains hemoglobin in hemodialysis patients.
2018;107(1):16-30.
Kidney Int. 2015;88(5):1187-94.
14. Nakazawa R, Kaneko M, Hoshi H, et al. Iron absorption in patients
with chronic uremia on maintenance hemodialysis and in healthy 31. Fishbane SN, Singh AK, Cournoyer SH, et al. Ferric pyrophosphate
volunteers measured with a simple oral iron load test. Nephron. citrate ( Triferic™) administration via the dialysate maintains
1996;73(3):403-6. hemoglobin and iron balance in chronic hemodialysis patients.
15. Ganz T, Nemeth E. Iron balance and the role of hepcidin in chronic Nephrol Dial Transplant. 2015;30(12):2019-26.
kidney disease. Semin Nephrol. 2016;36(2):87-93. 32. Auerbach M, Macdougall I. The available intravenous iron
16. Muñoz M, Gómez-Ramírez S, Bhandari S, et al. The safety of formulations: history, efficacy, and toxicology. Hemodial Int.
available treatment options for iron-deficiency anemia. Ex Op Drug 2017;21(1):83-92.
Saf. 2018;17(2):149-59. 33. Coyne DW, Kapoian T, Suki W, et al. Ferric gluconate is highly
17. Pergola PE, Fishbane S, Ganz T, et al. Novel oral iron therapies for efficacious in anemic hemodialysis patients with high serum
iron deficiency anemia in chronic kidney disease. Adv Chronic ferritin and low transferrin saturation: results of the Dialysis Patients’
Kidney Dis. 2019;26(4):272-91. Response to IV Iron with Elevated Ferritin (DRIVE) Study. J Am Soc
18. Yokoyama K, Hirakata H, Akiba T, et al. Ferric citrate hydrate for the Nephrol. 2007;18(3):975-84.
treatment of hyperphosphatemia in nondialysis-dependent CKD. 34. Agarwal R, Kusek JW, Pappas MK, et al. A randomized trial of
Clin J Am Soc Nephrol. 2014;9(3):543-52. intravenous and oral iron in chronic kidney disease. Kidney Int.
19. Block GA, Fishbane S, Rodriguez M, et al. A 12-week, double-blind, 2015;88(4):905-14.
placebo-controlled trial of ferric citrate for the treatment of iron 35. Macdougall IC, White C, Anker SD, et al. Intravenous iron in
deficiency anemia and reduction of serum phosphate in patients patients undergoing maintenance hemodialysis. N Eng J Med.
with CKD stages 3–5. Am J Kidney Dis. 2015;65(5):728-36. 2019;380(5):447-58.
20. Fishbane S, Block GA, Loram L, et al. Effects of ferric citrate in 36. Schödel J, Ratcliffe PJ. Mechanisms of hypoxia signalling:
patients with nondialysis-dependent CKD and iron deficiency new implications for nephrology. Nature Rev Nephrol. 2019;
anemia. J Am Soc Nephrol. 2017;28(6):1851-8. 15(10):641-59.

MU-151.indd 984 29-01-2021 15:01:39


Newer Avenues in Management of CKD Anemia CHAPTER 151 985

37. Taylor M, Qu A, Anderson ER, et al. Hypoxia-inducible factor-2alpha 44. Akizawa T, Macdougall IC, Berns JS, et al. Long-term efficacy
mediates the adaptive increase of intestinal ferroportin during iron and safety of molidustat for anemia in chronic kidney disease:
deficiency in mice. Gastroenterol. 2011;140(7):2044-55. DIALOGUE Extension Studies. Am J Nephrol. 2019;49(4):271-80.
38. Liu Q, Davidoff O, Niss K, et al. Hypoxia-inducible factor regulates 45. Pergola PE, Spinowitz BS, Hartman CS, et al. Vadadustat, a novel oral
hepcidin via erythropoietin-induced erythropoiesis. J Clin Invest. HIF stabilizer, provides effective anemia treatment in nondialysis-
2012;122(12):4635-44. dependent chronic kidney disease. Kidney Int. 2016;90(5):1115-22.
39. Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev 46. Haase VH, Chertow GM, Block GA, et al. Effects of vadadustat on
Cancer. 2013;13(12):871-82. hemoglobin concentrations in patients receiving hemodialysis
40. Renders L, Budde K, Rosenberger C, et al. First-in-human Phase I previously treated with erythropoiesis-stimulating agents. Nephrol
studies of PRS-080#22, a hepcidin antagonist, in healthy volunteers Dial Transplant. 2019;34(1):90-9.
and patients with chronic kidney disease undergoing hemodialysis. 47. Brigandi RA, Johnson B, Oei C, et al. A Novel Hypoxia-Inducible
PLoS One. 2019;14(3):0212023. Factor-Prolyl Hydroxylase Inhibitor (GSK1278863) for anemia
41. Chen N, Hao C, Peng X, et al. Roxadustat for anemia in patients in CKD: a 28-Day, Phase 2A Randomized Trial. Am J Kidney Dis.
with kidney disease not receiving dialysis. N Engl J Med. 2016;67(6):861-71.
2019;381(11):1001-10. 48. Holdstock L, Meadowcroft AM, Maier R, et al. Four-Week Studies
42. Chen N, Hao C, Liu BC, et al. Roxadustat treatment for anemia of Oral Hypoxia-Inducible Factor-Prolyl Hydroxylase Inhibitor
in patients undergoing long-term dialysis. N Engl J Med. GSK1278863 for treatment of anemia. J Am Soc Nephrol.
2019;381(11):1011-22. 2016;27(4):1234-44.
43. Macdougall IC, Akizawa T, Berns JS, et al. Effects of molidustat in 49. Akizawa T, Nangaku M, Yamaguchi T, et al. A placebo-controlled,
the treatment of anemia in CKD. Clin J Am Soc Nephrol. 2019;14(1): randomized trial of enarodustat in patients with chronic kidney
28-39. disease followed by long-term trial. Am J Nephrol. 2019;49(2):165-74.

MU-151.indd 985 29-01-2021 15:01:39


CHAPTER

152 SGLT2 Inhibitors—Mechanisms


of Cardiorenal Protection
Ashwini Balasaheb Gadde, Vijay Kher

Abstract 
Sodium Glucose co-transporter-2 (SGLT2) inhibitors are a class antihyperglycemic agents, which act by selectively
inhibiting SGLT2 present in the proximal tubules in the kidney causing glucosuria and to some extent natriuresis. Though
originally invented as one of the oral hypoglycemic agent (OHA), many randomized controlled trials have confirmed
their role far beyond this, in cardiovascular (CV) and renoprotection. This is in sharp contrast to other OHAs which were
associated with either mild CV benefits or rather harm. The benefit possibly extends to nondiabetic population as well.
Though SGLT2 inhibitors act mainly by causing glucosuria, natriuresis, and by inhibiting tubuloglomerular feedback
(TGF), these effects are insufficient to explain the impressive CV and renal outcomes. In this review we aim to explain the
possible mechanisms of cardiorenal protection of SGLT2 inhibitors.

Introduction a paradigm shift in the management of CV and renal


complications of type 2 diabetic patients2-5 and may be in
Sodium Glucose co-transporter-2 (SGLT2) inhibitors
nondiabetic CKD patients as well.6
(-gliflozins) are a class antihyperglycemic agents, which
act by selectively inhibiting SGLT2 present in the proximal
tubules in the kidney, leading to increased glucosuria and Basic Mechanism of Action of
thereby decreasing blood glucose levels. Though initially SGLT2 Inhibitors9
approved in 2013 as one of the oral antihyperglycemic Approximately 90% filtered glucose reabsorption is
agents for type 2 diabetes, the outcomes in EMPA-REG mediated by SGLT2 channels located on S1 segment of
OUTCOME trial opened a window to its role far beyond proximal convoluted tubules (PCT). These channels are also
glycemic control.1 This study established a remarkable responsible for 5–14% of Sodium reabsorption depending
and unexpected cardiovascular (CV) and renal benefits upon the glycemic status. Thus, the physiological effects
of empagliflozin in patients with type 2 diabetes with of SGLT2 inhibitors are a consequence of both glucosuria
clinical cardiovascular disease (CVD). Later many studies and natriuresis.
supported and strengthened their role in wide class of Before going in detail, let’s concentrate on the glucose
patients, including those without established CVD proving absorption in diabetes mellitus and effects of SGLT2
their beneficial effects in diabetes mellitus as primary as inhibitors on tubuloglomerular feedback (TGF)—an
well as secondary prevention for CV & renal endpoints.2-7 important mechanism for glomerular hemodynamic
Table 1 summarizes the CV and renal outcomes in major alteration of in type 2 diabetes and diabetic nephropathy
trials. These trials have established SGLT2 inhibitors as (Fig. 1).

MU-152.indd 986 29-01-2021 15:01:34


SGLT2 Inhibitors—Mechanisms of Cardiorenal Protection CHAPTER 152 987

TABLE 1 Cardiovascular and renal outcomes of SGLT2 inhibitors in trials

Trial Population Cardiac end point Renal end points


Hazard ratio (confidence interval) Hazard ratio (confidence interval)
EMPA-REG OUTCOME1 T2D+CVD 3-point MACE* Doubling of serum creatinine with eGFR<45,
N=7020 eGFR>/30 0.86 (0.74–0.99) initiation of RRT or kidney-related death
0.54 (0.40–0.75)
CANVAS program2 T2D+CVD (if>/30 yrs) or 3-point MACE* Progression of albuminuria 0.73 (0.67–0.79)
n=10,142 >2 CV risk factors if >/50 yrs 0.86 (0.75–0.97) 40% reduction in eGFR, RRT, or renal-related
death 0.60 (0.47–0.77)
DECLARE -TIMI584 T2D+CVD/>2 CV risk factors 3-point MACE* >40% decrease in eGFR to <60, ESRD or
N=17,160 0.93 (0.0.84–1.03) non-significant. renal-related death
Composite of CV death or HHF 0.83 0.53 (0.43–0.66)
(0.73–0.95)
CREDENCE 3 T2D+ eGFRof 30 to <90, Composites of CV death or HHF 0.69 Relative risk of the primary outcome
N=4401 albuminuria and on stable (0.57–0.83) composite of ESRD, doubling of the serum
dose of ACEi/ARBs for >/4 3-point MACE 0.80 (0.67–0.95) creatinine level from baseline or death from
weeks 3 HHF 0.61(0.47–0.80) renal or cardiovascular disease.
Was 30% lower (hazard ratio, 0.70
(0.59–0.82)*
DAPA-HF5 Heart failure and an ejection The primary composite outcome of A composite of worsening renal function
N=4744 fraction of 40% or less worsening heart failure or CV death 0.71 (0.44–1.16) §
(irrespective of the diabetes 0.74 (0.65–0.85)
status)
DAPA-CKD6 Adults with CKD with an The composite of CV death or HHF Primary composite endpoint 
N=4304 eGFR ≥25 but ≤75 mL/ 0.71 (0.55 to 0.92). zz Time to ≥50% eGFR decline from baseline

min/1.73 m2 and a UACR (confirmed by ≥28-day serum creatinine)


≥200 mg/g but ≤5000 mg/g zz Time to ESRD defined as eGFR <15 mL/

on stable dose of ACEi/ARBs min/1.73 m2, need for chronic dialysis


for >/4 weeks (both confirmed after ≥28 days) and renal
transplantation
zz Time to renal or cardiovascular death

0.61 (0.51 to 0.72)


*Primary outcome, §Not significant
3-point MACE, major adverse cardiac events (composite of nonfatal stroke, nonfatal MI, cardiovascular death); ACEi/ARB, angiotensin converting
enzyme inhibitors/aldosteron receptor blockers; CKD, chronic kidney disease; CVD-cardiovascular disease; ESRD, end stage renal disease; HHF,
hospitalization for heart failure; RRT, renal replacement therapy; T2D, type two diabetes; UACR, urine albumin creatinine ratio

In hyperglycemic state, filtered glucose load efferent arteriolar vasoconstriction due to increase in renin
is increased. To handle that load, there is increased secretion and increased renin–angiotensin–aldosterone
expression of SGLT2 on PCT. Along with glucose, sodium system (RAAS) activation.8 High SNGFR further increases
reabsorption also increases. This increased work of Na and the work of reabsorption continuing this vicious cycle.
glucose reabsorption leads to increased cortical oxygen Hyperfiltration and increase in intraglomerular pressure
consumption and also tubular hypertrophy, resulting also causes proteinuria which is nephrotoxic and
in renal cortical ischemia, which promotes interstitial contributes to progression of diabetic nephropathy.
fibrosis.8 As more glucose and sodium are absorbed in SGLT2 inhibitors interfere with these essential
PCT, less sodium will be delivered to distal convoluted pathophysiological effects in a diabetic kidney (Fig. 2).
tubules (DCT), which is sensed by macula densa. This By inhibiting SGLT2 channels, hyper-reabsorption of Na
causes increase in single nephron glomerular filtration and glucose reabsorption is inhibited thus decreasing
rate (SNGFR) via TGF. Due to the decreased sodium in the tubular work load and oxygen consumption avoiding
DCT (TGF), there is afferent arteriolar dilatation and cortical ischemia. Because of the inhibited Na and

MU-152.indd 987 29-01-2021 15:01:34


988 SECTION 11 Nephrology

Fig. 1: Glucose absorption in diabetes mellitus and effects of SGLT2 on tubuloglomerular


feedback and in evolution of diabetic nephropathy
JGA, juxtaglomerular apparatus; SNGFR, single nephron glomerular filtration rate; MD, macula densa

Fig. 2: Effect of SGLT2 inhibitors on pathophysiological mechanisms on diabetic nephropathy

glucose reabsorption in PCT, more is delivered the decrease in intraglomerular hypertension, glomerular
DCT. This causes afferent arteriolar constriction and blood flow leading to decrease in proteinuria.
decreases SNGFR via TGF mechanism. Also because of These effects to some extent do explain the renal
increased load of sodium and potassium in DCT, tubular benefits of SGLT2 inhibitors but may be inadequate to
backpressure in the Bowman’s capsule increases, leading explain the mechanism for CV benefits. Although the
to decrease in SNGFR. These effects together lead to glucose lowering efficacy of SGLT2 inhibitors declines at

MU-152.indd 988 29-01-2021 15:01:36


SGLT2 Inhibitors—Mechanisms of Cardiorenal Protection CHAPTER 152 989

the lower eGFR range, the CV benefits are persistent across inhibitors is another pathway hypothesized in natriuresis
wide spectrum kidney disease (eGFRs of 30–60 mL min−1 and reduction in BP.
[1.73 m]−2, 60–90 mL min−1 [1.73 m]−2 and >90 mL min−1 In EMPA-REG trial, SGLT2 inhibitors reduced systolic/
[1.73 m]−2).10 Also, beneficial effects of SGLT2 inhibitors diastolic BP by around 5/2 mm Hg.1 A recent meta-analysis
extend to nondiabetic population as well (Table 1).6 This of 43 randomized controlled trials with 22,528 patients
implies that many “extrarenal” effects might play a role. assessed the seated clinic blood pressure effects of SGLT2
inhibitors in patients with type 2 diabetes mellitus. The
Metabolic Effects Secondary to Glucosuria reduction in blood pressure was over and above the
already receiving antihypertensive therapy.16
Reduction in HbA1c
Placebo substracted HbA1c difference in EMPA-REG was Diuretic Effect
mild (0.4%) but it showed impressive 38% reduction in
SGLT2 inhibitors decrease preload by both natriuresis and
CV death, 35% reduction in heart failure, and 46% risk
osmotic diuresis secondary to glucosuria. But those are
reduction was seen in composite renal outcome. This is
different from other conventional loop/thiazide diuretics.
in sharp contrast with other oral hypoglycemic agents
They do not cause reflex sympathetic activity thus
(OHAs), which were associated with either mild CV
causing no compensatory tachycardia.17 It is postulated
benefits or sometimes even harm. 11-13 So, mechanisms
that as opposed to diuretics, SGLT2 inhibitors promote
other than this need to looked into.
a greater decrease in interstitial fluid relative to blood
It is important to note that SGLT2 inhibitors do
volume.18 This may have significant benefits in reducing
not cause hypoglycemia. This is due to intact counter-
neurohormonal activation via their effects on RAAS. Also,
regulatory mechanisms including upregulation of hepatic
thiazide/loop diuretics are known to cause hyperuricemia
gluconeogenesis. Also, SGLT1 downstream prevents
and sometimes hyperglycemia, but these parameters are
glucose excretion during SGLT2 inhibition when the
positively affected by SGLT2 inhibitors.
filtered glucose falls below the transport capacity of
SGLT1.7
Decrease in Intraglomerular Hypertension
Weight Loss As explained earlier, natriuresis also causes increased
SGLT2 inhibitors reduce body weight and fat mass, delivery of sodium to macula dense which will lead to
especially epicardial fat which is important for leptin (a afferent arteriolar constriction via TGF mechanism.
proinflammatory adipokine) secretion. In a 52-week study This decreases the intraglomerular hypertension and
comparing canagliflozin with glimepiride, canagliflozin hyperfilteration occurring in early diabetic nephropathy.
reduced serum leptin levels by 25% and increased the This will also decrease proteinuria but at the cost of initial
levels of the anti-inflammatory adipokine adiponectin dip in eGFR (around 4–6 mL/min) in initial 3–4 weeks,1
by 17%. 14 The antinatriuretic, anti-inflammatory and which is reversible either after stopping medication or
antifibrotic effects of SGLT2 inhibitors (discussed further) sometimes even after continuous prolonged treatment.
antagonise the deleterious effects of leptin on heart and This suggests that it’s related to a functional change rather
kidneys.14 than structural changes, similar to angiotensin-converting
enzyme inhibitor (ACEi)/angiotensin receptor blockers
Decrease in Uric Acid15 (ARBs).
Though benefit of this effect on CV protection is unclear. Currently, ACEi/ARBs are the standard treatment for
this purpose, which dilates efferent arteriole. In EMPA-
Hemodynamic Effects Secondary REG as well as other studies related to SGLT2i, most of
to Natriuresis the patient population (almost 80%) was already on ACEi/
ARBs. Benefits of SGLT2i are additive to current optimal
Blood Pressure Reduction therapy. RAAS blockers are active through neurohormonal
Along with SGLT2 inhibition, direct inhibition of the pathways of hyperfiltration whereas SGLT2 inhibitors
cardiac sodium-hydrogen exchanger (NHE)1 by SGLT2 work via tubular pathway.

MU-152.indd 989 29-01-2021 15:01:36


990 SECTION 11 Nephrology

Though above mentioned hemodynamic and key enzymes in this respect. SIRT1 decreases oxidative
metabolic effects have positive impact on CV outcome, stress by enhancing antioxidant activity, AMPK reduces
in practice, this impact is unexpectedly strong. Previously the formation of reactive oxygen species, In addition, both
it was thought that these CV benefits are because of the enzymes inhibit energy storage (glycogen synthesis and
effect on atherosclerosis, but benefits are seen as early as 3 lipogenesis) while promoting energy utilization (fatty acid
months from the start of treatment. It is very unlikely that oxidation leading to ketonemia). Coordinated activation
atherosclerosis-related effect will show impact so early. So, of two signaling stimulates autophagy which is a lysosome
more possible mechanisms have been proposed and are mediated pathway that removes potentially dangerous
getting tested. constituents and recycles cellular components. Since type
2 diabetes is perceived by cells as a state of nutrient excess,
Cardiac Fuel Energetics it is accompanied by suppression of SIRT1 and AMPK.
SGLT2 acts as a sensor of energy overabundance in this
So to explain these remarkable cardiac and renal benefits,
situation. SGTL2 inhibitors block this sensing mechanism
Ferrannini et al. came up with a “Thrifty substrate”
and inhibits suppression of this SIRT1/AMPK pathway.
hypothesis.19 Glycosuria lowers insulin levels and raises
fasting and post-meal glucagon concentrations. This
causes restriction of glucose utilization and increase in lipid Hemoconcentration/Erythropoietin
mobilization. Increased delivery of free fatty acids (FFAs) to Activation
the liver stimulates ketogenesis. In conditions of prolonged SGLT2 inhibitors may not only deceive cells into believing
hyperketonemia, b-hydroxybutyrate is freely taken up by that they are fasting but also that they are hypoxic, which
the heart and oxidized in preference to fatty acids. The activate hypoxia-inducible factor-2α (HIF-2α) via this
benefit of preferential fuel selection was demonstrated in SIRT1/AMPK pathway. Also, decreased reabsorption in
a study of 3-hydroxybutyrate versus placebo in humans early PCT causes compensatory increased transport work
with chronic HF in which ketone infusion increased stroke in late PCT and medullary thick ascending loop. This leads
volume, cardiac output, and LVEF in a dose responsive to relative medullary hypoxia and stimulation of HIF 1&2
fashion.20 This fuel selection improves work efficiency with which cause increase in erythropoietin secretion. Elevated
respect to oxygen consumption especially, in failing heart hematocrit is also a surrogate marker of reduced plasma
and also in kidneys.21 But recently this theory has been volume as well as of recovery of tubulointerstitial function
challenged. Animal studies to prove this change in cardiac associated with SGLT2 inhibitor therapy. In mediation
energetics have been inconclusive. Also, the stressed analysis of EMPA-REG, changes in hematocrit contributed
heart already preferentially utilizes ketone bodies, and the 51.8% of the effect of empagliflozin versus placebo on
diabetic kidney is a ketogenic organ21,22 so what difference the risk of CV death, maximum of all the effect of SGLT2
does SGLT2 inhibitors do? It needs to be delineated and inhibitors.24
explored further.
Direct Effect on LV Mass
Cellular Reprogramming to Along with decreasing preload and afterload, it is possible
“Dormancy State” that SGLT2 inhibitors have direct beneficial impact
Another theory seems to be that instead of postulating of cardiac remodeling. The EMPA-Heart trial which
a drug-induced enhancement of fuel supply, Avogaro included patients with T2D and coronary artery disease
et al. suggest that SGLT2 inhibitors induce a “dormancy demonstrated a reduction in LV mass indexed to body
surface area in patients treated with empagliflozin. This
state” that mimics starvation.23 When cells are stressed
was thought to be in part due to a reduction in wall stress.25
by starvation or by hypoxia or reactive oxygen species,
injured organelles, and misfolded proteins, they
activate a transcriptional program to adapt to this low- Effect on NLRP3 Inflammasome
nutrient conditions. Sirtuin 1 (SIRT1) and adenosine Activation of the NLR family, pyrin domain-containing 3
monophosphate–activated protein kinase (AMPK) are the (NLRP3) inflammasome in the innate immune cells and

MU-152.indd 990 29-01-2021 15:01:36


SGLT2 Inhibitors—Mechanisms of Cardiorenal Protection CHAPTER 152 991

subsequent interleukin (IL)-1β release has been proposed 6. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al; Dapagliflozin in
as one of the pathogenic mechanisms in diabetes, Patients with Chronic Kidney Disease. N Engl J Med. 2020;383(15):
1436-46.
atherosclerosis, and heart failure. SGLT2 inhibitors
7. Verma S, McMurray JJV. SGLT2 inhibitors and mechanisms of
have been shown to inhibit this effect with respect to cardiovascular benefit: a state-of-the-art review. Diabetologia.
sulfonylureas.26 2018;61(10):2108-17.
8. Peti-Peterdi J, Harris RC. Macula densa sensing and signaling
Effect on Arterial Stiffness mechanisms of renin release. J Am Soc Nephrol. 2010;21(7):1093-6.
9. Nespoux J, Vallon V. SGLT2 inhibition and kidney protection. Clin Sci
A post-hoc analysis of trials and study comparing (Lond). 2018;132(12):1329-39.
dapagliflozin with hydrochlorothiazide demonstrated that 10. Wanner C, Lachin JM, Inzucchi SE, et al. Empagliflozin and clinical
empagliflozin was associated with not only decreased BP, outcomes in patients with type 2 diabetes mellitus, established
but showed positive effects on markers of arterial stiffness cardiovascular disease, and chronic kidney disease. Circulation.
and vascular resistance, that is, aortic pulse wave velocity, 2018;137(2):119-29.
11. Fitchett DH, Udell JA, Inzucchi SE. Heart failure outcomes in clinical
brachial flow mediated dilation, and shear rates.27,28
trials of glucose-lowering agents in patients with diabetes. Eur J
In addition to these mechanism, many others are Heart Fail. 2017;19(1):43-53.
being considered and getting investigated in animal/ 12. Son JW, Kim S. Dipeptidyl peptidase 4 inhibitors and the risk of
human-inhibition of sodium-hydrogen exchanger 1 on cardiovascular disease in patients with type 2 diabetes: a tale of
cardiac myocytes,29 increase in Circulating Pro-Vascular three studies. Diabetes Metab J. 2015;39(5):373-83.
13. Marso SP, Daniels GH, Brown-Frandsen K, et al. Liraglutide and
Progenitor Cells.30
cardiovascular outcomes in type 2 diabetes. N Engl J Med.
2016;375(4):311-22.
Conclusion 14. Garvey WT, Van Gaal L, Leiter LA, et al. Effects of canagliflozin versus
glimepiride on adipokines and inflammatory biomarkers in type 2
To conclude, though the exact mechanism of cardiorenal
diabetes. Metabolism. 2018;85:32-7.
protection is still unclear, one thing is clear that we need
15. Zhao Y, Xu L, Tian D, et al. Effects of sodium-glucose co-transporter
to expand the horizon of SGLT2 inhibitors from a “glucose 2 (SGLT2) inhibitors on serum uric acid level: a meta-analysis of
lowering agent” to “organ protective” agent. Studies are randomized controlled trials. Diabet Obes Metab. 2018;20(2):
underway trying to find its use in nondiabetic population 458-62.
(DAPA CKD, EMPA-KIDNEY, TRANSLATE). Results of DAPA-HF 16. Mazidi M, Rezaie P, Gao HK, et al. Effect of sodium-glucose
trial have shown positive results in nondiabetic heart failure cotransport-2 inhibitors on blood pressure in people with type
patients.5 There were initial safety concerns with respect to 2 diabetes mellitus: a systematic review and meta-analysis of 43
acute kidney injury, diabetic ketoacidosis, amputations, urinary randomized control trials with 22,528 patients. J Am Heart Assoc.
and genital tract infections, bladder cancer, and bone fractures. 2017;6(6):e004007.
However, recent trials have downplayed these risks, and 17. Herat LY, Magno AL, Rudnicka C, et al. SGLT2 inhibitor–induced
sympathoinhibition. J Am Coll Cardiol Basic Trans Science.
benefits are found to outweigh risks.
2020;5(2):169-79.
18. Hallow KM, Helmlinger G, Greasley PJ, et al. Why do SGLT2
inhibitors reduce heart failure hospitalization? A differential volume
References regulation hypothesis. Diabetes Obes Metab. 2018;20(3):479-87.
1. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular 19. Ferrannini E, Mark M, Mayoux E. CV Protection in the EMPA-REG
outcomes, and mortality in type 2 diabetes. N Engl J Med. OUTCOME Trial: a “Thrifty Substrate” hypothesis. Diabetes Care.
2015;373(22):2117-28. 2016;39(7):1108-14.
2. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and 20. Nielsen R, Moller N, Gormsen LC, et al. Cardiovascular effects of
cardiovascular and renal events in type 2 diabetes. N Engl J Med. treatment with the ketone body 3-hydroxybutyrate in chronic
2017;377:644-57. heart failure patients. Circulation. 2019;139(18):2129-41.
3. Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin and renal 21. Aubert G, Martin OJ, Horton JL, et al. The failing heart relies on
outcomes in type 2 diabetes and nephropathy. N Engl J Med. ketone bodies as a fuel. Circulation. 2016;133(8):698-705.
2019;380(24):2295-306. 22. Zhang D, Yang H, Kong X, et al. Proteomics analysis reveals diabetic
4. Wiviott SD, Raz I, Bonaca MP, et al. Dapagliflozin and cardiovascular kidney as a ketogenic organ in type 2 diabetes. Am J Physiol
outcomes in type 2 diabetes. N Engl J Med. 2019;380(4):347-57. Endocrinol Metab. 2011;300(2):E287-95.
5. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in 23. Avogaro A, Fadini GP, Del Prato S. Reinterpreting cardiorenal
patients with heart failure and reduced ejection fraction. N Engl J protection of renal sodium–glucose cotransporter 2 inhibitors via
Med. 2019;381(21):1995-2008. cellular life history programming. Diabetes Care. 2019;43(3):501-7.

MU-152.indd 991 29-01-2021 15:01:36


992 SECTION 11 Nephrology

24. Inzucchi SE, Zinman B, Fitchett D, et al. How does empagliflozin resistance in patients with type 2 diabetes. Diabetes Obes Metab.
reduce cardiovascular mortality? Insights from a mediation analysis 2015;17(12):1180-93.
of the EMPA-REG OUTCOME trial. Diabetes Care. 2018;41(2):356-63. 28. Solini A, Giannini L, SeghieriM, et al. Dapagliflozin acutely improves
25. Verma S, Mazer CD, Yan AT, et al. EMPA-heart cardiolink-6 trial: endothelial dysfunction, reduces aortic stiffness and renal resistive
a randomized trial evaluating the effect of empagliflozin on index in type 2 diabetic patients: a pilot study. Cardiovasc Diabetol.
left ventricular structure, function and biomarkers in people 2017;16(1):1-9.
with type 2 diabetes and coronary artery disease. Circulation. 29. Uthman L, Baartscheer A, Bleijlevens B, et al. Class effects of SGLT2
2019;140(21):1693-702. inhibitors in mouse cardiomyocytes and hearts: inhibition of
26. Kim SR, Lee S, Kim SH, et al. SGLT2 inhibition modulates NLRP3 Na+/H+ exchanger, lowering of cytosolic Na+ and vasodilation.
inflammasome activity via ketones and insulin in diabetes with Diabetologia. 2018;61(3):722-6.
cardiovascular disease. Nat Commun. 2020;11(1):2127. 30. Hess DA, Terenzi DC, Trac JZ, et al. SGLT2 Inhibition with
27. Chilton R, Tikkanen I, Cannon CP, et al. Effects of empagliflozin empagliflozin increases circulating provascular progenitor cells in
on blood pressure and markers of aterial stiffness and vascular people with type 2 diabetes mellitus. Cell Metab. 2019;30(4):609-13.

MU-152.indd 992 29-01-2021 15:01:36


CHAPTER

153 Optimizing Renal


Care in Elderly
Narinder Pal Singh, Rahul Mishra, Shaurya Kaul

Abstract 
The global burden of chronic kidney disease (CKD) is increasing particularly among elderly. Aging itself increases
vulnerability for poor renal health. High cost of management and poor outcomes of CKD necessitate optimization of renal
care to be started early in the course of the disease. Collaborative involvement of patient, caregiver, primary care provider
and multidisciplinary clinics is required to achieve optimal conservative care and renal replacement modality. Dietary
changes, lifestyle modification, managing the pill-burden, appropriate nutrient supplementation, and vaccination to take
care of risks and complications of CKD are key steps as per latest evidences available. For those progressing to end-stage
renal disease (ESRD), an approach, taking into consideration of patient’s perspectives and functional and cognitive status,
to decide appropriateness of renal replacement therapy (RRT) or end-of-life care is a very essential particularly for elderly
patients. There is need of progressive work for optimizing various facets of renal care in elderly patients particularly in
economically constrained regions where most of the CKD patients fail to receive appropriate care.

Introduction arteriosclerosis, glomerulosclerosis, interstitial fibrosis,


tubular atrophy), and functional decline in total-kidney
The global burden of kidney disease, which has been
GFR due to decrease in total nephron count. 3 Optimal
rising consistently, poses a major challenge to the primary
care of CKD risk factors can delay or even prevent this
care physician. Recent GBD reported that out of 697.5
million worldwide CKD patients, India harbored 115.1 progression of healthy aging of kidneys into CKD. Elderly
million. 1 As per latest census in India (2011), around population needs special attention to their physical and
104 million people were 60 years or above and by 2021 functional deficits including multiple co-morbidities
this number is predicted to reach 133.32 million. So, (hypertension, diabetes), cognitive impairment, frailty
the likely CKD burden among elderly is expected to and progressive sensory impairment. Owing to high
rise proportionately. Indian CKD registry2 reported that cost and poor outcomes of CKD management, there is a
diabetic nephropathy was most common cause (31%) for definitive need for optimization of care in elderly CKD
CKD and mean age of CKD patient was 50 years. Prevalence patients on individual basis. Optimal care goals include
of CKD and its risk factors like diabetes, hypertension, best outcomes at individual, population and society
obesity, and cardiovascular disease increases with levels for betterment of survival and quality of life. The
age. Age-related changes in kidney include structural management of advanced CKD in the elderly can be
changes like macrostructure changes (decrease in cortical challenging in regards to prediction of disease progression
volume, increase in surface roughness, and size of simple to ESRD, selection of RRT modality, and choice of optimal
renal cysts), microstructure changes (nephrosclerosis- vascular access (VA) for hemodialysis (HD). There is a

MU-153.indd 993 29-01-2021 15:01:29


994 SECTION 11 Nephrology

need for collaborative efforts between patient, caregiver, Providing recommended vaccinations—hepatitis B,
primary care provider, and interaction of multidisciplinary pneumococcal, influenza, and other routine vaccines
clinics. Current review will focus on optimizing renal are essential for optimum care of CKD patients.11 Focus
care including conservative medical management and should be laid upon patient’s values and perspectives
selection of RRT modalities such as HD, peritoneal dialysis about the disease, as deficiency about health literacy is
(PD) and kidney transplant (KT) for elderly to maximize widespread in CKD population.12,13 Information should
functional status and minimize treatment-associated be provided using simple terms and techniques like
morbidity. “teach back” to ensure attention and understanding.14
Routine screening for anxiety, depression, and cognitive
Pre-RRT Phase: Elderly Focused impairment may be important because CKD is associated
with increased risk for dementia in elderly and poor
Conservative Care
functional status (including poor overall cognition,
In India, a country with 1.3 billion people and around language, and memory). Various mediators accounted for
two thousand nephrologists, practices that promote cognitive derangement like anemia, increased oxidative
renal health and prevent renal disease progression stress, inflammatory markers, and changes in lipid and
are important. Optimal care for elderly involves mix of homocysteine metabolism are found in CKD patients.15
pharmacological and non-pharmacological interventions. Identifying functional changes is important in order to
Diet may be important in deciding course of CKD before optimize the management outcome, as studies have
dialysis. Moderate intake of protein, especially dietary shown association of cognitive impairment with negative
strategies that increase plant protein, low sodium, outcome and increased risk of 1-year mortality among
dietary fiber, vitamin D, and reduction in obesity may elderly severe CKD patients who were in-hospital.16-18 The
limit worsening of CKD. 4 Renoprotective effects of conservative pharmacological management is sometimes
maintaining muscle health, especially among elderly the only choice in elderly patients who are declining or not
who have high prevalence of sarcopenia, are documented fit for RRT (Flowchart 1).
and they must be stressed upon in routine care.5 Life
style changes can be recommended based on specific
comorbidity of CKD patients, for example, increasing the
Risks and Complications of CKD
aerobic exercise capacity in those with cardiovascular CKD, due to any cause, can progress through I to V stages.
disease, high dietary fiber and moderate intensity exercise Complications and their management begin usually by
for patients with chronic lung disease or rheumatoid stage III onward, and by stage IV aim is to prepare the
arthritis and weight reduction for diabetic patients. 6 patient for dialysis with suitable measures like VA to be
CKD patients are likely to encounter polypharmacy needed during stage V. Appropriate timely management
risks, potential drug-drug interactions (pDDI) and is required, particularly in elderly patients who may have
inappropriate prescriptions. Incorporating procedures multiple modifiable risk factors that could prevent or delay
and pharmacists to determine pDDI into the kidney this progression.
care model could help dealing these challenges. 7,8
Non-adherence is a possible outcome of multiple pill Diabetes Mellitus
burden among CKD patients. Whenever possible during Diabetes mellitus is the most common cause of CKD and
clinical management, tapering thereafter deprescription this holds true for both the developed and the developing
strategies should be planned. Recommendations have world. Diabetic glomerulopathy might be contributing
been made on approach to deprescribe PPIs, statins and to over half of the ESRD cases. Bringing down of blood
oral hypoglycemic agents in CKD patients to reduce pill glucose levels (to preserve average HbA1c 7%) reflects
burden.9 Practices involving identification and avoidance as delay in urinary albumin excretion, GFR decline, and
of agents like nephrotoxic antimicrobials, radio-contrast requirement for dialysis. The use of ACEI/ARBs can
exposure, combining ACEI and ARBs, NSAIDs and further prevent kidney damage in normotensive diabetics
diuretics that might precipitate AKI should be promoted.10 with proteinuria.19 In the elderly, frail, and CKD patients,

MU-153.indd 994 29-01-2021 15:01:29


Optimizing Renal Care in Elderly CHAPTER 153 995

Flowchart 1: Options for renal care in elderly Dapagliflozin) have been demonstrated in diabetic kidney
disease.22

Hypertension
With regards to CKD, hypertension is related as both the
cause and effect. Hypertension is a major risk factor that
increases the risk of progression of kidney disease and
increase the risk of cardiovascular complications. The
KDIGO (2012) guidelines have described the general
management strategies for controlling blood pressure in
non-dialysis-dependent CKD patients. A combination
of lifestyle changes and pharmacological management
are the key approach to achieve target BP goal in CKD
patient. Gradually lowering of blood pressure should
be recommended. Blood pressure readings based
upon ambulatory and self-measurements better reflect
hypertension compared to office blood pressure readings
the consequences of hypoglycemia are very serious and allow individualizing of antihypertensive treatment.
including injury, myocardial infarction, stroke, and death. Blood pressure targets need to be individualized and
Therefore, a tailored approach for target HbA1c levels medical management should be based on the age,
should be followed for elderly CKD patients to avoid risk of presence of illness and end-organ damage (cardiovascular
hypoglycemia.20 Metformin is usually the first antidiabetic and retinopathy), progression of renal disease, and
to begin with. For reducing risk of lactic acidosis, a rare tolerance to treatment. Vascular stiffness necessitates
event, FDA recommends avoiding its use if creatinine in periodic monitoring for postural hypotension as it causes
men >1.5 mg/dL and women >1.4 mg/dL. Sulfonylureas higher systolic blood pressure whereas the diastolic blood
undergo renal clearance; therefore, risk of hypoglycemia pressure could still decline. Slowing of the progression of
is more in advanced CKD patients and their use should be kidney disease and reduction in CVD risk are the goals
avoided if GFR <30 mL/min/1.73 m2. Among meglitinides, of antihypertensive therapy and target BP, as suggested
repaglinide is safe to use in CKD. Thiazolidinediones are by KDIGO guidelines for non-dialysis patients, 140/90
hepatically metabolized so can be used in CKD with caveat mm Hg in nondiabetics/diabetics without proteinuria
that fluid retention and increased risk of fracture rates in and 130/80 mm Hg in patients with proteinuria, is a
women especially with underlying renal osteodystrophy key strategy to prevent further renal function decline.
could be concerning. Insulin is safe for all CKD patients Initial fixed dose RAAS inhibitor (ACEI/ARBs) based
and long acting single dose can be used when oral combination therapy (coadministration of CCB, diuretic,
agents fail to obtain target sugar levels. However, more α-blocker, or β-blocker) is more effective and efficient
complicated regimens with multiple insulin dosing may than sequential monotherapy for achieving target blood
increase chances of error and hypoglycemia, especially pressure and reduces risk of adverse events by allowing
among elderly with cognitive impairment. Among GLP1 use of lower doses of each drug.
agonists, no dose adjustments are needed for dulaglutide
or albiglutide in CKD while exenatide and liraglutide Anemia
should be avoided with eGFR <30 mL/min/1.73 m2 due In the elderly especially frail elderly with CKD,
to poor renal clearance. DPP4 inhibitors (sitagliptin, anemia is related to poor function and quality of life,
saxagliptin, linagliptin, vildagliptin) are well tolerated increased frequency and duration of hospital stays
and have some renal clearance and require dosage and mortality. 23 Normocytic normochromic anemia a
adjustment. 21 Cardioprotective and renoprotective common complication seen in CKD patients. A thorough
effect of SGLT2 inhibitors (Empagliflozin, Canagliflozin, assessment of other causes of anemia starting with

MU-153.indd 995 29-01-2021 15:01:30


996 SECTION 11 Nephrology

complete blood count, peripheral smear, iron studies, elderly patients on dialysis as compared to those with no
B12 and folate levels should be performed before labeling RRT patients cannot be ruled out; however, they tend to
anemia of CKD. As per KDOQI 2012 guidelines, target spend lot of time around health-care facility and may not
hemoglobin between 10–11.5 gm/dL is desired in all get life satisfaction.27 Assessing HD accessibility for each
CKD patients. Consider ESA in pre-dialysis and dialysis elderly patient opting for RRT is necessary as caregivers
patients with Hb below 10 g/dL and between 9–10 g/dL, and patients feel overwhelmed and burdensome about
respectively.24 KDOQI Anemia Work Group recommends frequent multiple visits (at least thrice weekly) to
sufficient iron supplementation to maintain serum ferritin a faraway located dialysis center. The approach of
concentration >200 ng/mL in HD patients and >100 ng/ incremental dialysis in elderly may assist them in adjusting
mL in non-dialysis or on peritoneal dialysis CKD patients to dialysis and sustaining residual kidney function.28
for optimal erythropoiesis.24 Suggested incremental dialysis approach involves 1–2
dialysis session weekly, each around 1.5–2.5 hours
Vitamin and Electrolyte Abnormality duration until worsening of renal function necessitates
Vitamins and minerals supplementation is critical further modification. Home dialysis can be suitable for
for elderly care. For CKD patients who are already those able to perform it by themselves or with a family
deficient (S.vit. D <20 ng/mL) or insufficient (S.vit. member support. However, it may not be preferred choice
D = 20–29 ng/mL) in vitamin D, adequate vitamin for elderly with multiple comorbidity, frailty, and lack of
D is absolute requirement for preventing secondary support. Exploration of ideal RRT has led to understanding
hyperparathyroidism (SHPT) and its complications. that diverse modalities are used by patients during the
Supplementation with ergocalciferol or cholecalciferol entire course of CKD. A term coined as “Integrated Care”
should be done as per individual needs. However, once has been popularized.29,30 Originally, it suggested starting
SHPT develops, vitamin D receptor agonists and/or with PD and then changing to in-center HD (CHD). The
centers pursuing this approach had reported survival
calcimimetics are required. Ensuring normal reference
benefits and cost optimization.31,32 PD needs assistance
levels of electrolytes like calcium and phosphorous is
and can be preferred as home based dialysis modality in
essential for prevention of bone disorders among CKD
presence of an assistant. However, PD may not be suitable
patients. Dyselectrolytemias including hypo- and hyper-
for elderly having poor functional status and declining
natremia and hyperkalemia are well associated with aging
vision. PD is relatively contraindicated among those with
kidneys.25 Caution with use of potassium sparing drugs
severe pulmonary disease, irreducible hernias, active
among CKD patients is required to avoid serious adverse
inflammatory bowel disease, significant scar from previous
events. Metabolic acidosis in CKD patients is often due
abdominal surgery, colostomy, ileostomy, or gastric
to impaired ammonia excretion and its management, for
tubes.33 Decision about choice of dialysis modality should
example using sodium bicarbonate, improves nutritional
also match patient’s values with treatment characteristics
parameters as well as CKD progression.26
in order to maximize achievable quality of life in elderly.

Renal Replacement Therapy Vascular Access


CKD patients are prone to progress to ESRD. Options of RRT An effective VA is very essential for carrying out HD and
include HD, which can be provided in-center or at home, comprises of arteriovenous fistula (AVF), arteriovenous
PD, and KT. Economic constraint limits RRT availability graft (AVG), and central venous catheter (CVC). For long,
to majority of ESRD patients in India. Elderly patient on as first choice, AVF has been created in the non-dominant
HD are physically frail with multiple comorbidities and hand. Procedure began at a distal site to safeguard
functional dependencies. Initiation of dialysis in elderly proximal blood vessels for future use, thus radiocephalic
should use multidisciplinary approach taking into account fistulas became initial choice.34 This site also reported to
various factors including life expectancy, pros and cons have higher failure rate because of thrombosis and inability
of each dialysis modality, quality of life, and patient and to mature, requiring more surgeries and use of tunneled
caregiver preferences. Possibility of survival benefit for catheter for dialysis. Moreover, preserving a venous

MU-153.indd 996 29-01-2021 15:01:30


Optimizing Renal Care in Elderly CHAPTER 153 997

location in a frail elderly may not be prudent due to fixed Kidney Transplant—An Option in Elderly?
life anticipation, so, the most appropriate AVF, utilizing
Although, age itself is not a contraindication to KT, but
the most excellent vessels should be created first. 35,36
poor accessibility and associated comorbidities could
Complications such as stenosis, thrombosis, and distal
make elderly ineligible for getting a transplant organ.
hypoperfusion ischemic syndrome (DHIS) are concerning
There is lower 5-year survival probability among KT
and add to heavy cost and morbidity with access. Therefore,
recipients aged >65 years compared to aged 35–49 years
varying views arise stating creation of upper arm AVFs in
(61% vs. 75% respectively).42 Though Kaul et al. reported,
elderly as standard care or preferring AVGs and CVCs as
in a retrospective study of ESRD patients, that after
more suitable options depending on individual patient.
adjusting for albumin and BMI, KT group had better
De Silva et al. found similar survival for elderly aged >80
short-term and long-term survival as compared to PD
years using either AVFs or AVGs, though, that could be
group.43 So, despite increased age, in the absence of other
attributed to lesser number of patients receiving AVG
significant factor limiting the life expectancy, KT could
(25.4%) as compared to AVF (43.2%) that were chosen
be beneficial for elderly. 44 Factors that reduce KT rate
for tunneled dialysis.37 Risk of death is more than risk of
in elderly may be strict selection criteria, health-care
progression to ESRD in most elderly CKD, especially if age
providers’ apprehensions and decreased willingness
>85 years, so benefits of an invasive procedure should be
among older patients for kidney transplantation.45 Thus,
weighed against associated complication and additional
instead of kidney transplantation, most of the elderly with
cost arising out of any potential unnecessary procedure.27
ESRD undergo dialysis.
O’Hare et al. demonstrated that only about 25% and 33%
of patients started dialysis within 6 month and 1 year,
respectively, after access creation in 85–100 year old Social Support and End-of-Life Care
patients with eGFR <15 mL/min/1.73 m2.38 Therefore, life Like most non-communicable diseases, the treatment for
expectancy and rate of deterioration of disease should CKD is non-curative, thus patient has to learn living with
form basis for timing and type of access creation in it. This creates functional restrain thereby limiting social
elderly. For example, a tunneled catheter usually not involvement and a feeling of being isolated. Increased
the most appropriate access choice, could be considered requirement of assistance for executive functions,
for those with a life very limited life expectancy (e.g., <6 dependency for care including dialysis, adjusting to
months). CVCs are preferred in IJV and femoral vein since new pattern of daily routine and cognitive changes of
subclavian vein has high risk of thrombosis. Infections, as ageing may cause a lot of stress, resulting in a feeling of
the complication, amount to about 30–60% of HD CVCs helplessness. Social support including emotional and
removal and also, hospitalization rates are higher with instrumental is essential for overcoming. Instrumental
CVCs than AVF.39 Active surveillance and monitoring of support helps patient carrying out all routine and financial
AVF is recommended. Physical examination of VA is very activities while emotional support enables person to
useful tool to assess inconvenience during cannulation or feel loved and cared.46 Ultimately, terminal patient-care
clot aspiration, bleeding at cannulation sites post dialysis needs interdisciplinary management. End-of-life care
or failure to attain target blood flows while dialysis. If discussion involving patient and family members, taking
any such sign present, further evaluation with direct care of their preferences to reach a consensus regarding
flow rate [using Doppler US and magnetic resonance accepting any intervention or simply withdrawing dialysis
angiography (MRA)], or indirect flow rate measurement for palliative care and reviewing goals of advance care
modality [UD ( Transonic), timed ultrafiltration should be considered. Kirchhoff et al. observed that after
methods, ionic dialysance, differential conductivity, and intervening with patient-centered advance care planning,
glucose infusion] should be considered. 40 A low-cost a significant percentage of ESRD patients withdrew dialysis
method using hemoglobin dilution test was described by as compared to those without intervention.47 Informed
Tiranathanagul et al. which can be used in the resource choice about all available options, including their pros and
limited areas where ultrasound dilution test (UDT) is cons, can augment decision-making especially for patients
costly and unavailable.41 on HD among whom 9–13% succumb within 1 year.48

MU-153.indd 997 29-01-2021 15:01:31


998 SECTION 11 Nephrology

Conclusion 14. Liu Y-B, Li Y-F, Liu L, et al. Effectiveness of the teach-back method for
improving the health literacy of senior citizens in nursing homes.
Optimization of renal care in a developing country, where Jpn J Nurs Sci JJNS. 2018;15(3):195-202.
health-care infrastructure is already struggling, is challenging. 15. Fried LF, Lee JS, Shlipak M, et al. Chronic kidney disease and
Regardless, perpetual efforts for active care, prevention from functional limitation in older people: health, aging and body
and preparation for deteriorating CKD stages and befitting RRT composition study. J Am Geriatr Soc. 2006;54(5):750-6.
are needed. Optimal care of elderly patients with CKD may be 16. Di Rosa M, D’Alia S, Guarasci F, et al. Cognitive impairment, chronic
kidney disease, and 1-year mortality in older patients discharged
rewarding by decelerating fall in renal functioning, better care
from acute care hospital. J Clin Med. 2020;9(7):2202.
of complications, and enabling a rational choice of RRT with
17. Shirazian S, Grant CD, Aina O, et al. Depression in chronic kidney
well-timed carried out VA.
disease and end-stage renal disease: similarities and differences
in diagnosis, epidemiology, and management. Kidney Int Rep.
2016;2(1):94-107.
References 18. Etgen T, Chonchol M, Förstl H, et al. Chronic kidney disease and
1. Bikbov B, Purcell CA, Levey AS, et al. Global, regional, and national cognitive impairment: a systematic review and meta-analysis. Am J
burden of chronic kidney disease, 1990–2017: a systematic Nephrol. 2012;35(5):474-82.
analysis for the Global Burden of Disease Study 2017. The Lancet. 19. National Kidney Foundation. KDOQI Clinical Practice Guideline for
2020;395(10225):709-33. Diabetes and CKD: 2012 Update. Am J Kidney Dis Off J Natl Kidney
2. Rajapurkar MM, John GT, Kirpalani AL, et al. What do we know Found. 2012;60(5):850-86.
about chronic kidney disease in India: first report of the Indian CKD 20. Hahr AJ, Molitch ME. Management of diabetes mellitus in patients
registry. BMC Nephrol. 2012;13(1):10. with chronic kidney disease. Clin Diabetes Endocrinol. 2015;1(1):2.
3. Hommos MS, Glassock RJ, Rule AD. Structural and functional 21. Bittle PA. The use of dipeptidyl peptidase-4 inhibitors in patients
changes in human kidneys with healthy aging. J Am Soc Nephrol. with type 2 diabetes & chronic kidney disease. Nurse Pract.
2017;28(10):2838-44. 2017;42(6):31-8.
4. Filipowicz R, Beddhu S. Optimal nutrition for predialysis chronic 22. Górriz JL, Navarro-González JF, Ortiz A, et al. Sodium-glucose
kidney disease. Adv Chronic Kidney Dis. 2013;20(2):175-80. cotransporter 2 inhibition: towards an indication to treat diabetic
5. Zelle DM, Klaassen G, van Adrichem E, et al. Physical inactivity: a risk kidney disease. Nephrol Dial Transplant. 2020;35(Suppl 1):i13-23.
factor and target for intervention in renal care. Nat Rev Nephrol. 23. Musio F. Kidney disease and anemia in elderly patients. Clin Geriatr
2017;13(3):152-68. Med. 2019;35(3):327-37.
6. Peng S, Shen F, Wen A, et al. Detecting lifestyle risk factors for 24. Drüeke TB, Parfrey PS. Summary of the KDIGO guideline on anemia
chronic kidney disease with comorbidities: association rule and comment: reading between the (guide)line(s). Kidney Int.
mining analysis of web-based survey data. J Med Internet Res. 2012;82(9):952-60.
2019;21(12):e14204. 25. Schlanger LE, Bailey JL, Sands JM. Electrolytes in the aging. Adv
7. Molnar AO, Bota S, Jeyakumar N, et al. Potentially inappropriate Chronic Kidney Dis. 2010;17(4):308-19.
prescribing in older adults with advanced chronic kidney disease. 26. de Brito-Ashurst I, Varagunam M, Raftery MJ, et al. Bicarbonate
PloS One. 2020;15(8):e0237868. supplementation slows progression of CKD and improves
8. Santos-Díaz G, Pérez-Pico AM, Suárez-Santisteban MÁ, et al. nutritional status. J Am Soc Nephrol JASN. 2009;20(9):2075-84.
Prevalence of potential drug-drug interaction risk among chronic 27. Berger JR, Jaikaransingh V, Hedayati SS. End-stage kidney disease in
kidney disease patients in a Spanish Hospital. Pharmaceutics. the elderly: approach to dialysis initiation, choosing modality, and
2020;12(8):713. predicting outcomes. Adv Chronic Kidney Dis. 2016;23(1):36-43.
9. Triantafylidis LK, Hawley CE, Perry LP, et al. The role of deprescribing 28. Kalantar-Zadeh K, Crowley ST, Beddhu S, et al. Renal replacement
in older adults with chronic kidney disease. Drugs Aging. therapy and incremental hemodialysis for veterans with advanced
2018;35(11):973-84. chronic kidney disease. Semin Dial. 2017;30(3):251-61.
10. Fassett RG. Current and emerging treatment options for the elderly 29. Davies SJ, Van Biesen W, Nicholas J, et al. Integrated care. Perit Dial
patient with chronic kidney disease. Clin Interv Aging. 2014;9:191-9. Int J Int Soc Perit Dial. 2001;21(Suppl 3):S269-74.
11. Guidelines for vaccination in patients with chronic kidney disease. 30. Mendelssohn DC, Pierratos A. Reformulating the integrated care
Indian J Nephrol. 2016;26(7):15. concept for the new millennium. Perit Dial Int J Int Soc Perit Dial.
12. Morton RL, Tong A, Howard K, et al. The views of patients and 2002;22(1):5-8.
careers in treatment decision making for chronic kidney disease: 31. Ghaffari A, Kalantar-Zadeh K, Lee J, et al. PD First: peritoneal
systematic review and thematic synthesis of qualitative studies. dialysis as the default transition to dialysis therapy. Semin Dial.
BMJ. Available from https://www.bmj.com/content/340/bmj.c112 2013;26(6):706-13.
13. Taylor DM, Fraser SDS, Bradley JA, et al. A systematic review of the 32. Chuengsaman P, Kasemsup V. PD First Policy: Thailand’s response
prevalence and associations of limited health literacy in CKD. Clin J to the challenge of meeting the needs of patients with end-stage
Am Soc Nephrol. 2017;12(7):1070-84. renal disease. Semin Nephrol. 2017;37(3):287-95.

MU-153.indd 998 29-01-2021 15:01:31


Optimizing Renal Care in Elderly CHAPTER 153 999

33. Ahmed FA, Catic AG. Decision-making in geriatric patients with 41. Tiranathanagul K, Katavetin P, Injan P, et al. A novel simple
end-stage renal disease: thinking beyond nephrology. J Clin Med. hemoglobin dilution technique to measure hemodialysis vascular
2019;8(1):5. access flow. Kidney Int. 2008;73(9):1082-6.
34. Vascular Access Work Group. Clinical practice guidelines for vascular 42. Knoll GA. Kidney transplantation in the older adult. Am J Kidney Dis
access. Am J Kidney Dis Off J Natl Kidney Found. 2006;48(Suppl Off J Natl Kidney Found. 2013;61(5):790-7.
1):S248-73.
43. Kaul A, Behera MR, Kishore R, et al. Optimization of treatment
35. Nadeau-Fredette A-C, Goupil R, Montreuil B, et al. Arteriovenous
fistula for the 80 years and older patients on hemodialysis: is it modality in elderly end-stage renal disease population: peritoneal
worth it? Hemodial Int Symp Home Hemodial. 2013;17(4):594-601. dialysis versus transplant. Indian J Nephrol. 2018;28(6):433-40.
36. de Leur K, Oztürk C, Van Zeeland MLP, et al. Vascular access outcome 44. Tonelli M, Riella M. Chronic kidney disease and the aging
in the elderly dialysis patient in combination with the quality of life. population. Indian J Nephrol. 2014;24(2):71.
Vasc Endovascular Surg. 2013;47(6):444-8. 45. Huang E, Segev DL, Rabb H. Kidney transplantation in the elderly.
37. DeSilva RN, Patibandla BK, Vin Y, et al. Fistula first is not always Semin Nephrol. 2009;29(6):621-35.
the best strategy for the elderly. J Am Soc Nephrol JASN. 46. Silva SM da, Braido NF, Ottaviani AC, et al. Social support of adults
2013;24(8):1297-304. and elderly with chronic kidney disease on dialysis. Rev Lat Am
38. O’Hare AM, Bertenthal D, Walter LC, et al. When to refer patients Enfermagem. 2016;24:2752.
with chronic kidney disease for vascular access surgery: should age
47. Kirchhoff KT, Hammes BJ, Kehl KA, et al. Effect of a disease-specific
be a consideration? Kidney Int. 2007;71(6):555-61.
39. Santoro D, Benedetto F, Mondello P, et al. Vascular access for advance care planning intervention on end-of-life care. J Am
hemodialysis: current perspectives. Int J Nephrol Renov Dis. Geriatr Soc. 2012;60(5):946-50.
2014;7:281. 48. Chandrashekar A, Ramakrishnan S, Rangarajan D. Survival analysis
40. Valliant A, McComb K. Vascular access monitoring and surveillance: of patients on maintenance hemodialysis. Indian J Nephrol.
an update. Adv Chronic Kidney Dis. 2015;22(6):446-52. 2014;24(4):206-13.

MU-153.indd 999 29-01-2021 15:01:31


CHAPTER
Understanding the Role
154 of Vitamin D in Diabetic
Nephropathy
Srinath KM, Spoorthy Raj, Manjunath S Shetty

Abstract 
Diabetes mellitus includes a group of metabolic disorders which share the common phenotype of hyperglycemia. Diabetes
mellitus is the leading cause of end stage renal disease. With the prevalence of diabetes mellitus increasing enormously in
present times, the burden of end stage renal disease caused by diabetes mellitus assumes significance.
Three major pathways are implicated in the development of DKD (Diabetic Kidney Disease) initiated by hyperglycemia.
They are Activation of Protein Kinase C and polyol pathways, formation of advanced glycation end products, and
glomerular hyperfiltration leading to glomerular hypertension.
Vitamin D is known to prevent beta cell apoptosis, increase insulin synthesis and enhance peripheral insulin
sensitivity. Also, it is known to suppress RAS (Renin Angiotensin Aldosterone System), prevent podocyte loss and structural
derangement of slit diaphragm, suppress inflammation and prevent glomerulosclerosis.
Hence, addition of Vitamin D supplements to ACE inhibitors/ARBs is proposed as a novel mechanism to prevent and
halt the progression of Diabetic Kidney disease.

Introduction Over 20% of the newly diagnosed T2DM have


concurrent DKD and a further 20–40% develop diabetic
Diabetes mellitus (DM) poses a major global public health
nephropathy mostly within 10 years of diagnosis.4
problem with ever increasing incidence and prevalence in
Several genetic and environmental factors lead to
recent years.
pathogenesis of DN.
At present, diabetes is the most common cause of end
Hyperglycemic state of diabetes leads to various
stage renal disease (ESRD).
hemodynamic, biochemical, metabolic changes in
Increasing burden of diabetic kidney disease (DKD) is
kidneys including inflammation and oxidative stress.5
secondary to widespread prevalence of diabetes.
Considering the burden imposed by diabetes and
According to International Diabetic Federation, the
DKD, extensive research is done in search of therapeutic
number of diabetics is expected to exceed 435 million by
agent, which can reduce or stop the progression of
2030, with more than 90% having type 2 diabetes.1
diabetic kidney disease. One such novel therapeutic agent
The increased risk of all cause and cardiovascular
for diabetic nephropathy is vitamin D and its analogues.
mortality in patients with diabetes is due to the presence
of DKD.2
Approximately, 25–40% of type 1 DM patients and Risk Factors Associated with DKD
5–40% of patients with type 2 diabetes develop this Multiple factors are responsible for the development of
microvascular complication.3 diabetic nephropathy and its progression.

MU-154.indd 1000 29-01-2021 15:01:25


Understanding the Role of Vitamin D in Diabetic Nephropathy CHAPTER 154 1001

These factors can be categorized as susceptibility Pathogenesis of Diabetic Nephropathy


factors—which predispose patients to the risk of developing
Hyperglycemia is the necessary factor in the initiation
DKD and are considered as non-modifiable factors. Acute
of renal injury. Abnormal intracellular metabolism,
kidney injury (AKI), dietary factors, hyperglycemia, and
involving three major pathways that are associated with
hypertension which initiate kidney damage and worsen
the development of diabetic nephropathy shown in
the DKD are considered as modifiable factors.6
Flowchart 1.9
Hence, the risk factors for DKD are summarized as: „„ Activation of protein kinase C and polyol pathways
„„ Susceptibility factors: Old age, male sex, African- „„ Advanced glycation end-products formation

American race, and significant family history „„ Glomerular hyperfiltration leading to intra-glomerular
„„ Initiation factors: Hyperglycemia and AKI
hypertension
„„ Progression factors: Hypertension, obesity, dietary
It is also implicated that there is an excessive
factors, and smoking7 production of mitochondrial reactive oxygen species.
Among the various risk factors hyperglycemia and Interactions between metabolic changes induced by
hypertension are the most predominant risk factors. hyperglycemia with hemodynamic factors, including
vasoactive hormones such as angiotensin II, play a critical
Natural History of DKD8 role in inducing renal injury. These mechanisms result
The progressive model of the natural history of DKD has in cell injury and activate inflammatory cascade which
been shown in Figure 1. further perpetuates cell injury and results in vicious

Fig. 1: The progressive model of the natural history of diabetic kidney disease. Duration of diabetes, in years, is presented on the horizontal
axis. Timeline is well characterized for type 1 diabetes mellitus; for type 2 diabetes mellitus, timeline may depart from the illustration due to
the variable timing of the onset of hyperglycemia

MU-154.indd 1001 29-01-2021 15:01:26


1002 SECTION 11 Nephrology

Flowchart 1: Pathophysiology of diabetic nephropathy

Source: Wada J, Makino H. Inflammation and the pathogenesis of diabetic nephropathy. Clinical science. 2013 Feb 1;124(3):139-52.

cycle of cell injury—inflammation—further cell injury— „„ TGF-β1 (transforming growth factor-β1) is a well
fibrosis.9 determined molecule for the accumulation of ECM
The molecules responsible for inflammation in glycoproteins and subsequent glomerulosclerosis10
diabetic nephropathy include:
„„ Transcription factors, NF-κB Pathology of Diabetic Nephropathy
„„ Proinflammatory cytokines & signaling molecules— Diabetic nephropathy was first described as glomerulo­
IL-6, IL-18, IL-1, TNF, JAK2, & STAT 1&3 pathy, mainly affecting mesangial cells; however, with
„„ Chemokines CCL2 (MCP-1) and CCR2, CXCL12 further research it is found that glomerular epithelial cell
(stromal-cell-derived factor-1), CX3CL1 abnormalities like podocyte dysfunction (angiotensin
„„ Fractalkine and CX3CR1 II mediated reduced expression of Nephrin), apoptosis,
„„ Adhesion molecules: Intercellular adhesion molecule ultimately resulting in depletion of podocytes is central to
1 (ICAM1), Vascular cell adhesion protein 1 (VCAM1), development of proteinuria which is a hallmark feature of
E-selectin (SELE); Toll like receptors—(TLR2, TLR4) diabetic nephropathy. Also identified are the important
„„ Adipokines: Adiponectin, Leptin; Nuclear receptors— changes in other sites like tubules, interstitium, medulla,
VDR, NR1H4 (FXR) PPARα PPARγ, PPARδ and papilla. Renal function and prognosis correlate better

MU-154.indd 1002 29-01-2021 15:01:27


Understanding the Role of Vitamin D in Diabetic Nephropathy CHAPTER 154 1003

with structural lesions in tubules and cortical interstitium


than with classic glomerular changes.11
The steps resulting in diabetic nephropathy are:12
„„ Hypertrophy of glomerulus and hyperfiltration

„„ Glomerular and tubulo-interstitial inflammation

„„ Decrease in number of cells by apoptosis and

accumulation of ECM.

Vitamin D
Fat soluble vitamin D exists in two major forms, namely
ergocalciferol (D2) and cholecalciferol (D3).
The main source of vitamin D for human body is Fig. 2: Role of vitamin D in hepatic glucose homeostasis
endogenously biosynthesized in the skin. Vitamin D
absorbed from diet and synthesized endogenously is
biologically inert. Vitamin D in liver is converted into „„ Calcitriol in liver is known to induce calcium flux
25-hydroxyvitamin D (25(OH) D3) by liver enzyme and activate calcium/calmodulin dependent protein
25-hydroxylase, which can be measured clinically. 25(OH) kinase (CaMkkB), which further activates 5’ AMP
D3 is further hydroxylated to 1, 25(OH)2 D3 by kidney- (Adenosine monophosphate) activated protein
derived 1-alpha-hydroxylase which is an active form of kinase (AMPK). This results in down regulation of
Vitamin D. Phosphoenol pyruvate carboxy kinase (PEPCK) and
The activated form of vitamin D (1,25(OH)2D3) further Glucose 6 phosphatase (G6Pase) and thus reduce
binds with vitamin D receptors (VDRs) and activates hepatic glucose output. AMPKa activation can also
various transcription factors. It is now found that, VDRs downregulate sterol regulatory element binding
and 1-alpha-hydroxylase is expressed in tissues like protein 1 (SREBP1c),FAS and Acetyl co-A carboxylase
islets of pancreas, hepatocytes, vascular smooth muscle (ACC) which further reduces the hepatic triglyceride
cells, macrophages, mesangial cells, and podocytes. content shown in Figure 2.
Vitamin D acts in autocrine manner and exerts multiple „„ Increased intracellular ionized calcium and subsequent

non-calcemic effects through VDRs. This includes downstream signaling mediated by vitamin D is known
vascular, immunomodulatory, anti-inflammatory effects, to enhance glucose and arginine mediated insulin
suppression of RAS, and control of glucose homeostasis.13 release from β-cells of pancreas.19
„„ Suppress the transcription of renin, angiotensin

Role of Vitamin D in Insulin Synthesis and receptors and serve to inhibit local RAS mediated beta
Regulating Insulin Sensitivity cell injury.
„„ Hyperglycemia and increased free fatty acids result
Hyperglycemia results in endoplasmic reticulum stress,
inflammation (lipotoxicity) and loss of insulin sensitivity. in endoplasmic reticulum stress and reduced activity
Thus, hyperglycemia plays a key role in causation of of Akt pathway, which ultimately results in β-cell
resistance to insulin and β-cell failure.14-16 Diet induced apoptosis.
hypovitaminosis D in animal mice models revealed this   Vitamin D helps in beta cell survival and also
hypovitaminosis D is a result of impaired glucose tolerance, increases compensatory beta cell growth in insulin
decreased islet function related gene transcription, and resistant states by enhancing activity of Akt pathway.20
increased RAS expression.17 „„ Vitamin D also reduces accumulation of hepatic

The postulated molecular mechanism behind triglyceride and glucose output through Ca2+/CaMKK/
regulation of insulin homeostasis by Vitamin D includes: AMPK signaling activation.
„„ Promoter regions of insulin receptor genes have VDR „„ Vitamin D also inhibits VDR mediated PPAR activity

response elements (VDREs).18 thus inhibits adipogenesis.21

MU-154.indd 1003 29-01-2021 15:01:28


1004 SECTION 11 Nephrology

Vitamin D Role in Inflammation Evidence from Clinical Studies


Activation of NF-kB pathway results in increased Clinical trial evidence indicates that combined
transcription and production of proinflammatory treatment with losartan and paricalcitol completely
cytokines like TNF-α, IL-1, and IL-6. This proinflammatory prevented albuminuria as well as markedly reduced
milieu leads to leukocyte infiltration of pancreas resulting glomerulosclerosis while restoring glomerular filtration
in reduced β-cell mass, reduced insulin synthesis, islet barrier structure. 2 The vitamin D receptor activator
amyloid deposition, altered downstream insulin signaling (paricalcitol) in albuminuria lowering (VITAL) study
through IRS/AKT/PI3K resulting in insulin resistance and showed that addition of two micrograms of paricalcitol
also affects the maintenance of energy and blood sugar to RAS inhibitors safely lowers albuminuria in patients
balance in hypothalamus.22 with diabetic nephropathy and can be a novel approach
VDD promotes inflammation through NF-kB to reduce residual renal risk in patients with diabetic
pathway as evidenced by animal model, whereas vitamin nephropathy who are already on treatment with optimal
D supplementation has shown beneficial effects in doses of RAS inhibitors.24
preventing the same.22
Conclusion
Vitamin D Role in Diabetic Nephropathy Diabetic nephropathy is a common renal complication of DM
Diabetic nephropathy is associated with increased and a major cause of ESRD. RAS is the predominant mediator of
incidence of vitamin D deficiency. Probable explanation progressive injury to renal system in DN. Hence, RAS inhibitors
are being used as the mainstay of treatment for DN; however,
for the same includes reduced sunlight exposure (e.g., in
RAS inhibition leads to compensatory rise in the renin due to
elderly, sick, dark-skinned people, people wearing veil,
the disruption of renin feedback inhibition. Vitamin D plays a
losses of vitamin D-binding protein in proteinuric states). renal protective role in DM by suppressing rennin expression
Vitamin D is known to ameliorate the harmful effects by negative regulation of RAS. Combination therapy with RAS
of hyperglycemia on kidney by following mechanism:23 inhibitors and active vitamin D and its analogue markedly
„„ Vitamin D inhibits RAS by downregulating renin, ameliorates renal injury.
angiotensinogen and angiotensin II receptor However, Vitamin D is currently only recommended in
expression. treatment of patients with moderate CKD associated with
„„ Enhances the expression of nephrin and prevents
secondary hyperparathyroidism and vitamin D insufficiency.
Hence, further study is required to answer many questions and
structural derangement of slit diaphragm.
thereby uncover the potential renoprotective role of vitamin D
„„ Inhibits activation of ERKs, p38-MAPK, Wnt-b catenin
and its analogues in treatment of patients with CKD.
pathway, and down regulates proapoptotic signals
Bad, Bak, and upregulate anti-apoptotic signal Bcl2
and prevent podocyte apoptosis and podocyte loss. References
„„ Inhibits the expression of Smad 3 protein or by the 1. International Diabetes Federation: Diabetes Atlas, 7th edition.
physical interaction of VDR with Smad 3 protein Brussels, Belgium: IDF Executive Office; 2015. p. 7.
downregulates the intracellular level of Smad 3 and 2. Afkarian M, Sachs MC, Kestenbaum B, et al. Kidney disease and
increased mortality risk in type 2 diabetes. J Am Soc Nephrol.
thus, reduce the expression of TGF-β, and prevent
2013;24(2):302-8.
glomerulosclerosis. 3. Parving HH, Lewis JB, Ravid M, et al. Prevalence and risk factors for
„„ Inhibits the expression of fibronectin and extracellular
microalbuminuria in a referred cohort of type II diabetic patients: a
matrix proteins from mesangial cells and protect global perspective. Kidney Int. 2006;69(11):2057-63.
against glomerulosclerosis. 4. Gatwood J, Chisholm-Burns M, Davis R, et al. Evidence of chronic
„„ Inhibits the expression of MCP-1 (Monoc yte
kidney disease in veterans with incident diabetes mellitus. PloS
One. 2018;13(2):0192712.
Chemotactic Protein 1) which results in decreased 5. Chokhandre MK, Mahmoud MI, Hakami T, et al. Vitamin D & its
leukocyte recruitment to kidneys and reduces the renal analogues in type 2 diabetic nephropathy: a systematic review. J
inflammation. Diabetes Metab Disord. 2015;14:58.

MU-154.indd 1004 29-01-2021 15:01:28


Understanding the Role of Vitamin D in Diabetic Nephropathy CHAPTER 154 1005

6. Taal MW. Risk factors and chronic kidney disease. In: Skorecki K (Ed). 16. Alvarez JA, Ashraf A. Role of vitamin D in insulin secretion and
Brenner and Rector’s The Kidney, 10th edition. Amsterdam: Elsevier; insulin sensitivity for glucose homeostasis. Int J Endocrinol.
2015. pp. 669-92. 2010;2010:351385.
7. Alicic RZ, Rooney MT, Tuttle KR, et al. Diabetic kidney disease: 17. Leung PS. The modulatory action of vitamin D on the renin–
challenges, progress, and possibilities. Clin J Am Soc Nephrol. angiotensin system and the determination of hepatic insulin
2017;12(12):2032-45. resistance. Molecules. 2019;24(13):2479.
8. Diabetes Control and Complications Trial/Epidemiology of Diabetes 18. Wang TT, Tavera-Mendoza LE, Lapierre D, et al. Large-scale in silico
Interventions and Complications Research Group. Retinopathy and and microarray-based identification of direct 1,25-dihydroxyvitamin
nephropathy in patients with type 1 diabetes four years after a trial D3 target genes. Mol Endocrinol. 2005;19(11):2685-95.
of intensive therapy. N Engl J Med. 2000;342(6):381-9. 19. Kampf C, Lau T, Olsson R, et al. Angiotensin II type 1 receptor
9. Wada J, Makino H. Inflammation and the pathogenesis of diabetic inhibition improves the blood perfusion, oxygen tension and first
nephropathy. Clin Sci. 2013;124(3):139-52. phase of glucose-stimulated insulin secretion in re-vascularized
10. Williams Textbook of Endocrinology, 14th edition. 2019. pp. 1481-2. syngeneic mouse islet grafts. Diabetologia. 2005;48(6):1159-67.
11. Makino H, Yamasaki Y, Haramoto T, et al. Ultrastructural changes of 20. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity.
extracellular matrices in diabetic nephropathy revealed by high Annu Rev Immunol. 2011;29:415-45.
resolution scanning and immunoelectron microscopy. Lab Invest. 21. Zhang Y, Deb DK, Kong J, et al. Long-term therapeutic effect of
1993;68(1):45-55. vitamin D analog doxercalciferol on diabetic nephropathy: strong
12. Leung PS. The potential protective action of vitamin D in hepatic synergism with AT1 receptor antagonist. Am J Physiology-Renal
insulin resistance and pancreatic islet dysfunction in type 2 Physiol. 2009;297(3):791-801.
diabetes mellitus. Nutrients. 2016;8(3):147. 22. Holick MF. Sunlight, UV-radiation, vitamin D and skin cancer. Adv
13. Kumashiro N, Erion DM, Zhang D, et al. Cellular mechanism of Exp Med Biol. 2008;624:1-15.
insulin resistance in nonalcoholic fatty liver disease. Proc Natl Acad 23. Ahmed OM, Ali TM, Abdel Gaid MA, et al. Effects of enalapril
Sci USA. 2011;108(39):16381-5. and paricalcitol treatment on diabetic nephropathy and renal
14. Flamment M, Hajduch E, Ferré P, et al. New insights into ER expressions of TNF-α, p53, caspase-3 and Bcl-2 in STZ-induced
stress-induced insulin resistance. Trends Endocrinol Metab. diabetic rats. PloS One. 2019;14(9):0214349.
2012;23(8):381-90. 24. De Zeeuw D, Agarwal R, Amdahl M, et al. Selective vitamin D
15. Del Prato S. Role of glucotoxicity and lipotoxicity in the receptor activation with paricalcitol for reduction of albuminuria
pathophysiology of type 2 diabetes mellitus and emerging in patients with type 2 diabetes ( VITAL study): a randomised
treatment strategies. Diabet Med. 2009;26(12):1185-92. controlled trial. Lancet. 2010;376(9752):1543-51.

MU-154.indd 1005 29-01-2021 15:01:28


MU-154.indd 1006 29-01-2021 15:01:28

You might also like