You are on page 1of 18

REVIEWS

Present and future drug treatments


for chronic kidney diseases:
evolving targets in renoprotection
Norberto Perico, Ariela Benigni and Giuseppe Remuzzi
Abstract | At present, there are no specific cures for most of the acquired chronic kidney
diseases, and renal transplantation is limited by organ shortage, therefore present efforts
are concentrated on the prevention of progression of renal diseases. There is robust
experimental and clinical evidence that progression of chronic nephropathies is
multifactorial; however, intraglomerular haemodynamic changes and proteinuria play a
key role in this process. With a focus on renoprotection, we first examine more established
therapies — such as those that modulate the renin–angiotensin–aldosterone system —
that can be used for the treatment of proteinuric renal diseases. We then discuss
examples of novel drugs and biologics that might be used to target the inflammatory and
profibrotic process, and glomerular injury, highlighting results from recent clinical trials.

Chronic kidney disease (CKD) is a worldwide threat to CKD encompasses different types of renal disease,
public health, but the scale of the problem is probably not and the biggest group includes glomerular diseases of
fully appreciated. Estimates of the global burden of dis- which diabetic and hypertensive nephropathies are the
eases report that diseases of the kidney and urinary tract most common cause of CKD. They are considered to
contribute to approximately 830,000 deaths annually1. be degenerative renal diseases. There are also inflam-
CKD that requires renal-replacement therapy (RRT), matory glomerular diseases, the prototype being lupus
which consists primarily of kidney transplantation, nephritis. Other renal diseases affect primarily the
haemodialysis and peritoneal dialysis, are rising sharply tubulointerstitium and they include polycystic kidney
worldwide so that the global end-stage renal disease disease.
(ESRD) population will exceed 2 million patients by the At present, there is no specific cure for most of the
year 2010 (REF.1). The aggregate cost for treatment during acquired CKDs, nor is there any reasonable expectation
the coming decade will be more than US$1 trillion1. that gene therapy will be available soon enough to treat
Patients on RRT can be regarded as the tip of an iceberg, genetic forms of kidney diseases, such as polycystic
as the number of those with CKD not yet in need of RRT kidney disease. Renal transplantation is limited by organ
is much greater. However, only rough estimates exist for shortage3, a worldwide problem that is not likely to be
the prevalence of predialysis CKD. resolved in the near or immediate future. Therefore, the
It is increasingly being recognized that the burden of best therapeutic option at the present time is to concen-
CKD is not limited to its implications on demands for trate our efforts on preventing the progression of renal
Department of Medicine RRT, but has a major impact on the health of the overall diseases.
and Transplantation, population. Indeed, patients with reduced kidney function
Ospedali Riuniti di Bergamo - represent a population not only at risk for progression of Progression of chronic nephropathies
Mario Negri Institute for
kidney disease and for ESRD, but also at even greater risk Certain renal diseases, although rare, have a rapid course
Pharmacological Research,
Via Gavazzeni 11-24125, for cardiovascular diseases2. This suggests that patients that quickly leads to irreversible ESRD. More common
Bergamo, Italy. with CKD who are progressing towards ESRD carry the nephropathies progress less rapidly, but still evolve to
Correspondence to G.R. heaviest burden of cardiovascular disease, and that this ESRD within one to four decades after diagnosis. During
e-mail: frequently leads to death before ESRD is reached. So, the past 20 years, research in animals and humans have
gremuzzi@marionegri.it
doi:10.1038/nrd2685
CKD, through their impact on cardiovascular morbidity, provided significant insights on the mechanisms by
Published online may directly contribute to increasing mortalities, even in which CKD progresses and has indicated preventive
10 October 2008 emerging countries. strategies. Many studies have established that progressive

936 | nOvEMbER 2008 | vOlUME 7 www.nature.com/reviews/drugdisc


REVIEWS

deterioration of renal function is the result of compen- that are responsible for interstitial fibrosis23. Tissue injury
satory glomerular haemodynamic changes in response promotes the generation of reactive oxygen species and
to nephron loss due to various original insults. In the an endoplasmic reticulum stress response24. This leads
widely used experimental model of renal mass reduc- to the oxidative modification of membrane lipids, pro-
tion by partial five-sixths nephrectomy, the remnant teins and DnA that initiates cell-death responses, which
glomeruli undergo hypertrophy and the tone of afferent results in tissue inflammation24. loss of peritubular capil-
arterioles drops by a greater degree than that of efferent laries also reduces oxygen availability. The danger signal
ones. This increases glomerular capillary hydraulic pres- secondary to hypoxia links cell injury to the activation of
sure, leading to more filtrate formed per nephron4. These Toll-like receptors, which further enhances the produc-
changes serve initially to minimize the functional conse- tion of pro-inflammatory cytokines and chemokines and
quences of nephron loss, but ultimately are detrimental, contribute to the local recruitment of macrophages and
causing relentless injury of remaining intact nephrons5. lymphocytes24,25. In turn, they are capable or stimulating
Enhancing intraglomerular capillary pressure and per- the transformation of tubular cells into myofibroblasts.
fusion pressure result in stretching of the three major Modest derangement of these processes often escapes
cell types — endothelial cells, mesangial cells and podo- clinical or laboratory detection because of compensa-
cytes — in the glomerulus, leading to direct cell injury6 tory adaptations by the myriad of unimpaired nephrons.
(FIG. 1). The mechanical cell damage together with con- beyond a certain level of injury, however, compensatory
comitant induction of pro-inflammatory and profibrotic adaptations no longer keep pace with nephron loss. As
phenotypic alterations in glomerular cells promote the a result, glomerular filtration rate declines and organic
development of glomerulosclerosis7–15. Impairment of the nitrogenous wastes accumulate in plasma and other
selective function of the glomerular capillary wall is a body fluids. In line with these experimental observa-
further mechanism by which glomerular hypertension tions is the evidence that in humans with nephropathy,
contributes to glomerular injury, resulting in excessive more severe and persistent proteinuria means more
protein ultrafiltration in animal models16. An excessive rapid progression of disease26. The role of proteinuria as
protein load in podocytes is a factor underlying pro- a strong, independent predictor of ESRD has also been
gressive injury of these glomerular cells. These injured documented in a mass-screening setting27.
glomerular cells release transforming growth factor-β In summary, there is robust experimental and clini-
(TGF-β) in the microenvironment, which ultimately cal evidence that progression of chronic nephropathies
promotes activation of adjacent mesangial cells and is multifactorial, but intraglomerular haemodynamic
deposition of extracellular matrix17 (FIG. 1). changes and proteinuria have a key role in this process.
A key player of these glomerular haemodynamic
changes that are central to progressive renal injury is Renin–angiotensin–aldosterone system blockade
angiotensin II18. In vitro experimental studies have also Although traditionally the renin–angiotensin–aldosterone
revealed several non-haemodynamic effects of angio- system (RAAS) was considered a systemic endocrine
tensin II that are relevant in renal disease progression. system, an intrarenal local RAAS has also been described
They include stimulation of mesangial cell proliferation to operate independently. The overview of the RAAS
Mesangial cell
A key cell of the glomerulus and extracellular matrix deposition19, and production cascade is reported in Supplementary information S1
with enlarged cytoplasmic of plasminogen activator inhibitor, type 1 (PAI1) by (box).
processes that extend around endothelial cells20. Moreover, angiotensin II alters size- RAAS can be inhibited at various points of the cas-
the capillary lumen and selective properties of the glomerular capillary barrier by cade. Three strategies have already been developed by
insinuate themselves between
the glomerular basement
binding to angiotensin II subtype 1 receptors on podo- pharmaceutical companies: inhibition of angiotensin-
membrane and the overlying cyte foot processes. This leads to rearrangement of the converting enzyme (ACE), competitive inhibition of
endothelium. cell cytoskeleton and in the distribution or expression the binding of angiotensin II to cell-surface receptors
of protein components of the slit diaphragm, which even- and inhibition of the enzymatic action of renin. Most
Podocytes
tually allows excess plasma proteins to move into the trials with these drugs have been performed or are being
The visceral epithelial cells
of the glomerulus with large urinary space21 (FIG. 1). performed on diabetic nephropathy. ACE inhibitors and,
cytoplasmic processes that Exposure of proximal tubuli to the excessive filtered increasingly, angiotensin II receptor antagonists are now
extend from the main body. proteins leads to further intrinsic toxicity of this nephron widely prescribed for the treatment of proteinuric renal
segment17. Thus, both in vitro and in vivo protein overload diseases, including diabetic nephropathy.
Glomerulosclerosis
Mesangial accumulation
causes increased production of vasoactive, inflammatory
of hyaline material that mediators and growth factors such as endothelin 1 (ET1), ACE inhibitors and angiotensin II receptor blockers
progressively encroaches monocyte chemoattractant protein 1 (MCP1), RAnTES or their combination for renoprotection. In five-sixths
on capillary lumina, which and osteopontin. Transcription analysis by cDnA micro- nephrectomized rats, ACE inhibitors or angiotensin II
obliterates the Bowman’s
array of renal tubular cells from patients with proteinuric receptor antagonists slowed the development of protein-
space and results in global
sclerosis of the glomerulus. nephropathies22 identified more than 160 genes as being uria and limited renal structural damage compared with
regulated differently compared with those of proximal conventional antihypertensive drugs16. As a consequence
Slit diaphragm tubular cells from control subjects. of these effects, progressive loss of kidney function was
A thin membrane that bridges The activation of various molecules, including cyto- attenuated16. In this nephrectomy model, as in other
the gap between adjacent foot
processes of the podocytes
kines, growth factors and vasoactive substances, results in models of diabetic and non-diabetic renal disease, ACE
near the glomerular basement abnormal accumulation in the interstitium of extracell- inhibitors and angiotensin II receptor antagonists have
membrane. ular matrix collagen, fibronectin and other components the unique property of normalizing intraglomerular

nATURE REvIEwS | drug discovery vOlUME 7 | nOvEMbER 2008 | 937


REVIEWS

Degenerative/ Proliferation/
inflammatory extracellular Glomerulosclerosis
glomerular diseases matrix deposition

Nephron loss

Mesangial cells
Compensatory
Mechanical
glomerular capillary Endothelial cells
stretching
hypertension
Podocytes ↑Ang II synthesis

Glomerular Increased glomerular


hyperfiltration permeability Podocyte protein Gene
accumulation activation ↑TGF-β
to proteins

Cytoskeleton rearrangement/
Proteinuria loss of differentiated phenotype

Slit-diaphragm Foot-process
dysfunction effacement

Figure 1 | Mechanisms of progressive glomerular injury. A reduction in the number of nephrons as a consequence
of various glomerular diseases results in compensatory glomerular haemodynamic changes that are ultimately
detrimental. In particular, by mechanical stretching, the increased glomerular capillaryNature
pressure directly
Reviews | Druginjures
Discovery
glomerular cells — namely endothelial cells, mesangial cells and podocytes. Glomerular hypertension also impairs the
glomerular capillary size-permeable-selective function, which causes excessive protein ultrafiltration and eventually
proteinuria. The concomitant protein overload of podocytes leads to rearrangement of the cell cytoskeleton and loss
of differentiated phenotype. This eventually contributes to the structural and functional changes of foot processes and
slit diaphragm of podocytes, which further enhances protein traffic through the glomerular capillary wall. Injured
podocytes release transforming growth factor-β (TGF-β) in the microenvironment, which activates adjacent mesangial
cells to proliferate and produce excessive extracellular matrix components, and thus contributes to the development
of glomerulosclerosis. A key player in these haemodynamic and non-haemodynamic events of progressive glomerular
injury is angiotensin II (Ang II).

capillary pressure and size selectivity of the glomerular with30,31. Subsequent meta-analysis confirmed that pro-
capillary barrier, which limits protein traffic and conse- teinuria strongly correlated with renal disease progres-
quent nephrotoxicity28. sion, and treatments that fail to reduce proteinuria are
before 1995, several randomized trials evaluating the unlikely to exert significant renoprotection32,33. However,
effect of ACE inhibitors in humans had conflicting results, proteinuria is currently not recognized by the US Food
possibly because of the heterogeneity of the patients and and Drug Administration (FDA) as an outcome for CKD
treatments studied. More importantly, most studies treatment. nevertheless, in a recent meeting, discussion
included patients with no or mild proteinuria, which was has been initiated between leading nephrologists and
associated with slow disease progression and negligible FDA representatives for qualification of proteinuria as
treatment effect. This, combined with the small sample a surrogate marker in CKD (see Further information).
size, reduced the power of the analyses and increased In chronic nephropathies, high blood pressure is also a
the risk of random data fluctuations29. More convincing major determinant of disease progression, and blood-
evidence of a specific renoprotective and dialysis-saving pressure reduction is renoprotective34. However, it is
potential of ACE inhibitors and angiotensin II receptor controversial whether ACE inhibition therapy in combi-
antagonists was then provided by a series of large clinical nation with efforts to reduce blood pressure — to lower
trials in patients with diabetes and in patients without dia- than usual targets — in patients with CKD will provide
betes (TABLE 1). The REIn study30 showed that in patients additional renoprotection. This issue has been addressed
with non-diabetic nephropathy an ACE inhibitor (rami- by the REIn-2 trial, which showed that in patients with
pril) slowed the rate of decline in glomerular filtration non-diabetic proteinuric nephropathies receiving back-
rate and halved the risk of ESRD at equivalent levels of ground ACE-inhibitor therapy, no additional benefit from
blood-pressure control in comparison with conventional further blood-pressure reduction by the dihydropyridine
antihypertensive therapy. Treatment effect was a func- calcium-channel blocker felodipine was achieved in terms
tion of baseline proteinuria, and the highest benefit was of delaying decline in glomerular filtration rate and pro-
observed in those with most severe proteinuria to start gression to ESRD35.

938 | nOvEMbER 2008 | vOlUME 7 www.nature.com/reviews/drugdisc


REVIEWS

ACE inhibitors are also renoprotective in patients with effective inhibition of the RAAS. Finding that combined
type 1 diabetes. They reduced the incidence of ESRD as therapy also ameliorated concomitant cardiovascular
well as progression to overt nephropathy in those with risk factors such as hypertension and dyslipidaemia
macroalbuminuria36. Relatively small studies have also suggests that dual RAAS blockade could be the treatment
found that ACE inhibitors are renoprotective in type 2 of choice to limit renal and cardiovascular morbidity and
diabetes37, but there has been no adequately powered mortality in this population. Along this line, post-hoc
trial conducted yet that evaluates their effect on renal analyses of the OnTARGET trial45 will be important to
function decline or progression to ESRD. nevertheless, assess whether dual RAAS blockade may reduce renal
in the bEnEDICT trial38, evaluation of 1,204 patients with and cardiovascular events more effectively than an ACE
type 2 diabetes and hypertension (but no evidence of renal inhibitor or an angiotensin II receptor antagonist alone
involvement) has shown that ACE inhibitors may prevent in patients with renal dysfunction at inclusion.
the onset of nephropathy, which is the strongest risk fac-
tor for cardiovascular mortality in this population. More Renin inhibitors. Monotherapy with ACE inhibitors or
recently, the IRMA-2 (REF. 39), REnAAl40 and IDnT41 angiotensin II receptor antagonists provides incomplete
trials have provided evidence that angiotensin II receptor inhibition of the RAAS, particularly after long-term use
antagonists are also renoprotective, independently of their in which a reactive rise in plasma renin activity may
antihypertensive effect, in patients with type 2 diabetes limit efficacy. So, targeting renin, the rate-limiting step
and incipient or overt nephropathy (TABLE 1). in angiotensin II synthesis, may afford better blockade
nevertheless, more effective intervention strategies of the RAAS. The first renin inhibitors were peptide ana-
are needed to limit the still excessive renal and cardio- logues of the prosegment of renin or substrate inhibitory
vascular risk of people with diabetes. In this regard, it is peptides of the renin cleavage site of angiotensinogen46.
important to note that angiotensin II can be also gener- They effectively inhibited renin activity in animals and
ated by non-ACE pathways, such as chymase, that are humans46, but their limitation was that they had to be
not inhibited by ACE inhibitors (see Supplementary administered parenterally. This pitfall was overcome by
information S1 (box))42. The combination of an ACE novel orally active renin inhibitors (see Supplementary
inhibitor and an angiotensin II receptor antagonist has information S1 (box)). The first generation of orally
been suggested as a way to maximize RAAS blockade by active renin inhibitors — with a molecular mass of a
affecting both the bioavailability and activity of angio- tetrapeptide — were never used clinically because of the
tensin II. Moreover, an ACE inhibitor might block the short half-life, bioavailability of less than 2% and mild
compensatory increase in angiotensin II synthesis that blood-pressure-lowering effect47. The discovery of the
is observed during angiotensin II receptor antagonist new orally active renin inhibitor aliskiren (Tekturna;
therapy, and these antagonists might block the activity of novartis), which has a prolonged half-life and high bio-
angiotensin II produced by ACE-independent pathways. availability, in addition to the high specificity for human
So, dual RAAS blockade is probably a rational approach renin48, renewed interest in this class of antihypertensive
to achieve a more effective inhibition of the system and, drugs. Aliskiren directly blocks the function of renin by
conceivably, maximize renoprotection and cardiopro- occupying the enzymatic pocket of the molecule, thereby
tection in patients at risk. providing greater inactivation of the RAAS and possibly
Animal studies and clinical studies have consistently more renoprotective benefit than other RAAS blockers.
found that ACE inhibitors and angiotensin II receptor This possibility is supported by findings in hyper-
antagonists in combination reduce proteinuria more tensive double transgenic rats harbouring the human
effectively than either agent alone43. However, in the renin and angiotensinogen genes: aliskiren lowered
clinical studies, the combined therapy included an ACE blood pressure and albuminuria and normalized serum
inhibitor and an angiotensin II receptor antagonist at full creatinine49. Clinical trials in hypertensive patients
recommended doses, and the dual RAAS blockade had a have demonstrated the efficacy of aliskiren in lower-
more robust antihypertensive effect than monotherapy43. ing arterial blood pressure at comparable or at an even
The full dosages of ACE inhibitor and angiotensin II higher extent than angiotensin II receptor antagonists50.
receptor antagonist used in these studies did not allow Clinical effects of oral renin inhibitors on kidney dam-
the assessment of whether the superior antiproteinuric age remain to be determined. The only large clinical trial
effect of combined therapy really reflected more effective of aliskiren in patients with CKD (the AvOID study)
RAAS blockade or, rather, was just a function of a greater has just been completed51. This study was performed in
reduction in blood pressure. This issue was addressed in patients with type 2 diabetes who also have hyperten-
a cohort of patients with non-diabetic CKD given full sion and overt nephropathy. Participants were assigned
doses of the ACE inhibitor benazepril or of the angio- to receive aliskiren or placebo in addition to the maxi-
tensin II receptor antagonist valsartan, or half doses of mum recommended renoprotective dose of losartan
Albumnin/creatinine ratio the two drugs used in combination44. A similar reduc- (Cozaar; Merck). Aliskiren reduced the albumin/creatinine
Ratio between the tion in blood pressure was seen in the three treatment ratio by 20% compared with placebo during 24 weeks of
concentrations of albumin groups, whereas a more consistent proteinuria reduction follow-up51. These promising findings, however, should
and creatinine measured in was seen in those on dual therapy. Thus, the superior be tempered by the fact that the AvOID study has been
urine. It is used to detect
early chronic nephropathy,
antiproteinuric effect of dual compared with single relatively short-term and lacks relevant information
particularly diabetic drug RAAS blockade was not explained by a superior regarding long-term outcomes such as the progression
nephropathy. antihypertensive effect, but in all likelihood by a more of kidney disease to ESRD.

nATURE REvIEwS | drug discovery vOlUME 7 | nOvEMbER 2008 | 939


REVIEWS

Table 1 | Renoprotection by angiotensin II blockers in large cohorts of patients with nephropathy


Trial Patients Treatment (median end point/outcome Proteinuria
follow-up) changes
Non-diabetic nephropathies
AIPRI 583 normotensive Double-blind placebo controlled Doubling of serum Benazepril was
(REF. 207) or hypertensive trial with benazepril (3 years) creatinine or need for –29%, placebo
patients with renal dialysis: benazepril was +9%
insufficiency due to was 10.3%, placebo was
various disorders 20.1%
REIN 352 normotensive or Double-blind placebo controlled Doubling of serum Ramipril was
(REF. 30) hypertensive patients trial with ramipril; both plus creatinine or end-stage –50%, placebo
with non-diabetic conventional antihypertensive renal disease: ramipril had no change
proteinuric therapy (3.4 years) was 23.1%, placebo was
nephropathies 45.4%
REIN-2 338 patients Randomized controlled trial Time to end-stage renal Similar in both
(REF. 35) (non-diabetic) comparing conventional BP disease: conventional BP groups
with proteinuric control plus ramipril with control was 20%, intensive
nephropathies intensified BP control (ramipril BP control was 23%
plus felodipine) (3 years)
Diabetic nephropathies
RENAAL 1,513 patients with Double-blind placebo controlled End-stage renal disease: Losartan was
(REF. 40) type 2 diabetes and trial with losartan; both plus losartan was 19.6%, –35%, placebo
overt nephropathy conventional antihypertensive placebo was 25.5% there was
therapy (3.4 years) a trend to
increase
IDNT 1,715 hypertensive Double-blind placebo controlled Doubling of serum Irbesartan
(REF. 41) patients with type 2 trial comparing irbesartan with creatinine, onset of was –33%,
diabetes and overt amlodipine; all plus conventional end-stage renal disease amlodipine was
nephropathy antihypertensive therapy or death: irbesartan was –6%, placebo
(2.6 years) 32.6%, amlodipine was was –10%
41.1%, placebo was 39%
IRMA-2 590 hypertensive Double-blind placebo controlled Time to onset of Not measured
(REF. 39) patients with type trial comparing irbesartan macroalbuminuria:
2 diabetes and 300 mg with irbesartan 150 irbesartan 300 mg was
microalbuminuria mg; both plus conventional 5.2%, irbesartan 150 mg
antihypertensive therapy was 9.7%, placebo was
(3.4 years) 14.9%
BENEDICT 1,204 hypertensive Double-blind placebo controlled Development of persistent Not measured
(REF. 38) patients with type trial comparing trandolapril with microalbuminuria:
2 diabetes and trandolapril/verapamil, and with trandolapril was 6.0%,
normoalbuminuria verapamil; all plus conventional trandolapril/verapamil
antihypertensive therapy was 5.7%, verapamil was
(3.6 years) 11.9%, placebo was 10.0%
Type 1 409 patients with Double-blind placebo Doubling of serum Significantly
diabetes type 1 diabetes and controlled trial with captopril creatinine: captopril less proteinuria
mellitus nephropathy 300 mg; both plus conventional was 12.1%, placebo was in the captopril
(REF. 36) antihypertensive therapy 21.2% group than in
(2.7 years) the placebo
group
ACE, angiotensin converting enzyme; AIPRI, ACE inhibition in progressive renal insufficiency; BENEDICT, Bergamo nephrologic
diabetes complications trial; BP, blood pressure; IDNT, irbesartan diabetic nephropathy trial; IRMA-2, irbesartan in patients with
type 2 diabetes and microalbuminuria; REIN, ramipril efficacy in nephropathy; REIN-2, BP control for renoprotection in patients
with non-diabetic chronic renal disease; RENAAL, reduction of end points in non-insulin-dependent diabetes mellitus with the
angiotensin II antagonist losartan.

Aldosterone antagonists. An additional key component of potent in blocking mineralocorticoid receptors than
the RAAS is aldosterone — a mineralocorticoid hormone spironolactone. However, eplerenone is substantially
that is produced in the adrenal cortex, and in endothelial more selective than spironolactone and has little ago-
and vascular smooth muscle cells in the heart, blood nist activity for the oestrogen receptor and the proges-
vessels and brain. Spironolactone and eplerenone are terone receptor52. Therefore, eplerenone is associated
steroid analogues that strongly resemble aldosterone with a lower incidence of side effects. At variance with
structurally, thereby functioning as competitive antago- spironolactone, eplerenone has no active metabolites,
nists of the mineralocorticoid receptor52. Eplerenone is which results in a shorter effective half-life and therefore
a 9α,11α epoxy derivative of spironolactone and is less a more rapid time to peak response. Selective aldosterone

940 | nOvEMbER 2008 | vOlUME 7 www.nature.com/reviews/drugdisc


REVIEWS

receptor antagonism may have benefit for the kidney proteinuria63. by contrast, in patients with idiopathic
independent of its effects on blood pressure. Indeed, membranous nephropathy and nephrotic proteinuria ,
in recent years there has been a striking paradigm shift spironolactone given with full-dose ACE inhibitor
with respect to our understanding of the widespread therapy did not provide additional antiproteinuric
effects of aldosterone that are in addition to the classical effects (G. Remuzzi and P. Ruggenenti, unpublished
effects on sodium and potassium transport in the renal observations). However, a systematic review of 15 studies
tubules. This points to a possible contribution of this that give an aldosterone blocker in addition to an ACE
RAAS component to the development and progression inhibitor and/or angiotensin II receptor antagonist for
of chronic injury. Recent evidence indicates that in rats, treatment of patients with proteinuric CKD showed
aldosterone can be produced in the renal cortex53, and decreases in urinary protein excretion, but highly vari-
its mineralocorticoid receptor has been localized to pre- able changes in renal function64. The clinically significant
glomerular vasculature, mesangial cells and fibroblasts, heterogeneity of these studies precludes any recommen-
as well as distal tubular cells of the nephron54. Thus, the dation about using sequential blockade of the RAAS with
intrarenal sites of aldosterone production and expression an ACE inhibitor/angiotensin II receptor antagonist and
pattern of its receptor, together with the observation aldosterone antagonist to provide additional long-term
that mediators of renal disease progression promote the benefits to patients with chronic nephropathies. As there
local synthesis of aldosterone, suggest a possible direct is also concern with regards to the risk for developing
role of this mineralocorticoid hormone in kidney dam- hyperkalaemia, future randomized trials are necessary
age. Although the molecular pathways of aldosterone- to confirm the efficacy and safety of aldosterone receptor
mediated renal injury have not been fully elucidated, they antagonism on the progression of CKD.
all seem to contribute to the final common pathway of
renal fibrosis55 (FIG. 2). Anti-inflammatory and antifibrotic therapeutics
As aldosterone acts through production of growth In addition to components of the RAAS, a large variety
factors and reactive oxygen species, and inhibition of of inflammatory cells are recruited in the kidney tissues
extracellular matrix degradation — important mediators and are activated to produce mediators that trigger pro-
of renal fibrosis — blockade of the mineralocorticoid gression of renal disease towards ESRD. Inflammatory
receptor should prevent or limit renal injury. Indeed, it cells — apart from being involved in the development of
has been shown that spironolactone limits glomerulo- glomerular injury — are key players of the tubulointer-
sclerosis and tubulointerstitial fibrosis in rats with stitial damage, which eventually leads to interstitial fibro-
streptozotocin-induced diabetic nephropathy56, in a sis (FIG. 3). Mediators of tubulointerstitial injury would
genetic model of type 2 diabetes57, and in spontan- represent novel potential targets for therapeutics that
eously hypertensive rats of the stroke-prone substrain58. eventually could help to complement the renoprotection
Similarly, in Dahl salt-sensitive rats, eplerenone prevented afforded by RAAS inhibitors, and in the future could
podocyte damage, proteinuria and glomerulosclerosis59. be part of a multidrug approach to provide the ultimate
More importantly, inhibition of the aldosterone recep- treatment for progressive renal disease. In the following,
tor showed renoprotection in animals that have estab- we will examine these new therapeutic options; most of
lished renal injury. In rats with five-sixths nephrectomy, them are still in the preclinical development while a few
spironolactone induced regression of existing proteinuria are now reaching the clinical arena.
and glomerular damage60. Tubulointerstitial fibrosis and
vascular lesions improved in parallel with glomerulo- Endothelin receptor antagonists. The endothelin system
sclerosis. The renoprotective effects of spironolactone comprises a family of three isopeptides that have power-
were potentiated when rats were given the aldosterone ful vasoconstrictor and profibrotic properties of which
receptor antagonist in combination with the angiotensin ET1 is the predominant and biologically more relevant
II receptor antagonist losartan. These experimental isoform in human kidneys65. It is produced by glomerular
findings may have important clinical implications, as endothelial, epithelial and mesangial cells, and by renal
Dahl salt-sensitive rat they indicate that blockade of the aldosterone receptor tubular and medullary collecting duct cells. The renal
An experimental model of is instrumental to protect the kidney from further dam- medulla contains the highest concentration of ET1 of
hypertension and renal injury age even in the presence of an angiotensin II receptor any organ66. The effects of endothelin are mediated by
(glomerulosclerosis and antagonist. whether this novel renoprotective approach two cell-surface receptors belonging to the superfamily
tubulointerstitial damage)
induced in rats sensitive to
would translate into clinical benefits for patients with of rhodopsin-like G-protein-coupled receptors, namely
a high-salt diet. chronic nephropathies remains unclear, as available data the type-A (ETA) and the type-b (ETb) receptors67,68. ETA
are scarce and are mainly from non-controlled or small, receptors reside in vascular smooth-muscle cells and
Idiopathic membranous short-term controlled studies that are focused only on mediate vasoconstriction and cell proliferation69, whereas
nephropathy
the effect on urinary protein excretion. The results show ETb receptors mainly reside on endothelial cells and
The most common cause for
nephrotic syndrome in adults, consistent renoprotection (reduction of albuminuria or their activation results in vasodilation via prostacyclin
which occurs as idiopathic of proteinuria) in patients with type 1 or type 2 diabetes and nitric oxide70. However, ETb receptors on smooth-
(primary) disease of the kidney. and overt nephropathy when aldosterone antagonists are muscle cells can elicit vessel contraction71. besides its
given alone or as add-on therapy to RAAS blockade61,62. vasodilatory effect, the ETb receptor acts as a clearance
Nephrotic proteinuria
Urinary protein excretion of
Even in patients with non-diabetic chronic disease receptor for circulating ET1, and regulates water and
more than 3 g per day in the already treated with ACE inhibitors and/or angiotensin sodium handling. Selective and non-selective endothe-
setting of glomerular disease. II receptor antagonists, spironolactone effectively reduced lin-receptor antagonists have been developed and have

nATURE REvIEwS | drug discovery vOlUME 7 | nOvEMbER 2008 | 941


REVIEWS

showed promising results in animal models of hyperten- combination therapy in patients who do not fully respond
sion, heart failure and renal disease72. In particular, the to ACE inhibitors. The question of whether selective ETA
renoprotective effect of endothelin-receptor blockade or non-selective ETA/ETb receptor antagonists should
has been documented in several models of progressive be used is still a matter of investigation. Although both
nephropathies, including renal mass ablation73,74, acceler- antagonists reduce blood pressure, preliminary evi-
ated passive Heymann nephritis75, streptozotocin-induced dence in patients with CKD showed that selective ETA
diabetes76 and lupus nephritis77. Animal data suggest receptor antagonists have additional desirable effects on
that concomitant blockade of the endothelin system and renal haemodynamics80. Concomitant blockade of ETb
the RAAS produces greater haemodynamic changes78 receptors seems to offer no advantage over selective ETA
and more renoprotective effects75 than those seen with antagonism in terms of preserving natriuresis, diuresis
blockade of either system alone. This synergism is based and glomerular haemodynamics, but studies are needed
on the interaction of endothelin and angiotensin II at to discern the theoretical benefit. Despite the successful
the molecular level79, and would represent the basis for a use of endothelin-receptor antagonists in pulmonary
OH

O O
HO

Chaperone
O
Aldosterone
MR MR

Transcription
MR Na+/K+
ATPase
MR Target gene
Non-genomic K+
transactivation Na+
ATP

EGFR ADP
P ERK1/2 SGK1
phosphorylation

NADPH ENaC
oxidase
Na+
↑ICAM1 ↑Collagen
↑MCP1 ↑TGF-β
↑IL6 ↑ROS ↑PAI1

Inflammation Extracellular ↑Blood


matrix pressure
accumulation

Renal damage

Figure 2 | Mechanisms of aldosterone-induced kidney damage. Activation of the mineralocorticoid receptor (MR) by
aldosterone results in its dissociation from molecular chaperones, translocation into the nucleus and binding to hormone-
Nature Reviews | Drug Discovery
response elements in the regulatory region of target gene promoters to enhance expression. Stimulatory action of
aldosterone on Na+ reabsorption in renal tubular cells depends mostly on transcriptional regulation of Na+/K+-ATPase and
early induction of serum/glucocorticoid regulated kinase 1 (SGK1), which de-represses the surface expression and activity
of the epithelial Na+ channel (ENaC). Aldosterone enhances the gene expression of profibrotic molecules, including
collagen, transforming growth factor-β (TGF-β) and plasminogen activator inhibitor, type 1 (PAI1). Moreover, aldosterone
stimulates inflammation through the generation of reactive oxygen species (ROS) by increasing the expression of
Heymann nephritis
NADPH oxidase. Aldosterone can also exert rapid, non-genomic effects, which involves transactivation of the epithelial
An experimental model of growth-factor receptor (EGFR) and the phosphorylation of extracellular-signal-regulated kinase 1 (ERK1)/ERK2, thereby
glomerular disease that upregulating the expression of pro-inflammatory transcription factors and related molecules. In the kidney, the net effect
mimics membranous of aldosterone-induced inflammation and extracellular matrix accumulation is tissue fibrosis and ultimately renal
nephropathy in humans. scarring. ICAM1, intracellular adhesion molecule 1; IL6, interleukin 6; MCP1, monocyte chemoattractant protein 1.

942 | nOvEMbER 2008 | vOlUME 7 www.nature.com/reviews/drugdisc


REVIEWS

hypertension81, their possible antiproteinuric and reno- and nCT00296400, respectively; see Further infor-
protective effects in patients with CKD remain to be eval- mation) are currently underway to assess the effect of
uated. This could be due to the fact that pharmaceutical rosuvastatin on proteinuria and renal function during
companies are in favour of performing acute/short-term 1 year of follow-up. These trials may provide definitive
clinical studies, such as those in pulmonary hyperten- evidence to the renoprotection and safety of statins.
sion, but are reluctant to embark in trials with long-term nevertheless, more relevant would be trials with
outcomes, such as those required to define the potential clinically important outcomes of progression to ESRD.
renoprotective effects of endothelin-receptor antagonists. Hopefully, the results of the SHARP trial (ClinicalTrials.
Moreover, major side effects of hepatotoxicity and fluid gov database identifier nCT00125593; see Further
retention documented with this drug class raise concern information) that is currently being conducted in 6,000
for their long-term clinical use72. patients with CKD will help to provide this information
in the near future91.
Statins. Statins are inhibitors of the 3-hydroxy-3-methyl-
glutaryl coenzyme A (HMG-CoA) reductase, the rate- Cytokine and chemokine receptor antagonists and
limiting step in cholesterol synthesis — namely the chemokine inhibitors. Members of the tumour necrosis
conversion of HMG-CoA into mevalonate (see Supple- factor (TnF) superfamily of cytokines regulate several
mentary information S2 (box)). Statins therefore effec- cell responses including proliferation, differentiation,
tively reduce serum levels of low-density lipoprotein inflammation and cell death92. Some of these cytokines
cholesterol and, to a lesser extent, they reduce triglycer- such as TnF-α have been extensively studied in kidney
ide levels. However, statins also inhibit the synthesis of a disease and have been shown to mediate renal damage.
range of compounds including the so-called isoprenoids Antibody blockade of TnF-α reduces inflammation and
(farnesyl-PP, geranyl-geranyl-PP) that are derived from scarring in experimental crescentic glomerulonephritis93 and
the mevalonate pathway, which are involved in numer- ameliorates obstruction-induced renal fibrosis and dys-
ous biological processes in all types of cells, including function in rodents94. The recombinant TnF-α receptor
cell proliferation and generation of pro-inflammatory etanercept (Enbrel; Amgen/wyeth) has been used in the
cytokines82 (see Supplementary information S2 (box)). clinic and is highly effective in patients with rheumatoid
Studies in rat models of cyclosporine-induced nephro- arthritis95. However, clinical trials that use TnF-α block-
pathy or unilateral ureteral obstruction have shown that ers in patients with ANCA-associated vasculitis gave mixed
statins do protect from renal scarring83,84. Although the results96. Of note, in a large-scale, randomized trial, treat-
renoprotective effect can be attributed to the lipid-lowering ment with etanercept was found to be ineffective and
property of statins, their anti-inflammatory and antipro- resulted in an excess of treatment-related mortality96.
liferative actions could also have a role in attenuating renal Moreover, autoimmunity that is induced by anti-TnF-α
inflammation, tubular transdifferentiation and interstitial therapy remains an issue of concern in patients with
fibrosis85. Furthermore, in a severe model of progressive AnCA-associated vasculitis as it is in patients with rheu-
nephropathy resistant to ACE inhibition, combining matoid arthritis96. In addition, the recent observation of
ACE inhibitors and angiotensin II receptor antagonists serious infections and malignancies in randomized clini-
with a statin arrested proteinuria and protected against cal trials95, as well as the increased mortality in patients
impairment of renal function and structure86. with chronic heart failure97, give doubt over the possibility
Preliminary clinical data support the renoprotective of designing clinical trials for CKD and renal fibrosis.
effects of statins. A meta-analysis of 15 small, randomized, Several chemokines and their receptors act as pro-
placebo-controlled studies involving 1,384 participants moters of renal fibrosis98. The profibrotic activity of
with CKD found that statins reduced albuminuria and MCP1 and its receptor CCR2 (REFs 99,100), as well as of
proteinuria within a median of 6 months after initia- RAnTES and its receptor CCR1 (REF. 101), is mediated by
tion of therapy87. Individually, these studies, however, the recruitment of inflammatory cells into the glomeruli
showed heterogeneous effects and were often of poor and the tubulointerstitium. Evidence is available dem-
quality. More recently, another meta-analysis on 18,176 onstrating that blockade of CCR2 (REF. 99) or CCR1
patients with low glomerular filtration rate documented chemokine receptors ameliorated progressive fibrosis in
Crescentic that statins modestly reduced proteinuria but did not the kidney as shown in rats with unilateral ureter liga-
glomerulonephritis significantly change the rate of renal function decline tion101 and in mice with glomerulosclerosis and nephrotic
An aggressive form of
glomerulonephritis that
in comparison with placebo88. This conclusion chal- syndrome102. However, the attempt to block chemokines
is characterized by intensive lenges a previous meta-analysis study that showed a and their receptors in humans is more complex. Indeed
glomerular inflammation, small benefit of statins in slowing the rate of decline in translation of experimental findings into clinical trials is
which induces epithelial cell glomerular filtration rate, but a similar effect on pro- difficult owing to the large species differences in the tissue
proliferation and macrophage
teinuria89. Moreover, there are observations that raise expression of chemokine receptors, the heterogeneity of
maturation (cellular crescents).
concern about the safety of statins (particularly rosuva- the response to block them in animal models103 and the
ANCA-associated vasculitis statin) owing to the risk of increasing urinary protein limited specificity in humans of the chemokine recep-
An inflammatory disease excretion90. So, only the results of ongoing large trials in tor antagonists (CCR2 and CCR5 antagonists) that have
involving blood vessels, also of patients with CKD will help to elucidate the real thera- been developed for clinical use so far. A possible prom-
the kidney, characterized by
the concomitant presence of
peutic potential of this class of drugs to kidney disease. ising alternative is the inhibition of MCP1 production
antineutrophil cytoplasmic with this in mind, the PlAnET 1 and PlAnET 2 studies with bindarit (2-methyl-2-[[1-(phenylmethyl)-1H-in-
antibodies (ANCA). (ClinicalTrials.gov database identifier nCT00296374 dazol-3-yl]methoxy]propanoic acid)104. In murine lupus

nATURE REvIEwS | drug discovery vOlUME 7 | nOvEMbER 2008 | 943


REVIEWS

Increased
potent pro-inflammatory mediator that induces cell
Excessive migration, enhances cell adhesion and stimulates the
glomerular
protein tubular
permeability
reabsorption oxidative burst108. So, the prevention of C5 cleavage
to proteins
Proximal tubular cell would have substantial anti-inflammatory consequences
in a relatively late stage of the complement activation
cascade without affecting the activation and function
of the early components. Amelioration of the course of
Nuclear signals for
Cell injury vasoactive and lupus nephritis was documented in mice after treatment
inflammatory genes with a monoclonal antibody that is specific for the C5
component109. The specific blockade of C5 cleavage can
be achieved either by a humanized monoclonal antibody
mRNA: ET1,
(eculizumab) or its 25-kDa single-chain version lack-
RANTES,MCP1, ing the whole constant region including the Fc part of
TGF-β the antibody (pexelizumab). Eculizumab was recently
approved by the FDA and the European Commission —
under the trade name Solaris (Alexion Pharmaceuticals)
Corresponding — for the treatment of patients with paroxysmal nocturnal
proteins
haemoglobinuria based on the demonstration that this
monoclonal antibody inhibited complement-mediated
Interstitium
Lymphocyte/ intravascular haemolysis110. with regards to kidney dis-
macrophage ease, after favourable Phase I safety results in patients
EMT infiltration with systemic lupus erythematosus (SlE), a Phase II trial
Cell
proliferation using eculizumab was initiated in patients with prolifer-
ative lupus nephritis in the United States, but unfor-
Interstitial ↑ECM protein Interstitial TGF-β, EGF, Cytokines/
fibrosis synthesis fibroblasts bFGF, PDGF chemokines tunately was then stopped owing to difficulties in the
recruitment of such patients110. On the other hand, a ran-
↓ECM
degrdation domized controlled trial in 200 patients with idiopathic
Figure 3 | Mechanisms of renal interstitial fibrosis. The injury of tubular membranous nephropathy — a disease in which C5b-9
epithelium as well as the protein overload of proximal tubular cells as a consequence activation of the podocytes have a pathogenetic role, at
of the increased glomerular permeability to proteins in the setting of glomerular least in experimental animals111 — found no difference
Nature
disease, activates intracellular signals that cause increased Reviewsof| Drug
production Discovery
vasoactive, between eculizumab and placebo in the primary out-
inflammatory mediators and growth factors, such as endothelin 1 (ET1), RANTES, come variable of urinary protein excretion112. However,
monocyte chemoattractant protein 1 (MCP1) and transforming growth factor-β since complement activity was not fully inhibited in the
(TGF-β). These substances are released into the interstitium and promote local majority of patients, and because of the short-term treat-
recruitment of inflammatory cells, which in turn are stimulated to release cytokines/ ment, the study may have been insufficient to document
chemokines and growth factors. In particular, TGF-β induces epithelial to a true therapeutic effect of eculizumab.
mesenchymal transition (EMT) and ultimately fibroblast generation. Furthermore,
growth factors stimulate interstitial fibroblasts to proliferate and increase the
production of extracellular matrix (ECM) proteins. These events, combined with the Anti‑TGF‑β antibodies, inhibitors of TGF‑β production
reduction in ECM degradation, eventually lead to interstitial fibrosis and renal and receptor kinases. Transforming growth factor-β
scarring. EGF, epidermal growth factor; bFGF, basic fibroblast growth factor (also (TGF-β) encompasses a family of ligands, the prototype
known as FGF2); PDGF, platelet-derived growth factor. of which is TGF-β1. Although most research focuses on
the TGF-β1 isoform, the β2 and β3 isoforms also have
profibrotic effects113. They bind to specific transmem-
autoimmune disease, bindarit prevented renal MCP1 brane type I (also termed activin receptor-like kinase,
upregulation, limited glomerular injury and prolonged AlK5) and type II serine/threonine kinase receptors,
animal survival105. Similarly, anecdotal observation in which subsequently propagate the signal by phosphory-
patients with active lupus nephritis showed that bindarit lation of several SMAD transcription factors that
administration significantly reduced urinary albumin accumulate in the nucleus. Eventually they control the
and MCP1 excretion, and was well tolerated106. Therefore, expression of target genes, including those encoding for
targeting MCP1 could be a new way to provide renopro- extracellular matrix components114 (FIG. 4). So, targeting
tection. A clinical trial aimed to reduce albuminuria and of the TGF-β pathway may have therapeutic potential
renal disease progression with bindarit added on to RAAS for tackling profibrotic processes, as suggested by studies
blockade therapy is ongoing in patients with type 2 dia- using an anti-TGF-β monoclonal antibody, inhibitors of
betes and with microalbuminuria or macroalbuminuria TGF-β production, and inhibitors of TGF-β type I and
(national Monitoring Centre for Clinical Trials database type II receptor kinases. Although the mechanism of
identifier 2006-006191-38; see Further information). action is not fully understood, it has been shown that
pirfenidone — which is known to be a collagen syn-
Complement inhibition with anti‑C5 antibodies. thesis inhibitor and an antifibrotic p38 MAP kinase
Cleavage of the C5 component, which results in the inhibitor — suppresses both TGF-β gene expression
fragments C5a and C5b, is the critical event during the at the transcription level and protein expression115, in
activation of the complement cascade107. C5a is a highly addition to antagonizing TnF-α signalling116 and being

944 | nOvEMbER 2008 | vOlUME 7 www.nature.com/reviews/drugdisc


REVIEWS

O The functional role of TGF-β in vivo is emphasized


O by evidence showing that abrogation of TGF-β signalling
O
N
O
by long-term treatment with an anti-TGF-β antibody
H
in db/db mice with type 2 diabetes-like disease almost
NH
completely prevented the increase in collagen and
N N O
N
fibronectin expression and mesangial matrix expan-
N N NH2 sion121. Moreover, pirfenidone reduced the expression
N H
of glomerular TGF-β, attenuated the accumulation of
GW788388 SB431542 glomerular extracellular matrix components122 in the
five-sixths nephrectomy rat model, and improved renal
fibrosis and function in animals with ureteral obstruc-
TGF-β tion123. In addition, in a rat model of puromycin-induced
TGFBRII TGFBRI
(ALK5) nephritis the TGF-β type 1 receptor kinase inhibitor
Sb525334 reduced procollagen 1α(I) deposition in the
GW788388 P P SB431542, renal tissue124. Recently, when the inhibitor Gw788388
GW788388 was given orally for 5 weeks to db/db mice, it reduced
P the mRnA expression of critical factors in extracellular
matrix remodelling, namely COl-I, COl-III and PAI1,
R-SMAD2/3 and significantly lowered renal fibrosis119.
SMAD4 Overall, these observations provide the rationale
for adding agents that are capable of reducing TGF-β
activity or production in combination with inhibitors of
angiotensin II activity as a multimodal intervention to
implement renoprotection. Indeed, treatment with an
SMAD4 anti-TGF-β antibody, when added on the background
Fibronectin of chronic ACE inhibition, normalized proteinuria
Collagen I and abrogated glomerulosclerosis and tubular damage
PAI1
as compared with ACE inhibitor alone in diabetic rats
with overt nephropathy125. Despite encouraging results
in experimental animals by the multimodal intervention
that includes anti-TGF-β antibody, this regimen is not
so easily adaptable to patients with CKD. Anti-TGF-β
antibodies are just at the beginning of their clinical
Figure 4 | structures of TgF-β receptor inhibitors and their inhibitory actions. development in proteinuric chronic nephropathies, such
Nature Reviews
Transforming growth factor-β (TGF-β) binds to the TGF-β-receptor | Drug Discovery
II (TGFBRII), as focal segmental glomerulosclerosis. Certainly, however,
which forms a heteromeric receptor complex with TGF-β-receptor I (TGFBRI or activin more data are needed about their renoprotective effects
receptor-like kinase, ALK5)114. The resulting kinase activation leads to the recruitment before anti-TGF-β antibodies will be part of the thera-
and phosphorylation of specific-receptor regulated (R)-SMADs, which are the peutic options for renoprotection. Of note, in a small
intracellular effectors of TGF-β family members. TGF-β induces SMAD2/3 study in patients with advanced focal segmental glomer-
phosphorylation. This phospho-SMAD forms a heteromeric complex with SMAD4, ulosclerosis, pirfenidone showed a beneficial effect on
which is transported into the nucleus where it regulates the expression of target genes
slowing the loss of glomerular filtration rate, but had no
such as fibronectin, collagen I and plasminogen activator inhibitor, type 1 (PAI1).
effect on proteinuria during 13 months of treatment126.
Apart from stimulating extracellular matrix protein synthesis204, TGF-β induces fibrosis
by inhibiting matrix degradation through the suppression of the activity of matrix However, the safety profile of both anti-TGF-β antibodies,
metalloproteinases205 and activating epithelial to mesenchymal transition, a process pirfenidone and TGF-β receptor kinase inhibitors should
whereby epithelial cells are transformed into migratory myofibroblasts206. TGF-β be carefully explored in view of the fact that complete
receptor inhibitors block activation of TGFBRI (SB431542 and GW788388) and of inhibition of the potent immunosuppressive activity of
TGFBRII (GW788388). TGF-β could have serious adverse consequences.

Thiazolidinediones. Thiazolidinediones (TZDs) represent


a reactive oxygen species scavenger117. The inhibition of a class of compounds that are currently in use for the
TGF-β type I and type II receptor kinases is achieved by treatment of type 2 diabetes; they exert their hypogly-
small-molecule competitive antagonists of the receptor caemic properties through reduction of insulin resist-
kinase activity that interact with the ATP-binding site, ance127. These compounds act by stimulating perixosome
which prevents phosphorylation of SMAD proteins118,119 proliferator-activated receptor-γ (PPAR-γ), which are
Db/db mouse (FIG. 4). The AlK5 inhibitor Sb431542 is commonly used members of the nuclear hormone receptor superfamily
The db/db mouse is a in experimental in vitro and in vivo studies. Recently, a (FIG. 5). Synthetic TZDs, such as pioglitazone (Actos;
hyperinsulinaemic model of new TGF-β receptor kinase inhibitor, Gw788388, has Takeda) and rosiglitazone (Avandia; GlaxoSmithKline),
genetic diabetes that develops been developed that has an improved pharmacokinetic are extensively used as antidiabetic or insulin-sensitizing
abnormalities in renal
morphology and function
profile in comparison with Sb431542 (FIG. 4). In vitro, agents (FIG. 5). Troglitazone, the first PPAR-γ agonist, is
that mimic those in human Gw788388 inhibits both TGF-β type I and type II receptor no longer available for clinical use owing to its hepato-
diabetic nephropathy. kinase activities120. toxicity128. Apart from ameliorating insulin resistance,

nATURE REvIEwS | drug discovery vOlUME 7 | nOvEMbER 2008 | 945


REVIEWS

O O

NH NH
N N S S
O N O
O O
Rosiglitazone Pioglitazone

PPAR RXR-α Target genes Leptin


PPAR RXR-α TNF-α
aP2

PPRE

Figure 5 | structures and agonistic effects of thiazolidinediones. Thiazolidinediones, for example, rosiglitazone and
pioglitazone, bind to peroxisome proliferator-activated receptors (PPARs). After ligand binding, PPARs form heterodimers
Nature Reviews
with retinoid X receptor-α (RXR-α), thereby undergoing to conformational changes, which facilitate | Drug Discovery
their binding to the
peroxisome proliferative response element (PPRE) in the enhancer regions of target genes such as leptin, tumour-necrosis
factor-α (TNF-α) and adipocyte fatty acid-binding protein (aP2).

TZDs also decrease the expression of TGF-β in glomeruli disorganization and calcium accumulation135. It there-
independently of their effect on glucose129, as well as fore may contribute to renal injury via uncoupling of
suppress the glucose-induced production of TGF-β mitochondrial respiration and generation of reactive
from tubular cells130. So, TZDs may offer a clinically oxygen species136. It is well established that the effects of
promising approach to target the profibrotic TGF-β vitamin D on endocrine system functions extend beyond
activity specifically for diabetic nephropathy. the regulation of calcium and phosphate metabolism.
In streptozotocin-induced experimental diabetes, Several important activities of vitamin D may contrib-
pioglitazone suppressed the renal expression of TGF-β, ute to its renoprotective property, which include inhi-
reduced urinary albumin excretion and ameliorated bition of profibrotic growth factors and inflammatory
glomerular injury131. Treatment with rosiglitazone also cytokines and suppression of the RAAS137–139. Moreover,
reduced albuminuria in patients with type 2 diabetes the synthetic vitamin D analogue paricalcitol (Zemplar;
through the inhibition of TnF-α and increase of adipo- Abbott) inhibits renal inflammation by promoting
Puromycin-induced
nectin132. Preliminary data are already available that vitamin D receptor-mediated sequestration of nuclear
nephrotic syndrome
An experimental model of demonstrate the favourable effects of combining angio- factor-κb (nF-κb) signalling in a human proximal
nephrotic syndrome that tensin II receptor antagonists and pioglitazone to target tubular cell line140.
is characterized by heavy proteinuria and to achieve better renoprotection as com- Animal studies have revealed pivotal protective roles
proteinuria and progressive pared with angiotensin II receptor antagonists alone in of vitamin D in the renal and cardiovascular systems141.
glomerular and
tubulointerstitial injury
patients with type 2 diabetic nephropathy133. Although Evidence is also mounting showing that a vitamin D
induced by single or repeated TZDs seem promising for the implementation of reno- analogue is renoprotective in different experimental
injections of the puromycin protection in diabetic nephropathy, caution should be nephropathies142–144. A recent pilot clinical trial shows
aminonucleoside. taken given the recent signal of concern of increased risk that paricalcitol reduces albuminuria and inflammation
of myocardial infarction and, albeit non-significant, of in patients with CKD145,146. nevertheless, long-term ran-
Focal segmental
glomerulosclerosis death from cardiovascular causes134. domized, controlled trials are required to confirm these
A diagnostic term for a clinical benefits of vitamin D analogues.
pathological syndrome that Vitamin D analogues. Calcium-phosphate deposition is
has multiple aetiologies and a frequent histological finding in end-stage kidney biop- Growth‑factor inhibitors. Connective tissue growth
pathogenic mechanisms
characterized by proteinuria
sies, irrespective of the underlying cause of renal failure. factor (CTGF) is a downstream mediator of TGF-β
and focal segmental glomerular Calcium deposition in the renal parenchyma is asso- with a potent profibrotic activity that operates directly
consolidation or scarring. ciated with ultrastructural evidence of mitochondrial by binding to integrins and extracellular matrix or by

946 | nOvEMbER 2008 | vOlUME 7 www.nature.com/reviews/drugdisc


REVIEWS

Table 2 | Efficacy of CDK inhibitors in models of chronic renal diseases Therapeutics to target immunological processes
Cyclin‑dependent kinase/glycogen synthase kinase‑3
Model drug Therapeutic response refs inhibitors. Cyclin-dependent kinase and glycogen syn-
Mesangial proliferative glomerulonephritis thase kinase-3 (GSK3) are families of serine/threonine
Anti-Thy 1.1 antibody Roscovitine Decreases mesangial cell 162 kinases that share high structural similarity at their
(rat) proliferation and mesangial matrix ATP-binding and catalytic domains158. They have been
production; ameliorates renal recognized to participate in the pathogenesis and repair
insufficiency of renal tissue injury through their ability to promote
Crescentic glomerulonephritis engagement (G0 to G1) and progression (G1 to M) of the
Nephrotoxic form (rat) Roscovitine Decreases crescent formation 208 cell cycle; to influence proliferation, apoptosis and inflam-
mation in part by direct control of gene expression; and
Anti-total glomerular Roscovitine Decreases glomerular matrix 163
protein form (mice) deposition; ameliorates renal to modulate tissue morphogenesis159. Moreover, GSK3
insufficiency is known to control the nOTCH pathway160, which has
HIV-induced collapsing glomerulopathy recently been shown to participate in podocyte injury
and possibly to the development of glomerulosclerosis161.
Tg26 (mice) Flavopiridol Prevents and regresses existing 209
Small-molecule cyclin-dependent kinase/GSK3 inhibi-
nephropathy
tors have been proposed as therapeutic molecules for
Tg26 (mice) Roscovitine Prevents and regresses existing 164 CKD (TABLE 2). In addition, some cyclin-dependent
nephropathy
kinase/GSK3 inhibitors have recently been shown to
Membranous nephropathy inhibit the nOTCH signalling pathway160.
Passive Heymann Roscovitine Decreases renal-cell proliferation; 210 Cumulative evidence suggests the preclinical efficacy
nephritis (rat) stabilizes podocyte injury of cyclin-dependent kinase/GKS3 inhibitors as renopro-
Proliferative lupus nephritis tective agents. This has been documented mainly with
Roscovitine Decreases glomerular 165
roscovitine in experimental models of mesangial prolifer-
NZB × NZW (mice)
hypercellularity ative glomerulonephritis162, crescentic glomerulonephritis
and collapsing glomerulopathy163,164, and proliferative lupus
NZB × NZW (mice) Roscovitine Decreases tubulointerstitial 166
damage, immunoglobulin and nephritis165,166. However, clinical trials of roscovitine in
complement deposition, patients with immunoglobulin A (IgA) nephropathy
and proteinuria were halted over concerns of potential toxicity with its
Diabetic nephropathy prolonged use167.
Streptozotocin BIO Inhibits mesangial cell apoptosis; 211
-induced diabetes (rat) decreases proteinuria Gamma‑secretase inhibitors. nOTCH genes encode
large, single-transmembrane receptors that regulate
Polycystic kidney disease
a broad range of cell-fate decisions 168. Recently, the
jck (mice) Roscovitine Long-lasting arrest of renal 212 transmembrane receptor nOTCH has been shown to
cystogenesis be expressed on the podocyte surface in the setting of
cpk (mice) Roscovitine Long-lasting arrest of renal 212 animal and human glomerulopathies. Moreover, the
cystogenesis nOTCH pathway is involved in the development of
BIO, (2′2,3′E)-6-bromoindirubin-3-oxime; CDK,cyclin-dependent kinase; cpk, congenital glomerular disease161. binding of the ligand jagged 1
polycystic kidney (also known as Cys1); jck, juvenile cystic kidney (also known as Nek8).
(JAG1) to nOTCH renders the transmembrane receptor
susceptible to γ-secretase-mediated proteolytic cleavage,
activating the intracellular nOTCH pathway, which
enhancing the signalling of other growth factors including eventually results in the upregulation of target genes
epidermal growth factor (EGF), platelet-derived growth and cell apoptosis161. Thus, γ-secretase could represent a
factor (PDGF) and basic fibroblast growth factor (bFGF; new therapeutic target for glomerulopathies. Inhibitors
also known as FGF2)147. Inhibition of CTGF markedly of γ-secretase have been developed and are already in
reduced renal fibrosis in experimental models of dia- Phase II and III clinical trials for Alzheimer’s disease169
betic nephropathy, renal mass ablation and unilateral (FIG. 6). Among them, l-685458, an aspartyl protease
ureteral obstruction147–150. Therapy with an anti-CTGF transition-state mimic, is a potent inhibitor of amyloid-β-
Collapsing glomerulopathy
antibody is now in clinical trials in diabetic nephropathy protein precursor γ-secretase activity170. Dibenzazepine
Collapsing glomerulopathy is a (FibroGen Clinical Trials; see Further information). inhibits the proliferation of cells in intestinal crypts
morphological variant of focal Similarly, PDGF is a key mediator of fibrosis in different and adenomas, thereby suggesting benefit in colorec-
segmental glomerulosclerosis organs including the kidney151. The multikinase inhibi- tal neoplastic disease171. DAPT (N-[N-(3,5-difluoro-
that is characterized by
tor imatinib (Gleevec/Glivec; novartis), which is widely phenyl)-l-alanyl]-S-phenylglycine-1,1-dimethylester)
segmental and global collapse
of the glomerular capillaries, used in cancer therapy, also blocks PDGF-receptor- blocks the nOTCH pathway and inhibits chondro-
marked hypertrophy and activated tyrosine kinases and attenuates renal damage genesis in human mesenchymal stem cells172. Of note, all
hyperplasia of podocytes, and in experimental unilateral ureteral obstruction152, lupus these γ-secretase inhibitors not only prevented the devel-
severe tubulointerstitial disease. nephritis153 and diabetic nephropathy154,155. However, it is opment of albuminuria, but also lowered established albu-
Intestinal crypts
uncertain whether imatinib could be a useful therapeutic minuria in experimental puromycin-induced nephrotic
Mucosal epithelium that is approach in patients with CKD given its side effects on syndrome161. The anti-albuminuric effect was associated
extensively invaginated. haematopoiesis, heart and bone156,157. with preservation of podocytes and protection against

nATURE REvIEwS | drug discovery vOlUME 7 | nOvEMbER 2008 | 947


REVIEWS

O F
O N
O
H
F N O NH
N F
H N
O O O
O
DAPT H H Dibenzazepine
F O N N NH2
N
H
O O O

L-685458

γ-secretase inhibitors

TGF-β

Podocyte Podocyte
γ-secretase
JAG1 NOTCH–ICD
Apoptosis
NOTCH

JAG1 p300
RBPJ Target genes
MAML ↑TGF-β

Figure 6 | structure and function of γ-secretase inhibitors on podocytes. In podocytes, transforming growth
factor-β (TGF-β) activates the transcription of the NOTCH ligand jagged 1 (JAG1). NOTCH is a transmembrane receptor
to γ-secretase-mediated
that is expressed on the surface of podocytes. Binding of JAG1 renders NOTCH susceptibleNature Reviews | Drug Discovery
proteolytic cleavage, thereby resulting in the release of the NOTCH intracellular domain (ICD) from the plasma
membrane and its translocation into the nucleus. Here, NOTCH–ICD binds to the DNA-binding protein recombinant
signal-binding protein J (RBPJ) and forms a nuclear complex with the cofactors MAML and p300. This complex blocks
transcriptional inhibition of RBPJ and leads to upregulation of NOTCH target gene transcripts, including those that
encode TGF-β. In turn, TGF-β may activate intracellular pathways that promote cell apoptosis. Inhibitors of γ-secretase
prevent NOTCH cleavage and eventually podocyte apoptosis. DAPT, N-[N-(3,5-difluorophenyl)-l-alanyl]-S-phenylglycine-
1,1-dimethylester.

glomerulosclerosis161. These findings provide proof of glomerular capillary barrier to macromolecules, which
principle that inhibition of γ-secretase could be a new leads to proteinuria and eventually glomerulosclero-
modality for treatment of chronic glomerulopathies. sis and tubulointerstitial fibrosis. Corticosteroids and
cytotoxic drugs are currently used as induction and/or
Rituximab. Rituximab (MabThera; Roche) is an engi- maintenance therapy176, but attempts to modulate this
neered chimeric monoclonal antibody that is directed antibody-mediated response have not been consistently
against the CD20 molecule found on pre-b and mature successful, in addition to exposing patients to a number
b cells173. It was first approved in 1997 for the treat- of serious side effects. More specific and less toxic thera-
ment of relapsed or refractory b-cell non-Hodgkin’s pies that target antibody-producing b cells are therefore
lymphoma174. Although the exact mechanism by which needed. based on its property of selectively depleting
Glomerular basement rituximab exerts its effect remains unclear, it is likely CD20+ b cells, rituximab has been recently proposed as
membrane
that b-cell depletion occurs through antibody-mediated a therapeutic option for antibody-mediated glomerulo-
This is one of the components
of filtration barrier between apoptosis, antibody-dependent cellular cytotoxicity and pathies, namely membranous nephropathy, cryoglobuli-
the blood and urinary space, complement-mediated toxicity175. naemic glomerulonephritis and SlE.
which is composed of a central Antibody production by b cells is characteristic of Indeed, the hallmark of idiopathic membranous
dense layer, the lamina numerous primary and secondary glomerular diseases nephropathy (IMn) is the subepithelial deposition of
densa, and two thinner,
more electron-lucent layers,
of the kidney. Antibody binding to glomerular cells and/ IgG on the glomerular basement membrane, in some cases
the lamina rara externa or basement membranes promotes an inflammatory associated with CD20+ infiltrates177. A study in eight
and the lamina rara interna. response that alters the size-selective properties of the patients with persistent IMn and nephrotic syndrome

948 | nOvEMbER 2008 | vOlUME 7 www.nature.com/reviews/drugdisc


REVIEWS

first showed that four infusions of 375 mg per m2 rituxi- single dose of the anti-CD20 antibody179. However, ritux-
mab at weekly intervals significantly reduced proteinuria, imab did not lower proteinuria at the same extent in all
with two patients achieving full remission (proteinuria patients179,180. This was not explained by different effects
<1 g per day)178. Rituximab treatment was well tolerated. of the depleting antibody on b lymphocytes, but to the
Recently, similar results have been achieved with only a severity of the accompanying chronic renal lesions. It
has been suggested that the immune pathways of IMn
can be effectively affected by b-cell depletion therapy,
Box 1 | New therapies for genetic renal diseases
whereas non-immune mechanisms could be control-
Insights into hereditary diseases of the kidney are constantly evolving as new led by treatments, such as RAAS blockade179. Therefore,
information about their molecular and cellular mechanisms become available at an renal biopsy findings may help to tailor treatment to
increasingly rapid rate. A better understanding of the genetic and pathophysiology of individual patients with IMn.
these diseases and the availability of animal models that are orthologous to the human Type II mixed essential cryoglobulinaemia is often associ-
disease has enabled the development of preclinical studies and the identification
ated with hepatitis C virus (HCv) infection, and leads
of promising candidate drugs for clinical trials in some renal diseases for which, until
to membranoproliferative glomerulonephritis and usu-
recently, only genetic counselling and symptomatic therapies were available.
ally systemic vasculitis181. very few studies, however, are
recombinant α-galactosidase A for Fabry’s disease so far available of rituximab therapy in this disorder. In
Fabry’s disease, also known as angiokeratoma corporis diffusum universalis,
six patients with HCv-associated cryoglobulinaemia
is an X-chromosome-linked inborn error of metabolism caused by a deficiency of
α-galactosidase A, an enzyme that is necessary for the catabolism of ceramide and renal involvement, a prolonged regimen of rituxi-
trihexoside (a glycosphingolipid)194. This enzymatic defect interferes with mab resulted in reduced proteinuria and improved renal
intralysosomal degradation of glycosphingolipids and results in their accumulation function at 12–18 months of follow-up182.
in various tissues of the body outside the central nervous system with various SlE is the prototypical immune complex disorder,
manifestations. Renal involvement is common in male hemizygotes and is occasional in and involves a wide range of autoantibodies. nephritis
female heterozygotes. The disease presents with haematuria and proteinuria, which is common in SlE, perhaps invariably. In a small study
often progresses to nephrotic levels. Recently, recombinant human α-galactosidase A of ten patients with proliferative nephritis the standard
replacement therapy has become available, and two randomized controller trials have therapeutic protocol of rituximab and oral steroids suc-
demonstrated its safety and efficacy. In one short-term study, α-galactosidase A cessfully achieved complete remission in four patients
treatment improved neuropathic pain, decreased mesangial widening and increased
after 1 year of follow-up183. One patient attained tem-
renal function195. In another study repeated renal biopsies showed decreased
microvascular endothelial deposits of globotriaosylceramide196. On this basis, clinical porary complete remission and the other three attained
guidelines for the management of Fabry’s disease have recently been published197. partial remission. In other studies of SlE patients treated
They encourage enzyme-replacement therapy as early as possible in all males with with rituximab, only a small proportion also had renal
Fabry’s disease and in female carriers with substantial disease manifestations. disease184,185. Overall, rituximab provided some clinical
Novel disease-targeted treatments for AdPKd and biochemical benefits in the majority of patients.
Autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive nevertheless, rituximab use should be confined to
polycystic kidney disease (ARPKD) are the most well known of a large family of patients who are resistant to standard induction therapy
inherited diseases that are characterized by the development of renal cysts of tubular or to patients with contraindications to corticosteroids
epithelial cell origin198. Identification of the causative mutated genes and elucidation and immunosuppressive agents until more data are avail-
of the function of their encoded proteins is shedding insights on the mechanisms that able from ongoing clinical trials.
underlie tubular epithelial cell differentiation198. These advances in the understanding
of ADPKD pathogenesis open the possibility for novel disease-specific treatment Conclusions
strategies that overcome the current therapies directed towards limiting the
One of the milestones in nephrology has been the char-
morbidity and mortality from the complications of the disease, which include
hypertension, pain, cyst haemorrhage and infection, and nephrolithiasis. Two major
acterization — during the middle of the 1980s — of
approaches as potential treatment are targeting cell proliferation and lowering the intraglomerular mechanisms underlying the pro-
intracellular cyclic AMP199. They have been shown to be effective in preventing gressive nature of proteinuric chronic nephropathies
cyst formation and renal-disease progression in several models of PKD (see towards ESRD4 and the identification of specific treat-
Supplementary information S3 (table)). ments targeting the deleterious effect of excessive traffic
Caution, however, is necessary when directly extrapolating results form rodent of proteins through the glomerular capillary barrier16.
models of renal cysts for human PKD. Apart from differences in genotype, cysts may The evidence in animal models that ACE inhibitors,
originate from different nephron segments and this could influence the effectiveness besides lowering blood pressure, had selective antipro-
of different drugs. Nevertheless, clinical studies with some novel therapeutics are teinuric and renoprotective properties opened a new era
ongoing. Phase II clinical trials with the vasopressin 2 receptor antagonist tolvaptan
for the treatment of patients with CKD. This dictated
have been completed in patients with ADPKD, and a Phase III trial with this molecule
is under way worldwide198. In a pilot study the synthetic metabolically stable
the current and consolidated therapeutic approach for
somatostatin analogue octreotide inhibited renal growth and stabilized renal function proteinuric chronic nephropathies based on blockade
in patients with ADPKD200. Given the safety and efficacy of the treatment, a long-term of the RAAS with ACE inhibitors and/or angiotensin
Phase III study with octreotide has been started. Similarly, based on encouraging II receptor antagonists, which limit proteinuria and
results in three rodent models of PKD demonstrating that mTOR (mammalian target reduce renal function decline and risk of ESRD more
of rapamycin) inhibitors significantly retard cyst expansion and protect renal effectively than other antihypertensive treatments. Even
function201,202, Phase II clinical trials of rapamycin and everolimus, two mTOR inhibitors, more intriguing are the more recent experimental186 and
are being implemented203. Given the complexity of the cystic disease process, it seems clinical187,188 observations that regression of glomerular
likely that a combination strategy will be required for maximal therapeutic benefit. structural changes and remodelling of the glomeru-
Nevertheless in the next decade, much promise is expected of an effective therapy
lar architecture is achievable with RAAS blockade.
for a so far untreatable disease.
However, precisely which cells are involved in the process

nATURE REvIEwS | drug discovery vOlUME 7 | nOvEMbER 2008 | 949


REVIEWS

of scar tissue removal and regeneration of endothelial experimental models of acute renal injury192,193 — would
Type II mixed essential
cryoglobulinaemia and mesangial cells and podocytes is not yet known. be a promising future alternative to repair chronic renal
Cryoglobulinaemia refers to a Regression of glomerulosclerosis and neoformation of injury and promote renoprotection.
pathogenic condition caused glomerular tissue has been linked to progenitor or stem Moreover, there is a clear need to find further thera-
by the production of cells of renal or extrarenal origin189,190, and ACE inhibi- pies to better control antibody production in immune-
circulating immunoglobulins
that precipitate upon cooling.
tors or angiotensin II receptor antagonists may promote related nephropathies, and rituximab is a promising
Type II cryoglobulinaemia their mobilization and/or activation at the site of renal option. Certainly, it seems likely that biological agents
is defined as mixed injury191. For non-responders or partial responders, treat- that are directed at b-cell depletion or inhibition
cryoglobulins containing at ment procedure to remission and/or regression must will form part of the immunotherapeutic armamen-
least two immunoglobulins,
include a multi-pharmacological strategy to promote tarium in the future treatment of immune-mediated
and has no clear aetiology.
renoprotection through novel anti-inflammatory and glomerulopathies.
Systemic vasculitis antifibrotic molecules. Added on to RAAS inhibitors, Of note, recent insights into the genetic and patho-
systemic vasculitis comprises they might theoretically potentiate the tissue reparative physiology of some hereditary diseases of the kidney
a large group of inflammatory effects of resident or extrarenal stem cells. The possibility, allowed the identification of promising candidate drugs
diseases with a suggestive or
proven immunopathogenesis
however, exists of patients with CKD who are resistant for diseases for which only symptomatic treatments so
involving blood vessels of to the multimodal therapeutic options that have regen- far are available (BOX 1).
various sizes and affecting erative potential, particularly when pharmacological Overall, as existing drugs for the treatment of chronic
various organs including intervention is started late. This could reflect the very nephropathies have a favourable risk/benefit ratio, new
the kidney.
low number of remaining stem cell niches in the adult therapies to induce renoprotection will require a similar
injured kidney. In this setting, cell therapy with exogenous or better safety profile if they are to be widely used and
mesenchymal stem cells — as recently documented in accepted.

1. Dirks, J., Remuzzi, G., Horton, S., Schieppati, A. & 15. Durvasula, R. V. et al. Activation of a local tissue 27. Iseki, K., Ikemiya, Y., Iseki, C. & Takishita, S.
Rizvi, S. A. H. in Disease Control Priorities in angiotensin system in podocytes by mechanical strain. Proteinuria and the risk of developing end-stage renal
Developing Countries (eds Jamison, D. T. et al.) Kidney Int. 65, 30–39 (2004). disease. Kidney Int. 63, 1468–1474 (2003).
695–706 (Oxford University Press and The World The first direct evidence that podocytes produce 28. Remuzzi, G. & Bertani, T. Pathophysiology of
Bank, New York, 2006). angiotensin II in response to mechanical stress. progressive nephropathies. N. Engl. J. Med. 339,
2. Gridelli, B. & Remuzzi, G. Strategies for making more 16. Anderson, S., Meyer, T. W., Rennke, H. G. & Brenner, 1448–1456 (1998).
organs available for transplantation. N. Engl. J. Med. B. M. Control of glomerular hypertension limits 29. Giatras, I., Lau, J. & Levey, A. S. Effect of
343, 404–410 (2000). glomerular injury in rats with reduced renal mass. angiotensin-converting enzyme inhibitors
3. Collins, A. J. The hemoglobin link to adverse J. Clin. Invest. 76, 612–619 (1985). on the progression of nondiabetic renal disease:
outcomes. Adv. Stud Med. 3, S14–S17 (2003). Breakthrough observation that control of increased a meta-analysis of randomized trials.
4. Hostetter, T. H., Olson, J. L., Rennke, H. G., intraglomerular capillary pressure with ACE Angiotensin-Converting-Enzyme Inhibition and
Venkatachalam, M. A. & Brenner, B. M. Hyperfiltration inhibitors limits glomerular injury and renal-disease Progressive Renal Disease Study Group. Ann. Intern.
in remnant nephrons: a potentially adverse response progression in an animal model. Med. 127, 337–345 (1997).
to renal ablation. Am. J. Physiol. 241, F85–F93 17. Zoja, C., Benigni, A. & Remuzzi, G. Cellular responses 30. The GISEN Group (Gruppo Italiano di Studi
(1981). to protein overload: key event in renal disease Epidemiologici in Nefrologia). Randomised placebo-
5. Remuzzi, G., Ruggenenti, P. & Perico, N. Chronic renal progression. Curr. Opin. Nephrol. Hypertens. 13, controlled trial of effect of ramipril on decline in
diseases: renoprotective benefits of renin–angiotensin 31–37 (2004). glomerular filtration rate and risk of terminal renal
system inhibition. Ann. Intern. Med. 136, 604–615 18. Denton, K. M., Anderson, W. P. & Sinniah, R. Effects of failure in proteinuric, non-diabetic nephropathy.
(2002). angiotensin II on regional afferent and efferent Lancet 349, 1857–1863 (1997).
6. Cortes, P., Zhao, X., Riser, B. L. & Narins, R. G. arteriole dimensions and the glomerular pole. Am. J. The first documentation that an ACE inhibitor
Regulation of glomerular volume in normal and Physiol. Regul. Integr. Comp. Physiol. 279, slowed the rate of glomerular filtration decline
partially nephrectomized rats. Am. J. Physiol. 270, R629–R638 (2000). and halved the risk of ESRD in patients with
F356–F370 (1996). 19. Kagami, S., Border, W. A., Miller, D. E. & Noble, N. A. non-diabetic nephropathy. This was in
7. Hostetter, T. H. Progression of renal disease and renal Angiotensin II stimulates extracellular matrix protein comparison with conventional antihypertensive
hypertrophy. Annu. Rev. Physiol. 57, 263–278 synthesis through induction of transforming growth therapy at equivalent level of blood pressure
(1995). factor-β expression in rat glomerular mesangial cells. control.
8. Kang, D. H. et al. Impaired angiogenesis in the J. Clin. Invest. 93, 2431–2437 (1994). 31. Ruggenenti, P., Perna, A., Gherardi, G., Benini, R.
remnant kidney model: I. Potential role of vascular 20. Vaughan, D. E., Lazos, S. A. & Tong, K. Angiotensin II & Remuzzi, G. Chronic proteinuric nephropathies:
endothelial growth factor and thrombospondin-1. regulates the expression of plasminogen activator outcomes and response to treatment in a prospective
J. Am. Soc. Nephrol. 12, 1434–1447 (2001). inhibitor-1 in cultured endothelial cells. A potential cohort of 352 patients with different patterns of
9. Brooks, A. R., Lelkes, P. I. & Rubanyi, G. M. Gene link between the renin–angiotensin system and renal injury. Am. J. Kidney Dis. 35, 1155–1165
expression profiling of vascular endothelial cells thrombosis. J. Clin. Invest. 95, 995–1001 (1995). (2000).
exposed to fluid mechanical forces: relevance for focal 21. Macconi, D. et al. Permselective dysfunction of 32. Ruggenenti, P., Schieppati, A. & Remuzzi, G.
susceptibility to atherosclerosis. Endothelium 11, podocyte–podocyte contact upon angiotensin II Progression, remission, regression of chronic renal
45–57 (2004). unravels the molecular target for renoprotective diseases. Lancet 357, 1601–1608 (2001).
10. Ingram, A. J., Ly, H., Thai, K., Kang, M. & Scholey, intervention. Am. J. Pathol. 168, 1073–1085 33. Jafar, T. H. et al. Progression of chronic kidney disease:
J. W. Activation of mesangial cell signaling cascades (2006). the role of blood pressure control, proteinuria, and
in response to mechanical strain. Kidney Int. 55, 22. Rudnicki, M. et al. Gene expression profiles of human angiotensin-converting enzyme inhibition: a patient-
476–485 (1999). proximal tubular epithelial cells in proteinuric level meta-analysis. Ann. Intern. Med. 139, 244–252
11. Harris, R. C., Haralson, M. A. & Badr, K. F. nephropathies. Kidney Int. 71, 325–335 (2007). (2003).
Continuous stretch–relaxation in culture alters rat 23. Johnson, D. W., Saunders, H. J., Baxter, R. C., Field, 34. Bakris, G. L. Hypertension and nephropathy. Am. J.
mesangial cell morphology, growth characteristics, M. J. & Pollock, C. A. Paracrine stimulation of human Med. 115 (Suppl. 8A), 49S–54S (2003).
and metabolic activity. Lab. Invest. 66, 548–554 renal fibroblasts by proximal tubule cells. Kidney Int. 35. Ruggenenti, P. et al. Blood-pressure control for
(1992). 54, 747–757 (1998). renoprotection in patients with non-diabetic chronic
12. Durvasula, R. V. & Shankland, S. J. Podocyte injury 24. Schlondorff, D. O. Overview of factors contributing to renal disease (REIN-2): multicentre, randomised
and targeting therapy: an update. Curr. Opin. Nephrol. the pathophysiology of progressive renal disease. controlled trial. Lancet 365, 939–946 (2005).
Hypertens. 15, 1–7 (2006). Kidney Int. 74, 860–866 (2008). 36. Lewis, E. J., Hunsicker, L. G., Bain, R. P. & Rohde, R. D.
13. Morton, M. J. et al. Human podocytes possess 25. Eddy, A. Role of cellular infiltrates in response to The effect of angiotensin-converting-enzyme inhibition
a stretch-sensitive, Ca2+-activated K+ channel: proteinuria. Am. J. Kidney Dis. 37, S25–S29 (2001). on diabetic nephropathy. The Collaborative Study
potential implications for the control of glomerular 26. Ruggenenti, P., Perna, A., Mosconi, L., Pisoni, R. & Group. N. Engl. J. Med. 329, 1456–1462 (1993).
filtration. J. Am. Soc. Nephrol. 15, 2981–2987 Remuzzi, G. Urinary protein excretion rate is the best 37. Ravid, M. et al. Use of enalapril to attenuate decline in
(2004). independent predictor of ESRF in non-diabetic renal function in normotensive, normoalbuminuric
14. Endlich, N. et al. Podocytes respond to mechanical proteinuric chronic nephropathies. “Gruppo Italiano di patients with type 2 diabetes mellitus. A randomized,
stress in vitro. J. Am. Soc. Nephrol. 12, 413–422 Studi Epidemiologici in Nefrologia” (GISEN). Kidney controlled trial. Ann. Intern. Med. 128, 982–988
(2001). Int. 53, 1209–1216 (1998). (1998).

950 | nOvEMbER 2008 | vOlUME 7 www.nature.com/reviews/drugdisc


REVIEWS

38. Ruggenenti, P. et al. Preventing microalbuminuria in 61. Schjoedt, K. J. et al. Beneficial impact of 86. Zoja, C. et al. Effect of combining ACE inhibitor and
type 2 diabetes. N. Engl. J. Med. 351, 1941–1951 spironolactone in diabetic nephropathy. Kidney Int. statin in severe experimental nephropathy. Kidney Int.
(2004). 68, 2829–2836 (2005). 61, 1635–1645 (2002).
The BENEDICT trial documented for the first time 62. Epstein, M. et al. Selective aldosterone blockade with 87. Douglas, K., O’Malley, P. G. & Jackson, J. L. Meta-
the effect of ACE inhibitors on preventing the eplerenone reduces albuminuria in patients with type 2 analysis: the effect of statins on albuminuria. Ann.
onset of nephropathy in patients with type 2 diabetes. Clin. J. Am. Soc. Nephrol. 1, 940–951 Intern. Med. 145, 117–124 (2006).
diabetes. (2006). 88. Strippoli, G. F. et al. Effects of statins in patients with
39. Parving, H. H. et al. The effect of irbesartan on the 63. Bianchi, S., Bigazzi, R. & Campese, V. M. Antagonists chronic kidney disease: meta-analysis and meta-
development of diabetic nephropathy in patients with of aldosterone and proteinuria in patients with CKD: regression of randomised controlled trials. BMJ 336,
type 2 diabetes. N. Engl. J. Med. 345, 870–878 an uncontrolled pilot study. Am. J. Kidney Dis. 46, 645–651 (2008).
(2001). 45–51 (2005). 89. Sandhu, S., Wiebe, N., Fried, L. F. & Tonelli, M. Statins
40. Brenner, B. M. et al. Effects of losartan on renal and 64. Bomback, A. S., Kshirsagar, A. V., Amamoo, M. A. & for improving renal outcomes: a meta-analysis. J. Am.
cardiovascular outcomes in patients with type 2 Klemmer, P. J. Change in proteinuria after adding Soc. Nephrol. 17, 2006–2016 (2006).
diabetes and nephropathy. N. Engl. J. Med. 345, aldosterone blockers to ACE inhibitors or angiotensin 90. Guthrie, R. M. & Martin, D. R. The safety of
861–869 (2001). receptor blockers in CKD: a systematic review. Am. J. rosuvastatin: effects on renal and hepatic function.
41. Lewis, E. J. et al. Renoprotective effect of the Kidney Dis. 51, 199–211 (2008). Expert Opin. Drug Saf. 6, 573–581 (2007).
angiotensin-receptor antagonist irbesartan in patients 65. Karet, F. E. & Davenport, A. P. Localization of 91. Baigent, C. & Landry, M. Study of Heart and Renal
with nephropathy due to type 2 diabetes. N. Engl. J. endothelin peptides in human kidney. Kidney Int. 49, Protection (SHARP). Kidney Int. Suppl. S207–S210
Med. 345, 851–860 (2001). 382–387 (1996). (2003).
These three large trials (references 39–41) have 66. Kohan, D. E. Endothelins in the normal and diseased 92. Locksley, R. M., Killeen, N. & Lenardo, M. J. The TNF
provided evidence that angiotensin II receptor kidney. Am. J. Kidney Dis. 29, 2–26 (1997). and TNF receptor superfamilies: integrating
antagonists are renoprotective independently of 67. Arai, H., Hori, S., Aramori, I., Ohkubo, H. & mammalian biology. Cell 104, 487–501 (2001).
their antihypertensive effect in patients with type 2 Nakanishi, S. Cloning and expression of a cDNA 93. Khan, S. B. et al. Antibody blockade of TNF-α reduces
diabetes and with incipient or overt nephropathy. encoding an endothelin receptor. Nature 348, inflammation and scarring in experimental crescentic
42. de Gasparo, M., Catt, K. J., Inagami, T., Wright, J. W. 730–732 (1990). glomerulonephritis. Kidney Int. 67, 1812–1820
& Unger, T. International Union of Pharmacology. 68. Sakurai, T. et al. Cloning of a cDNA encoding a (2005).
XXIII. The angiotensin II receptors. Pharmacol. Rev. non-isopeptide-selective subtype of the endothelin 94. Meldrum, K. K. et al. TNF-α neutralization ameliorates
52, 415–472 (2000). receptor. Nature 348, 732–735 (1990). obstruction-induced renal fibrosis and dysfunction.
43. Taal, M. W. & Brenner, B. M. Combination ACEI and 69. Davenport, A. P. International Union of Pharmacology. Am. J. Physiol. Regul. Integr. Comp. Physiol. 292,
ARB therapy: additional benefit in renoprotection? XXIX. Update on endothelin receptor nomenclature. R1456–R1464 (2007).
Curr. Opin. Nephrol. Hypertens. 11, 377–381 (2002). Pharmacol. Rev. 54, 219–226 (2002). 95. Bongartz, T. et al. Anti-TNF antibody therapy in
44. Campbell, R. et al. Effects of combined ACE inhibitor 70. Verhaar, M. C. et al. Endothelin-A receptor antagonist- rheumatoid arthritis and the risk of serious infections
and angiotensin II antagonist treatment in human mediated vasodilatation is attenuated by inhibition of and malignancies: systematic review and meta-
chronic nephropathies. Kidney Int. 63, 1094–1103 nitric oxide synthesis and by endothelin-B receptor analysis of rare harmful effects in randomized
(2003). blockade. Circulation 97, 752–756 (1998). controlled trials. JAMA 295, 2275–2285 (2006).
45. Yusuf, S. et al. Telmisartan, ramipril, or both in 71. Moreland, S., McMullen, D. M., Delaney, C. L., Lee, 96. Huugen, D., Cohen Tervaert, J. W. & Heeringa, P.
patients at high risk for vascular events. N. Engl. J. V. G. & Hunt, J. T. Venous smooth muscle contains TNF-α bioactivity-inhibiting therapy in ANCA-
Med. 358, 1547–1559 (2008). vasoconstrictor ETB-like receptors. Biochem. Biophys. associated vasculitis: clinical and experimental
46. Staessen, J. A., Li, Y. & Richart, T. Oral renin Res. Commun. 184, 100–106 (1992). considerations. Clin. J. Am. Soc. Nephrol. 1,
inhibitors. Lancet 368, 1449–1456 (2006). 72. Remuzzi, G., Perico, N. & Benigni, A. New therapeutics 1100–1107 (2006).
47. Rongen, G. A., Lenders, J. W., Smits, P. & Thien, T. that antagonize endothelin: promises and frustrations. 97. Mann, D. L. et al. Targeted anticytokine therapy in
Clinical pharmacokinetics and efficacy of renin Nature Rev. Drug Discov. 1, 986–1001 (2002). patients with chronic heart failure: results of the
inhibitors. Clin. Pharmacokinet. 29, 6–14 (1995). 73. Benigni, A. et al. A specific endothelin subtype A Randomized Etanercept Worldwide Evaluation
48. Wood, J. M. et al. Structure-based design of aliskiren, receptor antagonist protects against injury in renal (RENEWAL). Circulation. 109, 1594–1602 (2004).
a novel orally effective renin inhibitor. Biochem. disease progression. Kidney Int. 44, 440–444 (1993). 98. Sean Eardley, K. & Cockwell, P. Macrophages and
Biophys. Res. Commun. 308, 698–705 (2003). 74. Nabokov, A. et al. Influence of specific and non-specific progressive tubulointerstitial disease. Kidney Int. 68,
49. Pilz, B. et al. Aliskiren, a human renin inhibitor, endothelin receptor antagonists on renal morphology 437–455 (2005).
ameliorates cardiac and renal damage in double- in rats with surgical renal ablation. Nephrol. Dial. 99. Kitagawa, K. et al. Blockade of CCR2 ameliorates
transgenic rats. Hypertension 46, 569–576 (2005). Transplant. 11, 514–520 (1996). progressive fibrosis in kidney. Am. J. Pathol. 165,
50. Stanton, A., Jensen, C., Nussberger, J. & O’Brien, E. 75. Benigni, A. et al. Renoprotective effect of 237–246 (2004).
Blood pressure lowering in essential hypertension with contemporary blocking of angiotensin II and 100. Lloyd, C. M. et al. RANTES and monocyte
an oral renin inhibitor, aliskiren. Hypertension 42, endothelin-1 in rats with membranous nephropathy. chemoattractant protein-1 (MCP-1) play an important
1137–1143 (2003). Kidney Int. 54, 353–359 (1998). role in the inflammatory phase of crescentic nephritis,
51. Parving, H. H., Persson, F., Lewis, J. B., Lewis, E. J. & 76. Benigni, A. et al. Unselective inhibition of endothelin but only MCP-1 is involved in crescent formation and
Hollenberg, N. K. Aliskiren combined with losartan in receptors reduces renal dysfunction in experimental interstitial fibrosis. J. Exp. Med. 185, 1371–1380
type 2 diabetes and nephropathy. N. Engl. J. Med. diabetes. Diabetes 47, 450–456 (1998). (1997).
358, 2433–2446 (2008). 77. Nakamura, T., Ebihara, I., Tomino, Y. & Koide, H. 101. Anders, H. J. et al. A chemokine receptor CCR-1
52. Delyani, J. A. Mineralocorticoid receptor antagonists: Effect of a specific endothelin A receptor antagonist antagonist reduces renal fibrosis after unilateral
the evolution of utility and pharmacology. Kidney Int. on murine lupus nephritis. Kidney Int. 47, 481–489 ureter ligation. J. Clin. Invest. 109, 251–259
57, 1408–1411 (2000). (1995). (2002).
53. Xue, C. & Siragy, H. M. Local renal aldosterone 78. Berthold, H., Munter, K., Just, A., Kirchheim, H. R. & 102. Vielhauer, V. et al. CCR1 blockade reduces interstitial
system and its regulation by salt, diabetes, and Ehmke, H. Contribution of endothelin to renal vascular inflammation and fibrosis in mice with
angiotensin II type 1 receptor. Hypertension 46, tone and autoregulation in the conscious dog. Am. J. glomerulosclerosis and nephrotic syndrome. Kidney
584–590 (2005). Physiol. 276, F417–F424 (1999). Int. 66, 2264–2278 (2004).
54. Del Vecchio, L., Procaccio, M., Vigano, S. & Cusi, D. 79. Imai, T. et al. Induction of endothelin-1 gene by 103. Topham, P. S. et al. Lack of chemokine receptor CCR1
Mechanisms of disease: the role of aldosterone in angiotensin and vasopressin in endothelial cells. enhances Th1 responses and glomerular injury
kidney damage and clinical benefits of its blockade. Hypertension 19, 753–757 (1992). during nephrotoxic nephritis. J. Clin. Invest. 104,
Nature Clin. Pract. Nephrol. 3, 42–49 (2007). 80. Goddard, J. et al. Endothelin-A receptor antagonism 1549–1557 (1999).
55. Remuzzi, G., Cattaneo, D. & Perico, N. The reduces blood pressure and increases renal blood flow 104. Cioli, V. et al. A new protein antidenaturant agent,
aggravating mechanisms of aldosterone on kidney in hypertensive patients with chronic renal failure: bindarit, reduces secondary phase of adjuvant
fibrosis. J. Am. Soc. Nephrol. 19, 1459–1462 a comparison of selective and combined endothelin arthritis in rats. J. Rheumatol. 19, 1735–1742
(2008). receptor blockade. Circulation 109, 1186–1193 (1992).
56. Fujisawa, G. et al. Spironolactone prevents early renal (2004). 105. Zoja, C. et al. Bindarit retards renal disease and
injury in streptozotocin-induced diabetic rats. Kidney 81. Rubin, L. J. et al. Bosentan therapy for pulmonary prolongs survival in murine lupus autoimmune
Int. 66, 1493–1502 (2004). arterial hypertension. N. Engl. J. Med. 346, 896–903 disease. Kidney Int. 53, 726–734 (1998).
57. Han, K. H. et al. Spironolactone ameliorates renal (2002). 106. Guglielmotti, A. & Dionisio, P. A pharmaceutical
injury and connective tissue growth factor expression 82. Pierre-Paul, D. & Gahtan, V. Noncholesterol-lowering composition for the treatment of autoimmune
in type II diabetic rats. Kidney Int. 70, 111–120 effects of statins. Vasc. Endovascular Surg. 37, diseases. WO9716185 (1997).
(2006). 301–313 (2003). 107. Walport, M. J. Complement. First of two parts. N. Engl.
58. Rocha, R., Chander, P. N., Khanna, K., Zuckerman, A. 83. Li, C. et al. Pravastatin treatment attenuates J. Med. 344, 1058–1066 (2001).
& Stier, C. T. Jr. Mineralocorticoid blockade reduces interstitial inflammation and fibrosis in a rat model of 108. Gerard, N. P. & Gerard, C. The chemotactic receptor
vascular injury in stroke-prone hypertensive rats. chronic cyclosporine-induced nephropathy. Am. J. for human C5a anaphylatoxin. Nature 349, 614–617
Hypertension 31, 451–458 (1998). Physiol. Renal Physiol. 286, F46–F57 (2004). (1991).
59. Nagase, M. et al. Podocyte injury underlies the 84. Vieira, J. M., Jr et al. Simvastatin attenuates renal 109. Wang, Y. et al. Amelioration of lupus-like autoimmune
glomerulopathy of Dahl salt-hypertensive rats and is inflammation, tubular transdifferentiation and disease in NZB/WF1 mice after treatment with a
reversed by aldosterone blocker. Hypertension 47, interstitial fibrosis in rats with unilateral ureteral blocking monoclonal antibody specific for
1084–1093 (2006). obstruction. Nephrol. Dial. Transplant. 20, complement component C5. Proc. Natl Acad. Sci.
60. Aldigier, J. C., Kanjanbuch, T., Ma, L. J., Brown, N. J. 1582–1591 (2005). USA 93, 8563–8568 (1996).
& Fogo, A. B. Regression of existing 85. Massy, Z. A., Keane, W. F. & Kasiske, B. L. Inhibition of 110. Hillmen, P. et al. The complement inhibitor eculizumab
glomerulosclerosis by inhibition of aldosterone. J. Am. the mevalonate pathway: benefits beyond cholesterol in paroxysmal nocturnal hemoglobinuria. N. Engl. J.
Soc. Nephrol. 16, 3306–3314 (2005). reduction? Lancet 347, 102–103 (1996). Med. 355, 1233–1243 (2006).

nATURE REvIEwS | drug discovery vOlUME 7 | nOvEMbER 2008 | 951


REVIEWS

111. Cybulsky, A. V., Quigg, R. J. & Salant, D. J. 133. Jin, H. M. & Pan, Y. Renoprotection provided by 158. Caenepeel, S., Charydczak, G., Sudarsanam, S.,
Experimental membranous nephropathy redux. Am. losartan in combination with pioglitazone is superior Hunter, T. & Manning, G. The mouse kinome:
J. Physiol. Renal Physiol. 289, F660–F671 (2005). to renoprotection provided by losartan alone in discovery and comparative genomics of all mouse
112. Appel, G. et al. Eculizumab (C5 complement inhibitor) patients with type 2 diabetic nephropathy. Kidney protein kinases. Proc. Natl Acad. Sci. USA 101,
in the treatment of idiopathic membranous Blood Press. Res. 30, 203–211 (2007). 11707–11712 (2004).
nephropathy (IMN): preliminary baseline and 134. Drazen, J. M., Morrissey, S. & Curfman, G. D. 159. Obligado, S. H., Ibraghimov-Beskrovnaya, O., Zuk, A.,
pharmacokinetic (PK)/pharmacodynamic (PD) data. Rosiglitazone — continued uncertainty about safety. Meijer, L. & Nelson, P. J. CDK/GSK-3 inhibitors as
J. Am. Soc. Nephrol. 13, 668A (2002). N. Engl. J. Med. 357, 63–64 (2007). therapeutic agents for parenchymal renal diseases.
113. Yu, L., Border, W. A., Huang, Y. & Noble, N. A. 135. Goligorsky, M. S., Chaimovitz, C., Rapoport, J., Kidney Int. 73, 684–690 (2008).
TGF-β isoforms in renal fibrogenesis. Kidney Int. 64, Goldstein, J. & Kol, R. Calcium metabolism in uremic 160. Lee, M. J. et al. Indirubin-3′-monoxime, a derivative of
844–856 (2003). nephrocalcinosis: preventive effect of verapamil. a Chinese anti-leukemia medicine, inhibits Notch1
114. Massague, J. & Gomis, R. R. The logic of TGFβ Kidney Int. 27, 774–779 (1985). signaling. Cancer Lett. 265, 215–225 (2008).
signaling. FEBS Lett. 580, 2811–2820 (2006). 136. Schrier, R. W., Shapiro, J. I., Chan, L. & Harris, D. C. 161. Niranjan, T. et al. The Notch pathway in podocytes
115. Shihab, F. S., Bennett, W. M., Yi, H. & Andoh, T. F. Increased nephron oxygen consumption: potential plays a role in the development of glomerular disease.
Pirfenidone treatment decreases transforming growth role in progression of chronic renal disease. Am. J. Nature Med. 14, 290–298 (2008).
factor-β1 and matrix proteins and ameliorates fibrosis Kidney Dis. 23, 176–182 (1994). First documentation that the NOTCH pathway has
in chronic cyclosporine nephrotoxicity. Am. J. 137. Aschenbrenner, J. K., Sollinger, H. W., Becker, B. N. & a role in the development of glomerular disease
Transplant. 2, 111–119 (2002). Hullett, D. A. 1,25-(OH2)D3 alters the transforming and that γ-secretase inhibitors not only prevented
116. Cain, W. C. et al. Inhibition of tumor necrosis factor growth factor beta signaling pathway in renal tissue. the development of albuminuria, but also lowered
and subsequent endotoxin shock by pirfenidone. J. Surg. Res. 100, 171–175 (2001). established albuminuria in experimental animals
Int. J. Immunopharmacol. 20, 685–695 (1998). 138. Zhang, Z. et al. 1,25-Dihydroxyvitamin D3 targeting of with nephrotic syndrome.
117. Giri, S. N., Leonard, S., Shi, X., Margolin, S. B. & NF-κB suppresses high glucose-induced MCP-1 162. Pippin, J. W., Qu, Q., Meijer, L. & Shankland, S. J.
Vallyathan, V. Effects of pirfenidone on the generation expression in mesangial cells. Kidney Int. 72, 193–201 Direct in vivo inhibition of the nuclear cell cycle
of reactive oxygen species in vitro. J. Environ. Pathol. (2007). cascade in experimental mesangial proliferative
Toxicol. Oncol. 18, 169–177 (1999). 139. Li, Y. C. et al. 1,25-Dihydroxyvitamin D3 is a negative glomerulonephritis with Roscovitine, a novel cyclin-
118. Callahan, J. F. et al. Identification of novel inhibitors of endocrine regulator of the renin–angiotensin system. dependent kinase antagonist. J. Clin. Invest. 100,
the transforming growth factor beta1 (TGF-β1) type 1 J. Clin. Invest. 110, 229–238 (2002). 2512–2520 (1997).
receptor (ALK5). J. Med. Chem. 45, 999–1001 (2002). 140. Tan, X., Wen, X. & Liu, Y. Paricalcitol inhibits renal 163. Griffin, S. V., Krofft, R. D., Pippin, J. W. & Shankland,
119. Petersen, M. et al. Oral administration of GW788388, inflammation by promoting vitamin D receptor- S. J. Limitation of podocyte proliferation improves
an inhibitor of TGF-β type I and II receptor kinases, mediated sequestration of NF-κB signaling. J. Am. renal function in experimental crescentic
decreases renal fibrosis. Kidney Int. 73, 705–715 Soc. Nephrol. 19, 1741–1752 (2008). glomerulonephritis. Kidney Int. 67, 977–986 (2005).
(2008). 141. Levin, A. & Li, Y. C. Vitamin D and its analogues: do they 164. Gherardi, D. et al. Reversal of collapsing
An interesting demonstration of the renoprotective protect against cardiovascular disease in patients with glomerulopathy in mice with the cyclin-dependent
effect of a new TGF-β receptor kinase inhibitor in kidney disease? Kidney Int. 68, 1973–1981 (2005). kinase inhibitor CYC202. J. Am. Soc. Nephrol. 15,
db/db mice with potential clinical application. 142. Mizobuchi, M. et al. Combination therapy with an 1212–1222 (2004).
120. Gellibert, F. et al. Discovery of angiotensin-converting enzyme inhibitor and a vitamin 165. Zoja, C. et al. Cyclin-dependent kinase inhibition limits
4-{4-[3-(pyridin-2-yl)-1H-pyrazol-4-yl] D analog suppresses the progression of renal glomerulonephritis and extends lifespan of mice with
pyridin-2-yl}-N-(tetrahydro-2H- pyran-4-yl)benzamide insufficiency in uremic rats. J. Am. Soc. Nephrol. 18, systemic lupus. Arthritis Rheum. 56, 1629–1637
(GW788388): a potent, selective, and orally active 1796–1806 (2007). (2007).
transforming growth factor-β type I receptor inhibitor. 143. Panichi, V. et al. Effects of 1,25(OH2)D3 in 166. Benigni, A. et al. Involvement of renal tubular
J. Med. Chem. 49, 2210–2221 (2006). experimental mesangial proliferative nephritis in rats. Toll-like receptor 9 in the development of
121. Ziyadeh, F. N. et al. Long-term prevention of renal Kidney Int. 60, 87–95 (2001). tubulointerstitial injury in systemic lupus. Arthritis
insufficiency, excess matrix gene expression, and 144. Tan, X., Li, Y. & Liu, Y. Paricalcitol attenuates renal Rheum. 56, 1569–1578 (2007).
glomerular mesangial matrix expansion by treatment interstitial fibrosis in obstructive nephropathy. J. Am. 167. Nelson, P. J. & Shankland, S. J. Therapeutics in renal
with monoclonal antitransforming growth factor-beta Soc. Nephrol. 17, 3382–3393 (2006). disease: the road ahead for antiproliferative targets.
antibody in db/db diabetic mice. Proc. Natl Acad. Sci. 145. Agarwal, R. et al. Antiproteinuric effect of oral Nephron Exp. Nephrol. 103, e6–e15 (2006).
USA 97, 8015–8020 (2000). paricalcitol in chronic kidney disease. Kidney Int. 68, 168. Artavanis-Tsakonas, S., Rand, M. D. & Lake, R. J.
122. Shimizu, T. et al. Pirfenidone prevents collagen 2823–2828 (2005). Notch signaling: cell fate control and signal
accumulation in the remnant kidney in rats with partial 146. Alborzi, P. et al. Paricalcitol reduces albuminuria and integration in development. Science 284, 770–776
nephrectomy. Kidney Int. Suppl. 63, S239–S243 inflammation in chronic kidney disease: a randomized (1999).
(1997). double-blind pilot trial. Hypertension 52, 249–255 169. Wolfe, M. S. Therapeutic strategies for Alzheimer’s
123. Shimizu, T. et al. Pirfenidone improves renal function (2008). disease. Nature Rev. Drug Discov. 1, 859–866
and fibrosis in the post-obstructed kidney. Kidney Int. 147. Abdel Wahab, N. & Mason, R. M. Connective tissue (2002).
54, 99–109 (1998). growth factor and renal diseases: some answers, more 170. Shearman, M. S. et al. L-685,458, an aspartyl
124. Grygielko, E. T. et al. Inhibition of gene markers of questions. Curr. Opin. Nephrol. Hypertens. 13, 53–58 protease transition state mimic, is a potent inhibitor
fibrosis with a novel inhibitor of transforming growth (2004). of amyloid β-protein precursor γ-secretase activity.
factor-β type I receptor kinase in puromycin-induced 148. Burns, W. C. et al. Connective tissue growth factor Biochemistry 39, 8698–8704 (2000).
nephritis. J. Pharmacol. Exp. Ther. 313, 943–951 plays an important role in advanced glycation end 171. van Es, J. H. et al. Notch/γ-secretase inhibition turns
(2005). product-induced tubular epithelial-to-mesenchymal proliferative cells in intestinal crypts and adenomas
125. Benigni, A. et al. Add-on anti-TGF-β antibody to ACE transition: implications for diabetic renal disease. into goblet cells. Nature 435, 959–963 (2005).
inhibitor arrests progressive diabetic nephropathy in J. Am. Soc. Nephrol. 17, 2484–2494 (2006). 172. Vujovic, S., Henderson, S. R., Flanagan, A. M. &
the rat. J. Am. Soc. Nephrol. 14, 1816–1824 149. Okada, H. et al. Connective tissue growth factor Clements, M. O. Inhibition of γ-secretases alters both
(2003). expressed in tubular epithelium plays a pivotal role in proliferation and differentiation of mesenchymal stem
126. Cho, M. E., Smith, D. C., Branton, M. H., Penzak, S. R. renal fibrogenesis. J. Am. Soc. Nephrol. 16, 133–143 cells. Cell Prolif. 40, 185–195 (2007).
& Kopp, J. B. Pirfenidone slows renal function decline (2005). 173. Reff, M. E. et al. Depletion of B cells in vivo by a
in patients with focal segmental glomerulosclerosis. 150. Yokoi, H. et al. Reduction in connective tissue growth chimeric mouse human monoclonal antibody to CD20.
Clin. J. Am. Soc. Nephrol. 2, 906–913 (2007). factor by antisense treatment ameliorates renal Blood 83, 435–445 (1994).
127. Lebovitz, H. E. & Banerji, M. A. Insulin resistance and tubulointerstitial fibrosis. J. Am. Soc. Nephrol. 15, 174. Coiffier, B. et al. Rituximab (anti-CD20 monoclonal
its treatment by thiazolidinediones. Recent Prog. 1430–1440 (2004). antibody) for the treatment of patients with relapsing
Horm. Res. 56, 265–294 (2001). 151. Ostendorf, T. et al. Specific antagonism of PDGF or refractory aggressive lymphoma: a multicenter
128. Lebovitz, H. E., Kreider, M. & Freed, M. I. Evaluation of prevents renal scarring in experimental glomerulo- phase II study. Blood 92, 1927–1932 (1998).
liver function in type 2 diabetic patients during clinical nephritis. J. Am. Soc. Nephrol. 12, 909–918 (2001). 175. Maloney, D. G. Concepts in radiotherapy and
trials: evidence that rosiglitazone does not cause 152. Wang, S., Wilkes, M. C., Leof, E. B. & Hirschberg, R. immunotherapy: anti-CD20 mechanisms of action and
hepatic dysfunction. Diabetes Care 25, 815–821 Imatinib mesylate blocks a non-Smad TGF-β pathway targets. Semin. Oncol. 32, S19–S26 (2005).
(2002). and reduces renal fibrogenesis in vivo. FASEB J. 19, 176. Ruggenenti, P., Cravedi, P. & Remuzzi, G. Latest
129. Isshiki, K. et al. Thiazolidinedione compounds 1–11 (2005). treatment strategies for membranous nephropathy.
ameliorate glomerular dysfunction independent of 153. Zoja, C. et al. Imatinib ameliorates renal disease and Expert Opin. Pharmacother. 8, 3159–3171 (2007).
their insulin-sensitizing action in diabetic rats. survival in murine lupus autoimmune disease. Kidney 177. Cohen, C. D. et al. CD20-positive infiltrates in human
Diabetes 49, 1022–1032 (2000). Int. 70, 97–103 (2006). membranous glomerulonephritis. J. Nephrol. 18,
130. Panchapakesan, U., Pollock, C. A. & Chen, X. M. The 154. Lassila, M. et al. Imatinib attenuates diabetic 328–333 (2005).
effect of high glucose and PPAR-γ agonists on PPAR-γ nephropathy in apolipoprotein E-knockout mice. 178. Remuzzi, G. et al. Rituximab for idiopathic membranous
expression and function in HK-2 cells. Am. J. Physiol. J. Am. Soc. Nephrol. 16, 363–373 (2005). nephropathy. Lancet 360, 923–924 (2002).
Renal Physiol. 287, F528–F534 (2004). 155. Floege, J., Eitner, F. & Alpers, C. E. A new look at First identification of a novel, specific therapy for
131. Ohga, S. et al. Thiazolidinedione ameliorates renal platelet-derived growth factor in renal disease. J. Am. idiopathic membranous nephropathy.
injury in experimental diabetic rats through anti- Soc. Nephrol. 19, 12–23 (2008). 179. Ruggenenti, P. et al. Rituximab for idiopathic
inflammatory effects mediated by inhibition of NF-κB 156. Kerkela, R. et al. Cardiotoxicity of the cancer membranous nephropathy: who can benefit? Clin. J.
activation. Am. J. Physiol. Renal Physiol. 292, therapeutic agent imatinib mesylate. Nature Med. 12, Am. Soc. Nephrol. 1, 738–748 (2006).
F1141–F1150 (2007). 908–916 (2006). 180. Fervenza, F. et al. Variation in response to rituximab in
132. Miyazaki, Y., Cersosimo, E., Triplitt, C. & DeFronzo, 157. Berman, E. et al. Altered bone and mineral the treatment of idiopathic membranous nephropathy
R. A. Rosiglitazone decreases albuminuria in type 2 metabolism in patients receiving imatinib mesylate. (IMN): preliminary results at 1 year. J. Am. Soc.
diabetic patients. Kidney Int. 72, 1367–1373 (2007). N. Engl. J. Med. 354, 2006–2013 (2006). Nephrol. 14, A570 (2006).

952 | nOvEMbER 2008 | vOlUME 7 www.nature.com/reviews/drugdisc


REVIEWS

181. Barsoum, R. S. Hepatitis C virus: from entry to renal 193. Morigi, M. et al. Human bone marrow mesenchymal 206. Kalluri, R. & Neilson, E. G. Epithelial-mesenchymal
injury — facts and potentials. Nephrol. Dial. stem cells accelerate recovery of acute renal injury and transition and its implications for fibrosis. J. Clin.
Transplant. 22, 1840–1848 (2007). prolong survival in mice. Stem Cells 26, 2075–2082 Invest. 112, 1776–1784 (2003).
182. Roccatello, D. et al. Long-term effects of anti-CD20 (2008). 207. Maschio, G. et al. Effect of the angiotensin-
monoclonal antibody treatment of cryoglobulinaemic 194. Fabry, J. Ein beitrag zur kenntnis der purpura converting-enzyme inhibitor benazepril on the
glomerulonephritis. Nephrol. Dial. Transplant. 19, hemorrhagica nodularis (purpura papulosa progression of chronic renal insufficiency.
3054–3061 (2004). hemorrhagica hebrae). Arch. Derm. Syphilol. 43, The Angiotensin-Converting-Enzyme Inhibition in
183. Sfikakis, P. P. et al. Remission of proliferative lupus 187–200 (1898). Progressive Renal Insufficiency Study Group.
nephritis following B cell depletion therapy is 195. Schiffmann, R. et al. Enzyme replacement therapy in N. Engl. J. Med. 334, 939–945 (1996).
preceded by down-regulation of the T cell Fabry disease: a randomized controlled trial. JAMA 208. Soos, T. J., Meijer, L. & Nelson, P. J. CDK/GSK-3
costimulatory molecule CD40 ligand: 285, 2743–2749 (2001). inhibitors as a new approach for the treatment of
an open-label trial. Arthritis Rheum. 52, 501–513 A key study showing the efficacy of proliferative renal diseases. Drug News Perspect. 19,
(2005). enzyme-replacement therapy with recombinant 325–328 (2006).
184. van Vollenhoven, R. F. et al. Biopsy-verified α-galactosidase A for Fabry’s disease. 209. Nelson, P. J., D’Agati, V. D., Gries, J. M., Suarez, J. R.
response of severe lupus nephritis to treatment 196. Thurberg, B. L. et al. Globotriaosylceramide & Gelman, I. H. Amelioration of nephropathy in mice
with rituximab (anti-CD20 monoclonal antibody) accumulation in the Fabry kidney is cleared from expressing HIV-1 genes by the cyclin-dependent
plus cyclophosphamide after biopsy-documented multiple cell types after enzyme replacement therapy. kinase inhibitor flavopiridol. J. Antimicrob. Chemother.
failure to respond to cyclophosphamide alone. Kidney Int. 62, 1933–1946 (2002). 51, 921–929 (2003).
Scand. J. Rheumatol. 33, 423–427 (2004). 197. Eng, C. M. et al. Fabry disease: guidelines for the 210. Milovanceva-Popovska, M. et al. R-roscovitine
185. Gottenberg, J. E. et al. Tolerance and short term evaluation and management of multi-organ system (CYC202) alleviates renal cell proliferation in nephritis
efficacy of rituximab in 43 patients with systemic involvement. Genet. Med. 8, 539–548 (2006). without aggravating podocyte injury. Kidney Int. 67,
autoimmune diseases. Ann. Rheum. Dis. 64, 913–920 198. Torres, V. E., Harris, P. C. & Pirson, Y. Autosomal 1362–1370 (2005).
(2005). dominant polycystic kidney disease. Lancet 369, 211. Lin, C. L. et al. Wnt/β-catenin signaling modulates
186. Remuzzi, A. et al. ACE inhibition reduces 1287–1301 (2007). survival of high glucose-stressed mesangial cells.
glomerulosclerosis and regenerates glomerular tissue 199. Torres, V. E. & Harris, P. C. Mechanisms of Disease: J. Am. Soc. Nephrol. 17, 2812–2820 (2006).
in a model of progressive renal disease. Kidney Int. autosomal dominant and recessive polycystic kidney 212. Bukanov, N. O., Smith, L. A., Klinger, K. W.,
69, 1124–1130 (2006). diseases. Nature Clin. Pract. Nephrol. 2, 40–55 Ledbetter, S. R. & Ibraghimov-Beskrovnaya, O.
187. Fioretto, P., Steffes, M. W., Sutherland, D. E., Goetz, (2006). Long-lasting arrest of murine polycystic kidney
F. C. & Mauer, M. Reversal of lesions of diabetic 200. Ruggenenti, P. et al. Safety and efficacy of long-acting disease with CDK inhibitor roscovitine. Nature 444,
nephropathy after pancreas transplantation. N. Engl. somatostatin treatment in autosomal-dominant poly- 949–952 (2006).
J. Med. 339, 69–75 (1998). cystic kidney disease. Kidney Int. 68, 206–216 (2005).
188. Ruggenenti, P. et al. In chronic nephropathies 201. Shillingford, J. M. et al. The mTOR pathway is Acknowledgements
prolonged ACE inhibition can induce remission: regulated by polycystin-1, and its inhibition reverses We are deeply grateful to P. Ruggenenti, M. Abbate and
dynamics of time-dependent changes in GFR. renal cystogenesis in polycystic kidney disease. B. Bikbov for their suggestions and criticisms. We also
Investigators of the GISEN Group. Gruppo Italiano Proc. Natl Acad. Sci. USA 103, 5466–5471 (2006). thank F. Gaspari for technical assistance in preparing the
Studi Epidemiologici in Nefrologia. J. Am. Soc. 202. Tao, Y., Kim, J., Schrier, R. W. & Edelstein, C. L. manuscript.
Nephrol. 10, 997–1006 (1999). Rapamycin markedly slows disease progression in a
189. Fogo, A. B. New capillary growth: a contributor to rat model of polycystic kidney disease. J. Am. Soc. FURTHER INFORMATION
regression of sclerosis? Curr. Opin. Nephrol. Nephrol. 16, 46–51 (2005).
ClinicalTrials.gov: http://clinicaltrials.gov
Hypertens. 14, 201–203 (2005). 203. Walz, G. Therapeutic approaches in autosomal
FibroGen Clinical Trials: http://www.fibrogen.com/trials
190. Oliver, J. A., Maarouf, O., Cheema, F. H., Martens, T. P. dominant polycystic kidney disease (ADPKD): is
National Monitoring Centre for Clinical Trials:
& Al-Awqati, Q. The renal papilla is a niche for adult there light at the end of the tunnel? Nephrol. Dial.
http://ossc-svil.test.cineca.it/index_ingl.htm
kidney stem cells. J. Clin. Invest. 114, 795–804 Transplant. 21, 1752–1757 (2006).
NKF, FDA Host Conference to Assess Proteinuria as
(2004). 204. Nakamura, T., Miller, D., Ruoslahti, E. & Border, W. A.
191. Bahlmann, F. H. et al. Stimulation of endothelial Production of extracellular matrix by glomerular a Surrogate Outcome in Chronic Kidney Disease:
progenitor cells: a new putative therapeutic effect of epithelial cells is regulated by transforming growth http://www.kidney.org/professionals/physicians/
angiotensin II receptor antagonists. Hypertension 45, factor-β1. Kidney Int. 41, 1213–1221 (1992). proteinuria.cfm
526–529 (2005). 205. Laiho, M., Saksela, O. & Keski-Oja, J. Transforming
192. Morigi, M. et al. Mesenchymal stem cells are growth factor-β induction of type-1 plasminogen SUPPLEMENTARY INFORMATION
renotropic, helping to repair the kidney and improve activator inhibitor. Pericellular deposition and See online article: S1 (box) | S2 (box) | S3 (table)
function in acute renal failure. J. Am. Soc. Nephrol. sensitivity to exogenous urokinase. J. Biol. Chem. All liNKs Are AcTive iN The oNliNe PdF
15, 1794–1804 (2004). 262, 17467–17474 (1987).

nATURE REvIEwS | drug discovery vOlUME 7 | nOvEMbER 2008 | 953

You might also like