throbber
Maturitas 71 (2012) 94– 103
`
`Contents lists available at SciVerse ScienceDirect
`
`Maturitas
`
`j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / m a t u r i t a s
`
`Review
`Chronic kidney disease and diabetes
`Ronald Pyram a,b, Abhishek Kansara a,b,c, Mary Ann Banerji a,c, Lisel Loney-Hutchinson a,c,∗
`
`a Division of Endocrinology SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, United States
`b Division of Endocrinology Brooklyn VA Medical Center, Brooklyn, NY, United States
`c Kings County Hospital Center, Department of Medicine, 451 Clarkson Ave, Brooklyn, NY 11203, United States
`
`a
`
`
`
`r
`
`
`
`t
`
`
`
`i
`
`
`
`c
`
`
`
`l
`
`
`
`e
`
`
`
`i
`
`
`
`n
`
`
`
`f
`
`
`
`o
`
`a
`
`
`
`b
`
`
`
`s
`
`
`
`t
`
`
`
`r
`
`
`
`a
`
`
`
`c
`
`
`
`t
`
`Article history:
`Received 28 October 2011
`Received in revised form 9 November 2011
`Accepted 9 November 2011
`
`Keywords:
`Chronic renal disease
`Diabetes
`Glomerular filtration rate
`Nephropathy
`Renal failure
`
`Contents
`
`Chronic kidney disease has a significant worldwide prevalence affecting 7.2% of the global adult popu-
`lation with the number dramatically increasing in the elderly. Although the causes are various, diabetes
`is the most common cause of CKD in the United States and an increasing cause of the same worldwide.
`Therefore, we chose to focus on diabetic chronic kidney disease in this review.
`The pathogenesis is multifactorial involving adaptive hyperfiltration, advanced glycosylated end-
`product synthesis (AGES), prorenin, cytokines, nephrin expression and impaired podocyte-specific insulin
`signaling. Treatments focus on lifestyle interventions including control of hyperglycemia, hypertension
`and hyperlipidemia as well treatment of complications and preparation for renal replacement ther-
`apy. This review examines the current literature on the epidemiology, pathogenesis, complications and
`treatment of CKD as well as possible areas of future disease intervention.
`Published by Elsevier Ireland Ltd.
`
`1.
`
`2.
`
`3.
`
`4.
`
`94
`Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`1.1.
`95
`Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`95
`Diabetic nephropathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`2.1.
`95
`Risk factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`2.2.
`96
`Pathological disease progression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`2.3. Mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`96
`2.4.
`97
`Complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`97
`Evidence of modifiable risk factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.1.
`97
`Diet, weight reduction, exercise and smoking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.2.
`Blood glucose control. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`98
`3.3.
`98
`Blood pressure control . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.4. Management of complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`98
`3.5. Medication management of hypertension in patients with CKD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`99
`3.6.
`Preparation for initiation of renal replacement therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
`Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
`Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
`Competing interests. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
`Provenance and peer review . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
`References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
`
`∗ Corresponding author at: Kings County Hospital Center, Department of
`Medicine, 451 Clarkson Ave, Brooklyn, NY 11203, United States.
`Tel.: +1 718 270 6324.
`E-mail address: lisel.loney-hutchinson@downstate.edu (L. Loney-Hutchinson).
`
`0378-5122/$ – see front matter. Published by Elsevier Ireland Ltd.
`doi:10.1016/j.maturitas.2011.11.009
`
`1. Introduction
`
`Chronic kidney disease (CKD) affects a significant portion of the
`world population with a prevalence of 7.2% in adults over age 30
`and dramatically increasing to 23.4–35.8% over age 65 [1]. Further-
`more, population based studies in the United States reveals a CKD
`prevalence ranging from 1.4 to 43.3% [2]. The causes of CKD are
`
`MPI EXHIBIT 1145 PAGE 1
`
`

`

`R. Pyram et al. / Maturitas 71 (2012) 94– 103
`
`95
`
`various and include glomerular kidney disease, tubular and inter-
`stitial kidney disease, obstructive uropathy, pre renal and vascular
`disorders, diabetes and hypertension. Globally, diabetes is the most
`common cause of CKD.
`There are multiple etiologies involved in the pathogenesis of
`diabetic CKD. The initial mechanism of damage involves adaptive
`hyperfiltration which leads to long term damage of functioning
`nephrons. Additional mechanisms of disease include advanced
`glycosylated end products (AGEs), vascular endothelial growth fac-
`tor (VEGF), prorenin and the renin–angiotensin system, cytokines,
`nephrin expression and impaired podocyte signaling. Importantly,
`CKD is associated with an increased risk of cardiovascular disease,
`mortality and end stage renal disease. Most patients are likely to
`die than develop end stage renal disease. Treatments are limited
`but focus on treatment of associated causes including diabetes and
`hypertension, slowing progression of disease, treatment of compli-
`cations and preparation for renal replacement therapy. This review
`examines the current literature on the epidemiology, pathogene-
`sis, complications and treatment of CKD as well as possible areas of
`future disease intervention.
`
`1.1. Epidemiology
`
`Diabetes mellitus is a disease that affects over 23.5 million
`American adults [3] and type 2 diabetes accounts for over 90%
`[4,5]. Diabetes is the most common cause of CKD. The prevalence
`of some degree of CKD among adults with type 2 diabetes is 40%
`[3,6–11]. The 2009 annual report published by the United States
`Renal Data Systems, reports the prevalence rate of CKD in type 2
`diabetes according to stage was 8.9% for stage I, 12.8% for stage
`II, 19.4% for stage III, and 2.7% for stages IV and V combined. The
`lifetime risk of developing CKD in type 1 diabetes is 25% [5,12–17].
`The older population is especially affected by diabetic nephropa-
`thy. The incidence of end stage renal disease (ESRD) related to
`diabetes among the elderly markedly increased from the 1980s
`up until the late 1990s at which time there was slight decrease
`and leveling off to approximately 350–370/100,000 [4,18,19]. This
`more recent decline in CKD is likely due to earlier recognition and
`interventions.
`In the 2010 Annual report published by the United States Renal
`Data Systems, the median age of incident ESRD population was
`64.2 in 2008 which varied by race; the median age ranged from
`59.2 among African Americans to 66.8 among whites. The rate of
`prevalence of CKD remains highest among the African American
`and Native American population, and lowest among the Caucasian
`and Asian population paralleling the rate of diabetes in these pop-
`ulations [20,21]. The trend of increasing prevalence is expected to
`continue to grow as the elderly population continues to rise with
`increased life expectancy.
`CKD in diabetes causes significant disability and contributes
`greatly to health care costs; annually approaching $23 billion with
`ESRD in the US [22]. McFarlane et al. [23] evaluated the risk of hos-
`pitalization, cardiovascular risk and death in patients with chronic
`kidney disease and reported that the adjusted risk of hospitaliza-
`tion increased as the estimated glomerular filtration rate (eGFR)
`decreased ranging from an increase of 14% with an estimated GFR
`of 45 to 59 ml/min/1.73 m2 to an increase of 315% with an estimated
`GFR of less than 15 ml/min/1.73 m2.
`
`2. Diabetic nephropathy
`
`CKD
`in kidney
`loss
`irreversible
`is defined as progressive,
`function. The National Kidney Foundation (NKF) Kidney Disease
`Outcomes Quality Initiative (K/DOQI) defined CKD as lasting three
`or more months with either kidney damage defined by structural or
`
`Table 1
`Stages of chronic kidney disease.
`
`Stage
`
`Description
`
`eGFR (ml/min/1.73 m2)
`
`I
`II
`III
`IV
`V
`
`Normal or increased eGFRa
`Mildly decreases eGFRa
`Moderately reduced eGFR
`Severely reduced eGFR
`Kidney failure
`
`>90
`60–89
`30–59
`15–29
`<15 or dialysis
`
`a With evidence of structural kidney damage such as albuminuria, abnormal uri-
`nary sediment (i.e. casts, tubular epithelial cells), abnormal imaging studies, renal
`transplant recipients [26].
`
`functional abnormalities of the kidney with or without decreased
`eGFR, or a GFR of less than 60 ml/min/1.73 m2. These abnormal-
`ities include markers of kidney damage [1] such as albuminuria,
`abnormal urinary sediment (casts, epithelial cells), abnormal imag-
`ing (polycystic kidneys, hydronephrosis), blood and urine markers
`of ‘tubular syndromes’ and renal transplant recipients. In addi-
`tion to the gold standard isotopic measures, several equations
`are used to estimate GFR. The Modification of Diet in Renal Dis-
`ease (MDRD) equation [1], validated in patients with diabetic
`(type 2) and non-diabetic kidney disease and in transplant recip-
`ients is most commonly used. It is verified in US and European
`whites and African-Americans, but not with other racial/ethnic
`groups, extremes of age, pregnancy and certain other conditions
`[1]. The Chronic Kidney Disease Epidemiology Collaboration (CKD-
`EPI) equation is a new estimate of glomerular filtration and may be
`more accurate [24,25] at higher levels of eGFR.
`Stages of chronic kidney disease: The stages of chronic kidney
`disease as described in Table 1 are defined on the basis of eGFR [26]
`with evidence of kidney damage.
`
`2.1. Risk factors
`• Elevated blood pressure: Elevated systolic blood pressure is well
`known to accelerate diabetic nephropathy [27–29]. The devel-
`opment of impaired renal function was associated with higher
`mean blood pressures even in patients who were normotensive
`with and without proteinuria [28] in long-term follow-up of type
`2 diabetes patients.
`• Diabetes: Strong evidence for the role of glycemia was provided
`by the DCCT (type 1 diabetes) and the UKPDS (type 2 diabetes)
`intervention trials where improved glycemia resulted in signifi-
`cantly lower progression rates for albuminuria [30]. The duration
`of diabetes is associated with progression of nephropathy in long-
`term follow-up in Saudis with duration of diabetes >10 years
`[31]. The odds of nephropathy were 4.6-fold higher in urban
`African-Americans with duration of diabetes greater than 5 years
`as compared to <1 year [32].
`• Cholesterol: Elevated levels are associated with increased risk of
`nephropathy [26].
`• Microalbuminuria: The prevalence of microalbuminuria
`in
`South
`Indian diabetics was 36%, similar to urban African-
`Americans with type 2 diabetes for <1 year [32,33]. Microal-
`buminuria predicts progression to clinical proteinuria and
`decreased renal function in patients with type 2 diabetes [34–36]
`as well as increased CVD and mortality [30,37,38]. The rate
`of progression in UKPDS, from diagnosis of diabetes without
`microalbuminuria to microalbuminuria is 2% per year; from
`microalbuminuria to macroalbuminuria is 2.8% per year; and
`macroalbuminuria to elevated creatinine or renal replacement
`therapy
`is about 2.3% per year. Patients without microal-
`buminuria at diagnosis remained free of nephropathy for a
`median period of about 19 years, while those who developed
`microalbuminuria progressed to macroalbuminuria (or worse)
`in 11 years [39]. Microalbuminuria is infrequently reversible to
`
`MPI EXHIBIT 1145 PAGE 2
`
`

`

`96
`
`R. Pyram et al. / Maturitas 71 (2012) 94– 103
`
`Table 2
`Effects of RAAS in pathogenesis of diabetic nephropathy.
`
`Aldosterone
`
`Profibrotic
`
`Mitogenic
`
`Angiotensin II
`
`↑
`
`Production of reactive
`oxygen species
`↓ Nitric oxide production
`↑ Glomerular hypertension
`↑ Vascular endothelial growth
`factor
`
`Prorenin
`
`Elevated in type 1 diabetics
`with microalbuminuria
`
`increased VCAM expression causing vascular injury, mesangial
`cell growth, enhanced expression of growth factors, extracellu-
`lar membrane proteins, ROS and activation of protein kinase C,
`releasing cytokines and growth factors.
`• Protein kinase C (PKC): Hyperglycemia activates isoforms of
`protein kinase C through diacylglycerol (DAG), which activates
`MAP kinase and vasotropic substances such as angiotensin II,
`endothelin and prostanoids causing glomerular hyperfiltration.
`PKC activation increases ROS and the actions of fibrotic factors
`such as TGF-beta and connective tissue growth factor (CGTF)
`resulting in glomerular hypertrophy and mesangial expansion.
`ROS increases cytokines and extracellular membrane proteins
`type IV collagen leading to glomerulosclerosis and renal failure
`[59].
`• Vascular endothelial growth factor (VEGF) expression
`in
`podocytes is upregulated by hyperglycemia, increasing vascular
`permeability in the nephron [60].
`• Aldose reductase pathway: Its role in CKD is unclear [61,62].
`
`Nephrin: Nephrin, a protein found in podocytes is crucial in
`maintaining an intact filtration barrier. Lower renal expression of
`nephrin in kidney biopsies of patients with diabetes is reported
`[63].
`mTOR (mammalian Target Of Rapamycin) is a serine/threonine
`protein kinase which integrates multiple signals including insulin,
`energy balance and oxidative stress and regulates cell growth
`and survival. In glomeruli from patients with diabetic nephropa-
`thy, mTOR activated target genes were increased including VEGF,
`SREBP, mitochondrial genes as well as mTOR mRNA [64]. mTOR may
`play a role in glomerular hypertrophy and podocyte enlargement.
`Renin–Angiotensin System Aldosterone (RAAS) (see Table 2):
`
`• Prorenin: Increased serum prorenin levels precede and predict
`the onset of microalbuminuria in normotensive type 1 diabetes
`[65]. Levels were also higher in non-diabetic siblings of these
`patients [66,67] and may be a marker for increased risk of
`nephropathy in non-diabetic patients at high risk for diabetes.
`• Angiotensin II (Ang II) Ang II stimulates synthesis of matrix
`proteins, increases VEGF, oxidative stress, and decreases NO pro-
`duction and loss of endothelial integrity. Ang II causes efferent
`arteriolar vasoconstriction and increased intraglomerular pres-
`sure leading to renal hyperfusion [68,69].
`• Aldosterone is known to be mitogenic and increases renal fibro-
`sis through profibrotic TGF-beta [70,71].
`
`Microalbuminuria: Elevated levels of inflammatory markers
`are proportional to the degree of albuminuria [68]. In patients with
`type 2 diabetes and persistent microalbuminuria, elevated levels
`of biomarkers of inflammation and endothelial dysfunction pre-
`dicted development of nephropathy over a 2-year follow-up period
`[72]. Inflammatory factors lead to accumulation of macrophages
`in the tubular interstitium, producing free radicals, inflammatory
`cytokines and proteases that induce tubular damage [18,71].
`
`normoalbuminuria in type 2 diabetes (21% over 2 years) [40]. In
`contrast, this occurs in up to 50% of type 1 diabetes with short
`duration microalbuminuria [41]. Thus, although microalbumin-
`uria precedes proteinuria and decreased renal function, not all
`microalbiminuria will progress. The challenge is in identifying
`those with microalbuminuria who are likely to progress from
`those who will not. The GFR may also decline in the absence of
`any microalbuminuria [36,42].
`• Smoking: In types 1 and 2 diabetes, albuminuria was greater in
`smokers [43].
`• Genetic factors:
` An insertion /deletion polymorphism (specif-
`ically the deletion allele) of the ACE gene predicted severe
`structural kidney changes in patients with microalbuminuria
`[44]. Not all studies confirm this. Identifying the genetic risk of
`diabetic nephropathy [45] requires further study.
`• Age and BMI:
` Advancing age and obesity are also risk factors [28].
`
`2.2. Pathological disease progression
`
`Classical hemodynamic and structural causes of diabetic
`nephropathy, best characterized in type 1 diabetes are summarized
`below [46–49]:
`• Glomerular hyperfiltration: Vasodilatation and glomerular
`hyperfiltration, resulting in increased glomerular filtration rate,
`are known to occur early in type 1 diabetes, but not always in type
`2 diabetes [46,49–52]. While hyperfiltration frequently predicts a
`decline in renal function, it is not invariable.
`• Glomerular lesions without clinical disease: This stage
`is
`characterized by glomerular lesions-glomerular basement mem-
`brane thickening and mesangial expansion; without excess
`albumin excretion. Typical Kimmelstiel–Wilson lesions (nodular
`glomerulosclerosis) are found in only a small proportion early in
`nephropathy [49].
`• Incipient diabetic nephropathy: This stage develops after about
`10–15 years of type 1 diabetes characterized by microalbu-
`minuria and structural lesions [53,54]; the GFR may be well
`preserved. It is of clinical interest because interventions, espe-
`cially blood pressure control may prevent progression to overt
`nephropathy.
`• Overt diabetic nephropathy: There is persistent and progres-
`sively worsening proteinuria, a decline in GFR, frequently leading
`to ESRD. This stage is characterized by advanced glomerular
`lesions-diffuse and nodular glomerulosclerosis, fibrinoid caps,
`capsular drops and arteriolar hyalinosis [49].
`
`2.3. Mechanisms
`
`The mechanisms by which diabetes induces renal damage are
`not adequate to formulate a cohesive model of nephropathy.
`Various physical and metabolic factors together result in mesan-
`gial hypertrophy, glomerular basement thickening, podocyte and
`endothelial dysfunction.
`Glomerular hyperfiltration
`[55–57]: Various mediators
`include
`system, vascular endothelial
`the
`renin–angiotensin
`growth
`factor (VEGF), nitric oxide and transforming growth
`factor-beta (TGF-B).
`Hyperglycemia: Hyperglycemia is linked to multiple metabolic
`perturbations (Fig. 1).
`• Advanced glycosylated end-products (AGEs) are covalently
`glycosylated proteins [58] whose synthesis increases with hyper-
`glycemia. These accumulate in the extracellular matrix and the
`glomerular basement membrane altering the elasticity, ionic
`charge and thickness [59]. AGEs bind with cell surface receptors
`(RAGE) and initiate cellular signaling cascades associated with
`
`MPI EXHIBIT 1145 PAGE 3
`
`

`

`R. Pyram et al. / Maturitas 71 (2012) 94– 103
`
`97
`
`Hyperglycemia
`
`se Reducta
`Aldo
`
`
`Pathway
`
`se
`
`d Glycation
`Advance
`
`
`VCAM-1
`
`s
`
` End-product
`
`
`RAGERAGE
`
`Protein Kinase C
`
`DAG
`
`Diminished
`NO
`production
`
`Increased AGEs
`
`Increased
`prostaglandins
`
`Increased PKC
`
`Abnormal
`glomerular
`
`filtrati
`on
`
`
`
`
` injury
`
`Vascular
`
`
`Accumulation
`in ECM,
`
`
`
`GGBM
`
` elasticity,
`Altered
`
`
`ionic charge,ionic charge,
`
` of GBM
`thickness
`
`
`
`Renal dysfunctionRenal dysfunction
`
`
`
`
`
`AGE-RAGE
`-mesangial
` cell growth
`
`-stimulati
`
`on of GFs
`
`
`-ROS activation
`releasing
` cytokines
`
`
`Fibrotic factors
`
`
`
`
`Vasotropic
`substances
`-Ang,
`VEGF,
`PGE2
`
`ROS
`
`Increased
`cytokines,
`ECM
`proteins
`
`Glomerular
`hyperfiltratio
`n
`
`
`GlomerularGlomerular
`
`n,
`hyperfiltratio
`
`mesangial
`expansion
`
`Glomerulosclerosis
`
`Fig. 1. Pathways of hyperglycemia causing renal damage. AR: aldose reductase; NO: nitric oxide; AGEs: advanced end-glycation products; PKC: protein kinase C; VCAM:
`1-vascular cell adhesion molecule-1; RAGE: receptor for AGE; ECM: extracellular matrix; GBM: glomerular basement membrane; GFs: growth factors; ROS: reactive oxygen
`species; Ang: angiotensin; VEGF: vascular endothelial growth factor; PGE2: prostaglandin E2; DAG: diacylglycerol.
`
`2.4. Complications
`
`Anemia: Nearly 1 in 5 patients with stage III chronic kidney
`disease have anemia and likely due to decreased erythropoietin.
`Anemia is associated with a poor quality of life, increased fatigue,
`weakness, cognitive dysfunction, memory impairment and pro-
`gression of renal disease (ESRD) and increased mortality from CVD
`[73–76].
`Cardiovascular disease: As shown in Fig. 2, the prevalence
`of atherosclerotic vascular disease, congestive heart failure, renal
`replacement therapy and death was significantly higher in patients
`with both CKD and diabetes (49.1%, 52.3%, 3.4%, and 19.9%, respec-
`tively) compared with patients with neither underlying disease
`[77] in 1 million US elderly Medicare patients. The devastating CV
`consequences of CKD have been confirmed in numerous studies
`[78,79].
`Bone disease: Renal osteodystrophy or chronic kidney disease-
`mineral and bone disorder (CKD-MBD) encompasses all disorders
`of bone and mineral metabolism that are associated with CKD.
`
`CHF
`
`PVD
`
`ASCVD
`
`RRT
`
`Death
`
`60
`
`50
`
`40
`
`30
`
`20
`
`10
`
`0
`
`2000-2001/ 100 patient-years
`
`Incident events:
`
`The spectrum ranges from high bone turnover secondary to ele-
`vated PTH levels (secondary hyperparathyroidism) to those with
`low bone turnover associated with normal-to-low PTH levels (ady-
`namic bone disease, osteomalacia), including abnormal mineral
`metabolism, altered bone structure and composition, and extra-
`skeletal calcification.
`Secondary hyperparathyroidism ensues with a decline in GFR,
`secondary phosphorus retention, hypocalcemia and
`impaired
`1,25-dihydroxyvitamin-D production [80]. FGF-23 plays a key
`role in renal phosphate excretion and homeostasis; and sup-
`presses 1,25(OH)2-vitamin D3. Persistent hyperphosphatemia and
`1,25(OH)2-vitamin D3 are principal stimuli for its production. In
`patients with CKD, FGF-23 levels increase in response to phosphate
`retention which in turn suppresses 1-alpha-hydroxylase and renal
`synthesis of 1,25(OH)2-vitamin D3 which then leads to increased
`secretion of PTH. FGF-23 has been positively associated with left
`ventricular hypertrophy and CV mortality, especially in patients
`with low eGFR [81,82]. Recently, Klotho has been identified as a cru-
`cial cofactor essential for the biological effect of FGF-23 [80]. CKD
`may be a state of Klotho deficiency [83], with the lowest values in
`CKD stage V patients. Klotho protein may have anti-apoptotic and
`anti-senescent effects on endothelial cells including effects on vas-
`cular calcifications, endothelial dysfunction and kidney injury and
`repair. The ultimate role of FGF23 and its cofactor, Klotho protein,
`await further study.
`The prevalence of adynamic bone disease or bone’s resis-
`tance to the action of PTH ranges from 5% to 70% with a higher
`prevalence
`in advanced stages of CKD [84]. The CKD milieu
`promotes low bone formation. This milieu includes altered vitamin-
`D/calcium/phosphorus metabolism, acidosis, diabetes, age and
`AGEs (which increase circulating cytokines) [85–89].
`
`3. Evidence of modifiable risk factors
`
`3.1. Diet, weight reduction, exercise and smoking
`
`Epidemiological studies reveal a relationship between lifestyle
`variables including diet, obesity, exercise and smoking and CKD.
`
`DM-/CKD-
`
`DM+/CKD-
`
`DM-/CKD+
`
`DM+/CKD+
`
`
`Fig. 2. Incident event rates in 2000–2001 in over 1 million elderly US Medicare ben-
`eficiaries (over age 67 years). CHF: congestive heart failure; PVD: peripheral vascular
`disease; ASCVD: atherosclerotic cardiovascular disease; RRT: renal replacement
`therapy; DM: diabetes mellitus; CKD: chronic kidney disease.
`
`MPI EXHIBIT 1145 PAGE 4
`
`

`

`98
`
`R. Pyram et al. / Maturitas 71 (2012) 94– 103
`
`moderate blood pressure group. This was not the case in patients
`who had already displayed overt macroalbuminuria at baseline, as
`their renal function continued to decline regardless of which blood
`pressure group they were enrolled in [106].
`Further data from the ABCD study using hypertensive patients,
`treated also with Nisoldipine or Enalapril (switched to solely
`Enalapril during course of trial due to its cardiovascular bene-
`fit). After 5-year follow up, the intensive (mean BP 133/78) and
`moderate (mean BP 139/86) groups’ results were similar to the
`prior mentioned normotensive study arm. Renal function remained
`stable for those who had normoalbuminuria or microalbumin-
`uria at baseline, independent of initial therapy (Nisoldipine or
`Enalapril). Once again, those with overt macroalbuminuria con-
`tinued to show decline in renal function, with the rate of decline
`of about 5 ml/min/year. This decline was still markedly less than
`untreated hypertensives (10–12 ml/min/year), hence, very much
`suggestive of a significant benefit even in more advance CKD [106].
`Ravid et al. conducted a 5-year randomized study of 94 type
`2 diabetes with normal blood pressure and renal function but
`with microalbuminuria. Even in these normotensive patients there
`was a benefit of tighter blood pressure control with enalapril
`on renal function. In the enalapril group, the initial 24 h urine
`microalbumin of 143 mg/24 h initially decreased to a mean of
`123 mg/24 h, but rose to 140 mg/24 h after 5 years of treatment.
`In contrast, the placebo group began with a mean microalbumin
`level of 123 mg/24 h with an astonishing increase in microal-
`buminuria to a mean of 310 mg/24 h [107]. Additionally, there
`were significant differences in kidney function which decreased
`by 13% in the placebo group over the course of 5 years, while
`remaining stable in the enalapril group. These findings lead to
`the conclusion that intensive blood pressure control is benefi-
`cial for renal protection, especially when started early and may
`be less valuable in advanced CKD. Currently, the recommenda-
`tions from the American Society of Hypertension [3], patients with
`hypertension with eGFR > 50 ml/min/1.73 m2 or greater, should be
`started on anti-hypertensive medications. The medication regimen
`for patients with systolic blood pressures >20 mm Hg above goal
`should include an angiotensin converting enzyme inhibitor (ACEI)
`or an angiotensin receptor blocker (ARB) plus a thiazide diuretic or
`a calcium channel blocker. Patients with a systolic blood pressures
`<20 mm Hg above goal can be started just on an ACE inhibitor or
`ARB.
`
`3.4. Management of complications
`
`Management is focused on correcting or stabilizing the follow-
`ing complications.
`Treatment for hyperkalemia includes potassium restriction
`<60 mmol/d, and kayexalate, a potassium binder that lowers serum
`potassium levels via GI loss. Additional interventions include the
`re-evaluation and/or adjustment of medications with the potential
`to induce or exacerbate hyperkalemia (i.e. ACEi, potassium sparing
`diuretics, beta-blockers) [108].
`Volume overload can best be managed with fluid restriction
`and diuretic therapy [108,109].
`Although still somewhat controversial, many studies have
`shown the potential benefit of aggressive treatment of metabolic
`acidosis. Sodium bicarbonate supplementation resulted in a sig-
`nificant decrease in progression to dialysis at the end of 2 years
`[96,97]. Therefore, sodium bicarbonate may be a safe and effective
`treatment for metabolic acidosis, with the possible added benefit
`of reducing the rate of decline in renal function.
`Controlling hyperphosphatemia is important because of the
`increased risk development of secondary hyperparathyroidism,
`renal osteodystrophy and cardiovascular mortality, mostly seen in
`the hemodialysis population [109,110]. This can be achieved with
`
`However, evidence of benefit is limited as there are few robust
`intervention studies [90–98].
`The diet recommended is low protein and low carbohydrate
`diet. In the early stages of CKD, protein intake should not exceed
`0.8–1.0 g kg/wt d, while in later stages of CKD protein should be
`limited to 0.8 g kg/wt d. Dietary restriction can help improve renal
`function by decreasing urinary albumin excretion, presumably
`reducing the decline in eGFR [90,91]. In addition, a diet consist-
`ing of no more than 130 g of carbohydrates per day ha

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket