APPLICATION OF NITROGEN-CONTAINING SATURATED HETEROCYCLIC COMPOUND

Information

  • Patent Application
  • 20230330093
  • Publication Number
    20230330093
  • Date Filed
    September 03, 2021
    3 years ago
  • Date Published
    October 19, 2023
    11 months ago
Abstract
The present invention provides a use of a nitrogen-containing saturated heterocyclic compound. Specifically, the present invention provides a use of a nitrogen-containing saturated heterocyclic compound represented by the following formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicine for the treatment and/or prevention of chronic kidney disease:
Description
TECHNICAL FIELD

The present invention relates to a use of a nitrogen-containing saturated heterocyclic compound or a pharmaceutically acceptable salt thereof in the preparation of a medicine for the treatment and/or prevention of chronic kidney disease.


BACKGROUND OF THE ART

Chronic kidney disease (CKD) has the characteristics of high prevalence, low awareness, poor prognosis and high medical costs, which is another disease that seriously endangers human health after cardiovascular and cerebrovascular diseases, diabetes and malignant tumors. In recent years, the prevalence of CKD has increased year by year, and the global prevalence rate of the general population has reached 14.3%, the cross-sectional epidemiological study in China shows that the prevalence of CKD in people over 18 years old is 10.8%. Chronic kidney disease (CKD) is a progressive disease. If the disease is not treated in a timely and effective manner, the condition worsens and progresses, as the course of the disease progresses, patients with chronic kidney disease will develop renal failure. In such a case, without artificial dialysis or kidney transplantation, survival will become difficult. There is currently no particularly effective method for the treatment of chronic kidney disease, therefore, it has great clinical value to actively find medicines that are effective in treating chronic kidney disease and have low toxic and side effects.


CN103562191B discloses a nitrogen-containing saturated heterocyclic compound represented by the following structural formula or a pharmaceutically acceptable salt thereof, which shows an inhibitory effect on renin and can be used to treat hypertension.




embedded image


In the formula, R1 represents a cycloalkane group, etc., R22 represents a substituted aryl etc., R represents lower alkane group, etc., T represents a carbonyl group, Z represents —O— etc., R3, R4, R5 and R6 are the same or different, and represent a hydrogen atom etc.


CN106928218A discloses a salt of a novel pharmaceutically acceptable morpholine derivative (nitrogen-containing saturated heterocyclic ring), including its malate, tartrate, hydrochloride, acetate and naphthalene diphosphate, wherein, tartrate has three crystalline salt forms: crystal form A, crystal form B and dehydrate, malate, hydrochloride and acetate each have one crystalline salt form, and naphthalene diphosphate is amorphous. Compared with the known free bases of morpholine derivatives, the salts of morpholine derivatives have one or more improved properties, such as better crystalline state, which greatly improve water solubility, light stability and thermal stability, etc. The above salt of morpholine derivative or its crystal form can be used to treat and/or prevent hypertension.


SUMMARY OF THE INVENTION

The technical problem to be solved by the present invention is to provide a use of a nitrogen-containing saturated heterocyclic compound or a pharmaceutically acceptable salt thereof in the preparation of a medicine for the treatment and/or prevention of chronic kidney disease in view of the absence of a particularly effective medicine for treating chronic kidney disease in the prior art.


The entire contents of the patent CN103562191B and the patent application CN106928218A described in the background art are hereby incorporated into this description by reference.


In one embodiment of the present invention, provided is a use of a nitrogen-containing saturated heterocyclic compound represented by the following formula I or a pharmaceutically acceptable salt thereof in the preparation of a medicine for the treatment and/or prevention of chronic kidney disease:




embedded image


In a further embodiment of the present invention, the pharmaceutically acceptable salt is hydrochloride, sulfate, phosphate, hydrobromide, acetate, fumarate, oxalate, citrate, methanesulfonate, benzenesulfonate, p-toluenesulfonate, maleate, malate, tartrate, acetate or naphthalene disulfonate.


In a further embodiment of the present invention, the pharmaceutically acceptable salt is malate, tartrate, hydrochloride, acetate or naphthalene disulfonate.


In one embodiment of the present invention, the pharmaceutically acceptable salt is the malate of the compound of formula I, which is a compound formed by the compound of formula I and malic acid at a molar ratio of 1:1, and the structural formula is as follows:




embedded image


In one embodiment of the present invention, the malate is a crystal form, the X-ray powder diffraction pattern thereof has characteristic peaks at 2θ of 7.767°±0.2°, 13.897°±0.2°, 14.775°±0.2°, 17.098°±0.2°, 18.999°±0.2°, 20.153±0.2°, 20.960°±0.2°, 21.423°±0.2°, 26.348°±0.2° and 27.892°±0.2°. In particular, the X-ray powder diffraction pattern of the malate crystal further has characteristic peaks at 2θ of 5.598°±0.2°, 7.357°±0.2°, 10.395°±0.2°, 11.108°±0.2°, 16.037°±0.2°, 16.523°±0.2°, 19.410°±0.2°, 22.645°±0.2°, 26.630°±0.2°, 26.891°±0.2°, 27.380°±0.2°, 31.056°±0.2°, 33.306°±0.2°, 33.775°±0.2° and 39.231°±0.2°. More specifically, the malate crystal of the compound of formula I has an X-ray powder diffraction pattern as shown in FIG. 1.


In one embodiment of the present invention, the chronic kidney disease includes hypertension with nephropathy, hypertension with nephropathy with abnormal glucose metabolism, chronic renal insufficiency with chronic heart failure or chronic kidney disease with abnormal glucose metabolism.


In one embodiment of the present invention, the chronic kidney disease refers to the G1, G2, G3a, G3b, G4 stages of chronic kidney disease, preferably the G2, G3a or G3b stage.


In one embodiment of the present invention, the hypertension in hypertension with nephropathy is grade 1 hypertension, grade 2 hypertension or grade 3 hypertension, preferably grade 1 hypertension or grade 2 hypertension.


In one embodiment of the present invention, the dosage forms of the medicine include tablet, capsule, intravenous injection, inhalant, aerosol, lyophilized agent, patch, gel, spray, or suppository, preferably tablet.


In one embodiment of the present invention, the medicine is unit dose.


In one embodiment of the present invention, the unit dose of the medicine contains the nitrogen-containing saturated heterocyclic compound represented by formula I or the pharmaceutically acceptable salt thereof in a range of 25 mg to 200 mg, for example, 25 mg, 50 mg, 100 mg, 150 mg, 200 mg of the nitrogen-containing saturated heterocyclic compound represented by formula I or the pharmaceutically acceptable salt thereof.


The “Kidney Disease Outcomes Quality Initiative” (KDOQI) working group under the American Kidney Foundation (NKF) formulated the definition and staging criteria for CKD in 2002.


Chronic kidney disease is defined as abnormal kidney structure or function>3 months. The diagnostic criteria for chronic kidney disease is that any of the indicators in Table 1 below is present for ≥3 months.









TABLE 1





Diagnostic criteria for chronic kidney disease


Kidney damage signs
















(1) Albuminuria [AER ≥ 30 mg/24 h;



ACR ≥ 30 mg/g(or ≥3 mg/mmol)];



(2) Abnormal urine sediment;



(3) Renal tubule-associated pathological



changes;



(4) Histological abnormality;



(5) Structural abnormalities detected by



imaging;



(6) History of kidney transplantation



Decreased GFR
eGFR < 60 mL/(min · 1.73 m2)





Note:


meeting at least one item:


AER: Urinary albumin excretion rate;


ACR: Urinary albumin to creatinine ratio;


GFR: glomerular filtration rate







Chronic kidney disease staging: Chronic kidney disease is divided into 5 stages according to the glomerular filtration rate (GFR), see Table 2 below.









TABLE 2







Glomerular Filtration Rate (GFR)


categories - Description and range












GFR




Staging
[mL/(min · 1.73 m2)]
Description














G1
≥90
Normal or high



G2
60-89
Mildly decreased



G3a
45-59
Mildly to moderately





decreased



G3b
30-44
Moderately to severely





decreased



G4
15-29
Severely decreased



G5
<15
Kidney failure









Definition of hypertension: systolic blood pressure (SBP)≥140 mmHg (1 mmHg=0.133 kPa) and/or diastolic blood pressure (DBP)≥90 mmHg, in the case of not using antihypertensive drugs and that office blood pressure is measured 3 times on different days.


That SBP≥140 mmHg and DBP<90 mmHg is isolated systolic hypertension. When the patient has a history of hypertension and is currently using antihypertensive drugs, although the blood pressure is lower than 140/90 mmHg, it should still be diagnosed as hypertension.


According to the elevated blood pressure level, hypertension is further divided into Grade 1, 2 or 3. The classification and definition of blood pressure levels are shown in Table 3 below.









TABLE 3







Classification and definition of blood pressure levels











Systolic

Diastolic



blood

blood



pressure

pressure


Classification
(mmHg)

(mmHg)













Normal
<120
and
<80


Elevated
120-139
and/or
80-90


Hypertension
≥140
and/or
≥90


Grade 1 hypertension (mild)
140-159
and/or
90-99


Grade 2 hypertension (moderate)
160-179
and/or
100-109


Grade 3 hypertension (severe)
≥180
and/or
≥110


Isolated systolic hypertension
≥140
and
<90









Heart failure is a clinical syndrome, defined as a group of complex clinical syndromes in which the ability of the ventricle to fill with or eject blood is impaired due to any abnormality in the structure or function of the heart. The main clinical manifestations are dyspnea and fatigue (restricted activity tolerance), and fluid retention (pulmonary congestion and peripheral edema).


According to the left ventricular ejection fraction (LVEF), heart failure is divided into heart failure with reduced ejection fraction (HFrEF), heart failure with preserved ejection fraction, HFpEF) and heart failure with mid-range ejection fraction (HFmrEF), the definitions of the 3 types of heart failure are shown in Table 4 below:









TABLE 4







Classification and definition of heart failure












Left





ventricular




Symptoms
ejection




and/or
fraction



Classification
signs
(%)
Others













HFrEF
Presented
<40



HFmrEF
Presented
40-49
1. Natriuretic peptide increased a;





2. Meeting at least one of the following:





(1) Left ventricular hypertrophy





and/or left atrium enlargement;





(2) Abnormal diastolic function b


HFpEF
Presented
≥50
1. Natriuretic peptide increased a;





2. Meeting at least one of the following:





(1) Left ventricular hypertrophy





and/or left atrium enlargement;





(2) Abnormal diastolic function b





Notes:


HFrEF, heart failure with reduced ejection fraction;


HFmrEF, heart failure with median ejection fraction;


HFpEF, heart failure with preserved ejection fraction;



a B-type natriuretic peptide (BNP)>35 ng/L and/or N-terminal B-type pronatriuretic peptide (NT-proBNP)>125 ng/L;




b E/e′ ≥ 13, e′ average value (ventricular septum and free wall) <9 cm/s;



— none






Abnormal glucose metabolism includes pre-diabetes and diabetes.


Pre-diabetes is impaired glucose regulation, which means that the blood sugar level has exceeded the normal range, but has not yet reached the diagnostic criteria for diabetes, namely, the intermediate state between normal people and diabetic patients.


Diabetes is a group of common metabolic diseases characterized by hyperglycemia, honey-urine, impaired glucose tolerance and abnormal insulin release in test.


The positive and progressive effects of the present invention are: studies on the effectiveness and safety of nitrogen-containing saturated heterocyclic compound salt administered continuously for 8 weeks to rhesus monkeys (middle-aged/elderly) with chronic renal insufficiency with chronic heart failure have shown that, the nitrogen-containing saturated heterocyclic compound or the pharmaceutically acceptable salt thereof provided by the present invention can effectively prevent or treat chronic kidney disease, the chronic kidney disease includes hypertension with nephropathy, hypertension with nephropathy with abnormal glucose metabolism, chronic renal insufficiency with chronic heart failure or chronic kidney disease with abnormal glucose metabolism. At the same time, the nitrogen-containing saturated heterocyclic compound or the pharmaceutically acceptable salt thereof provided by the present invention is very safe, and there is no adverse event related to administration.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows the XRPD pattern of the malate of the compound of formula I used in the examples of present invention;



FIG. 2 shows drug concentration-time graph of the compound of formula I in monkeys after intravenous (iv) and oral (po) administration in Example 3, in which the doses were respectively 1 mg/kg (intravenous), and 1 mg/kg, 3 mg/kg, and 9 mg/kg (oral).





DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS

The present invention will be further explained by way of examples below, but the present invention is not limited to the scope of the described examples. In the following examples, the experimental methods without specific conditions are selected according to conventional methods and conditions, or according to the product specification.


The malate of the compound of Formula I used in the following examples was prepared and confirmed according to the preparation method described in Example 1 in Patent Application Publication CN106928218A. Valsartan was purchased through commercial channels. The rhesus monkeys used were purchased from Sichuan Premey Xingzhi Biotechnology Co., Ltd. (Premey Primate Research Center).












Abbreviations








Abbreviations
English full name





CKD-EPI
Chronic Kidney Epidemiology


GFR
glomerular filtration rate


eGFRcreat-cys
Estimated glomerular filtration rate(creatinine-cystatin C)


UACR
urine albumin creatine ratio


SBP
Systolic blood pressure


DBP
Diastolic blood pressure


MALB
Micro albumin


CR-U
Urine creatinine


E
Peak Velocity of Mitral Blood Flow at early diastole


Ea
Peak Velocity of Mitral Annulus at early diastole


LVEF
Left Ventricular Ejection Fraction


CVD
cerebrovascular disease


DKD
diabetic kidney disease









Example 1: Study on the Effectiveness and Safety of the Malate of the Compound of Formula I Administered Continuously for 8 Weeks to Rhesus Monkeys with Chronic Renal Insufficiency with Chronic Heart Failure (Middle-Aged/Elderly)

1. Research Purpose


The improvement of renal function by 8-week continuous administration of the malate of the compound of formula I, and the effective dose in which the renal function can be improved or delayed, the intensity of action and the risk of CVD events were evaluated, in order to provide basis for the dosage design of clinical trials, patient selection criteria and safety. Primary efficacy endpoints (ENDPOINT): 1. Improvement of renal function: glomerular filtration rate eGFRcreat-cys, Cystatin C (CysC), creatinine and UACR; 2. Improvement of cardiac function: analysis of changes of cardiac structure and systolic and diastolic functions before and after administration by echocardiographic indicators; 3. Safety indicators: blood pressure, blood potassium, glucose and lipid metabolism related indicators, etc.


2. Test System


2.1 Rhesus Monkeys with Spontaneous Chronic Renal Insufficiency


Animal species: Macaca mulatta (Rhesus Macaque)


Level: Normal level. The pre-test quarantine was qualified, including physical examination, 2 tubercle bacillus tests, parasite, salmonella, shigella and B virus inspections.


Animal logo: A stainless steel number plate engraved with Arabic numerals was worn on the neck ring, and the chest was tattooed.


Supplier: Sichuan Primed Shines Bio-tech Co., Ltd.Production license number: SCXK (Sichuan) 2019-027.


Animal certificate number: 0016710, 0016944, 0016966.


2.2 Selection Criteria


23 male/female animals, aged 14-24 years (equivalent to adults aged 40-70 years) were selected. The body weight thereof was 8.45-14.93 kg for males, and was 6.43 kg for one female.


They suffered from long-term abnormal glucose and lipid metabolism for more than 3 years. The abnormal glucose metabolism was fasting blood glucose (FPG)>4.8 mmol/1, vs. age-matched control group 4.1±0.3 mmol/1.


CKD-EPI rating G3a-G3b, glomerular filtration rate eGFR: 30˜59 ml/min/1.73 m2; or moderate to severe increase in proteinuria (A2-A3): urine albumin/creatinine ratio (UACR)>15 mg/g-350 mg/g (4-6 hour urine collection), vs. age-matched control group 3±2 mg/g.


Normal blood pressure, grade 1 hypertension or grade 2 hypertension (consistent with clinical patient standard).


HFrEF: LVEF 30%-49% (normal value: 50%-70% Simpson biplane method) or HFpEF (moderate or above damage): Ea<8 or E/Ea>10 (based on clinical diagnostic criteria).


2.3 Exclusion Criteria

    • 1) Chronic liver disease (including known active hepatitis) and/or screening for alanine aminotransferase (ALT) or aspartate aminotransferase (AST)>3× upper limit of normal (ULN).
    • 2) Exclusion criteria related to dyslipidemia: TC>7 mmol/1; TG>3.5 mmol/l.
    • 3) Any history of other diseases that may affect the evaluation of drug efficacy.


Table 5 below shows the underlying causes and characteristics of rhesus monkeys with chronic DKD renal insufficiency in baseline period—each dose group.









TABLE 5







The underlying causes and characteristics of rhesus monkeys with chronic DKD renal insufficiency in baseline period-each dose group

































Years



















of














Kidney




abnormal














function




glucose














diagnosis




and

Blood











Body
CKD-

eGFR

CYS C
lipid
FPG
pressure

RBC

K+





Animal
Age
weight
EPI
UACR
(ml/min/
CR-P
mg/
meta-
mmol/
(SBP/DBP)
Anemia
1012/
HGB
mmol/
EF
E/Ea


Group
No.
yrs
kg
rating
mg/g
1.73 m2)
mg/dL
L
bolism
L
mmHg
diagnosis
L
g/L
L
%
/



























Placebo
6653
19
10.92
G3a-
27.8
47
1.23
1.80
≥3
4.90
103/53
Normal
5.23
121
4.70
68.36
11.43


group



A2















(n = 4)
231
18
10.19
G3a-
14.5
58
1.19
1.42
≥3
5.79
134/63
Normal
5.57
135
5.37
54.05
10.87






A2
















1029
21
12.71
G3a
10.5
53
1.00
1.71
≥3
5.35
162/72
Normal
5.69
143
3.92
50.79
10.63



4713
21
11.88
G3a
8.5
47
1.42
1.55
≥3
4.86
143/65
Normal
6.09
140
4.38
58.81
26.16



Mean ± SD
20 ± 2
11.43 ±
/
15.3 ±
51 ± 5
1.21 ±
1.62 ±
/
5.23 ± 0.44
135 ± 24/
/
5.65 ± 0.35
135 ± 10
4.59 ± 0.61
58.00 ±
14.77 ±





1.10

8.7

0.17
0.17


 63 ± 8  




7.65
7.60


Valsartan
2091
18
9.35
G3a-
37.3
55
1.28
1.44
≥3
7.77
131/67
Normal
6.79
153
4.63
64.02
8.40


group



A2















(n = 3)
4861
17
11.23
G3a-
63.6
50
1.31
1.62
≥3
5.75
125/60
Normal
6.77
152
4.27
58.75
13.80






A2
















6645
23
9.97
G3b-
15.0
40
1.50
1.76
≥3
6.06
148/82
Normal
6.09
137
5.07
59.26
18.35






A2
















Mean ± SD
19 ± 3
10.18 ±
/
38.7 ±
48 ± 7
1.36 ±
1.61 ±
/
6.53 ± 1.09
135 ± 12/
/
6.55 ± 0.40
147
4.66 ± 0.40
60.68 ±
13.52 ±





0.96

24.3

0.12
0.16


 70 ± 11 




2.91
4.98


The
144
18
6.43
G3b-
118.5
55
0.90
1.49
≥3
5.46
132/63
Normal
6.41
162
5.43
63.71
7.09


group of



A2















malate
5073
14
9.75
G3a
10.5
48
1.53
1.63
≥3
5.32
 159/105
Normal
5.92
143
4.74
42.11
11.46


of
6651
24
8.45
G3a-
25.4
51
1.10
1.61
≥3
4.23
127/74
Normal
5.64
139
4.03
71.95
9.21


compound



A2















of Formula
257
21
9.45
G3b-
279.3
41
0.99
2.52
≥3
4.59
180/97
Normal
5.50
121
5.20
/
/


I



A2















5 mg/kg
287
15
12.87
G3a
2.4
53
1.43
1.47
≥3
6.08
154/70
Normal
5.74
134
4.75
49.98
5.91


(n = 6)
4721
17
10.38
G3b-
125.8
43
1.79
1.55
≥3
5.31
149/71
Normal
5.45
127
4.72
59.46
14.04






A2
















Mean ± SD
18 ± 4
9.56 ±
/
93.7 ±
49 ± 5
1.29 ±
1.71 ±
/
5.17 ± 0.66
150 ± 19/
/
5.78 ± 0.35
138 ± 14
4.81 ± 0.48
57.44 ±
9.54 ±





2.13

105.9

0.35
0.40


 80 ± 17 




11.6
3.2





Note:


For No. 257 animal which is with thoracic deformity, cardiac ultrasound images cannot be collected.













TABLE 5





Baseline period-the underlying causes and characteristics of rhesus monkeys with chronic DKD renal insufficiency in each dose group


(continued).

































Years











of











ab-






Kidney




normal






function




glucose






diagnosis




and





Body
CKD-

eGFR

CYS C
lipid



Animal
Age
weight
EPI
UACR
(ml/min/
CR-P
mg/
meta-


Group
No.
yrs
kg
rating
mg/g
1.73 m2)
mg/dL
L
bolism





The group
4829
20
12.13
G3a
3.5
54
1.15
1.57
≥3


of malate of
6501
17
10.55
G3a
2.5
50
1.14
1.87
≥3


compound
287
15
14.53
G3a-
15.3
48
1.36
1.72
≥3


of Formula I



A2







2 mg/kg
4721
18
10.79
G3a-
122.1
46
1.42
1.69
≥3


(n = 5)



A2








2091
19
9.88
G3a-
29.3
58
1.06
1.56
≥3






A2








Mean ± SD
18 ± 2
11.58 ± 1.84
/
34.5 ± 50.1
51 ± 5
1.23 ± 0.16
1.68 ± 0.13
/


The group
1567
16
11.47
G3a-
32.8
51
1.25
1.67
≥3


of malate of



A2







compound
4861
18
9.77
G3a-
30.0
54
1.03
1.80
≥3


of Formula I



A2







1 mg/kg
5453
19
14.93
G3a
2.9
53
1.09
1.72
≥3


(n = 5)
6653
20
10.46
G3a
1.6
50
1.05
1.84
≥3



5073
15
10.34
G3a
4.1
52
1.21
1.75
≥3



Mean ± SD
18 ± 2
11.39 ± 2.07
/
14.3 ± 15.7
52 ± 1
1.13 ± 0.10
1.76 ± 0.07
/








Blood










FPG
pressure

RBC

K+





Animal
mmol/
(SBP/DBP)
Anemia
1012/
HGB
mmol/
EF
E/Ea


Group
No.
L
mmHg
diagnosis
L
g/L
L
%
/





The group
4829
4.09
126/58
Normal
5.11
124
4.69
54.41
17.11


of malate of
6501
4.61
111/56
Normal
5.30
129
5.80
70.41
7.73


compound
287
6.87
184/78
Normal
5.62
133
4.77
57.45
6.69


of Formula I











2 mg/kg
4721
5.53
143/83
Normal
4.98
119
4.07
57.90
17.43


(n = 5)












2091
7.7
137/74
Normal
6.13
145
5.40
62.76
8.89






Mean ± SD
5.76 ± 1.51
140 ± 27/
Normal
5.43 ± 0.46
130 ± 10
4.95 ± 0.67
60.59 ± 6.25
11.57 ± 5.26





 70 ± 12 








The group
1567
5.03
101/54
Normal
5.69
127
4.50
65.25
10.50


of malate of











compound
4861
6.09
105/56
Normal
5.69
127
4.51
55.89
9.53


of Formula I











1 mg/kg
5453
5.75
125/60
Normal
5.85
140
4.40
54.86
9.22


(n = 5)
6653
6.29
121/59
Normal
5.49
124
4.78
69.87
8.68



5073
4.59
129/70
Normal
5.64
136
4.79
47.92
10.36



Mean ± SD
5.55 ± 0.72
116 ± 13 
Normal
5.67 ± 0.13
131 ± 7 
4.60 ± 0.18
58.76 ± 8.75
 9.66 ± 0.77





60 ± 6 









3. Experimental Design


3.1 Grouping and Dosage Design


In the trial, animals were brought in three stages. In the first stage, there were two groups: 3 animals (No. 257, No. 4721, No. 287) in a group with 5 mg/kg of the malate of compound of formula I, and 2 animals (No. 1029, No. 4713) in the placebo group. In the second stage, there were three groups: 3 animals (No. 144, No. 5073, No. 6651) in a group with 5 mg/kg of the malate of compound of formula I, 3 animals (No. 2091, No. 4861, No. 6645) in the Valsartan group, and 2 animals (No. 6653, No. 231) in the placebo group. In the third stage, there were two groups: 5 animals (No. 4829, No. 6501, No. 287, No. 4721, No. 2091) in a group with 2 mg/kg of the malate of compound of formula I, and 5 animals (No. 1567, No. 4861, No. 5453, No. 6653, No. 5073) in a group with 1 mg/kg of the malate of compound of formula I.


Among them, 2 animals (No. 287 and No. 4721) in the first stage were subjected to drug withdrawal and elution for about 9 months, and 4 animals (No. 5073, No. 2091, No. 4861 and No. 6653) in the second stage were subjected to drug withdrawal and elution for about 5 months, and then they entered the baseline period of the third stage (the baseline period was 1 month).


See Table 6 below for details.









TABLE 6







Group design information











Number of
Dose
Dosing


Group name
animals
(mg/kg/time)
frequency





Placebo group
4
/
/


Valsartan group
3
2.67
once or twice a day


The group with
6
5
Once a day


5 mg/kg of the





malate of compound





of Formula I





The group with
5
2
Once a day


2 mg/kg of the





malate of compound





of Formula I





The group with
5
1
Once a day


1 mg/kg of the





malate of compound





of Formula I





Remarks: In the Valsartan group, the drug was administered once a day during the administration period in the first and second weeks, and twice a day from the third week to the eighth week.






3.2 Dosing Information


Route of administration: Oral administration.


Dosing frequency: the malate of compound of formula I was administered once a day; in the Valsartan group, the drug was administered once a day during the administration period in the first and second weeks, and twice a day from the third week to the eighth week.


Dosage calculation: the dose for the next week was calculated based on the body weight weighed each time.


Administration time: 08:00-09:00 administration.


3.3 Main Efficacy Indicators


Glomerular filtration rate eGFRcreat-cys: creatinine (Cr—P), urea nitrogen (BUN) and cystatin (CysC) were detected, and the eGFR value was obtained by calculation. It was performed once before the administration and once every two weeks after the administration.





eGFRmale=135×min(Cr/0.9,1)−0.207×max(Cr/0.9,1)−0.601×min(CysC/0.8,1)−0.375×max(CysC/0.8,1)−0.711×0.995Age×3





eGFRfemale=135×min(Cr/0.7,1)−0.284×max(Cr/0.7,1)−0.601×min(CysC/0.8,1)−0.375×max(CysC/0.8,1)−0.711×0.995Age×3×0.969

    • UACR: 4 h/6 h urine was collected, urine microalbumin (malb) and urine creatinine (Cr—U) were detected, and the UACR value was obtained by calculation. This was performed once before administration and once after administration. UACR=MALB/Cr—U.
    • Blood pressure: the blood pressure was checked after anesthesia, including systolic blood pressure (SBP), diastolic blood pressure (DBP), mean blood pressure (MBP) and heart rate (HR). This was performed once before administration and once at weeks 4 and 8 respectively after administration.
    • Cardiac GE Doppler ultrasound: historical cardiac ultrasonography once within 1 year (selected animals), once before administration (within 30 days (D-30) of the baseline period), and once at week 8 after administration.


3.4 Secondary Efficacy Indicators


Serum potassium, twice before administration, once every 2 weeks during the administration period;


Glucose and lipid metabolism and liver function: FPG, FRA, LDL-c, HDL-c, TG, TC, NT-proBNP, ALT, AST, TBIL, etc., twice before administration, once at weeks 4 and 8 respectively after administration;


Other biochemical indicators and hematology testing, once each before and after administration;


Body weight: once before administration, once a week during the administration period;


After administration, the changes in food intake and behavior were observed for 24 hours every day.


3.5 Testing Methods and Equipments


Testing methods: see Table 7 and Table 8 below.


Hematology testing equipment: Siemens ADVIA 2120i Hematology Systems.


Blood biochemical and urine index detection equipment: Roche cobas6000 analyzer series C501 module detection. ELISA kit was used for NT-proBNP test.









TABLE 7







Biochemical testing items









Measurement item
Unit
Measurement method





Serum potassium ion(K+)
mmol/L
Ion selective electrode




method, indirect method


N-terminal pro-B-type natriuretic
pg/mL
Enzyme-linked


peptide (NT-proBNP)

immunosorbent assay


Urine-Micro albumin (Malb)
g/L
Immunoturbidimetry,




2-point endpoint


Urine-creatinine (Cr-U)
mg/dL
Enzyme colorimetry


Blood urea nitrogen (BUN)
mg/dL
Colorimetric method,




rate method


Plasma creatinine (Cr-P)
mg/dL
Enzyme colorimetry


Cystatin C (CysC)
mg/L
Immunoturbidimetry


Total cholesterol (TC)
mmol/L
Enzyme colorimetry


Triglycerides (TG)
mmol/L
Enzyme colorimetry


High density lipoprotein (HDL-c)
mmol/L
Homogeneous enzyme




Colorimetry


Low density lipoprotein (LDL-c)
mmol/L
Homogeneous enzyme




Colorimetry


Blood glucose (FPG)
mmol/L
Hexokinase method


Fructosamine (FRA)
μmol/L
Rate method


Alanine aminotransferase (ALT)
IU/L
IFCC rate method


Aspartate aminotransferase (AST)
IU/L
Colorimetry


Total bilirubin (TBIL)
μmol/L
Diazo method


Alkaline phosphatase (ALP)
IU/L
Colorimetric method,




rate method


Total protein (TP)
g/L
Colorimetry, 2-point




end point


Albumin (ALB)
g/L
colorimetry


Direct bilirubin (DBIL)
μmol/L
Diazo method, 2-point




end point


γ-glutamyl transpeptidase (GGT)
IU/L
Enzyme colorimetry
















TABLE 8







Hematology test items









Index
Unit
Measurement method





White blood cell count (WBC)
10{circumflex over ( )}9/L
2D laser


Red blood cell count (RBC)
10{circumflex over ( )}12/L
2D laser


Hemoglobin (HGB)
g/L
Cyanmethemoglobin


Hematocrit (HCT)
%
2D laser


Mean red blood cell volume (MCV)
fL
2D laser


Mean red blood cell hemoglobin (MCH)
pg
2D laser


Mean red blood cell hemoglobin
g/L
2D laser


concentration (MCHC)




Red blood cell volume distribution
%
2D laser


width (RDW)




Hemoglobin distribution width (HDW)
g/L
2D laser


Directly measured mean hemoglobin
g/L
2D laser


concentration (CHCM)




Average hemoglobin content (CH)
pg
2D laser


Mean red blood cell CH distribution
pg
2D laser


width (CHDW)




Total platelets (PLT)
10{circumflex over ( )}9/L
2D laser


Plateletcrit (PCT)
%
2D laser


Platelet volume distribution width
%
2D laser


(PDW)




Mean platelet volume (MPV)
fL
2D laser









3.6 Cardiac Doppler Ultrasound


Anesthesia method: 15 mg/kg of ketamine hydrochloride was used for intramuscular injection anesthesia. According to the animal anesthesia state, the supplemental anesthesia will be performed after judgment by veterinarian. Each supplemental anesthesia dose was ½ of the initial dose.


Detection method: The animal was placed in the left decubitus position, and the 6S-RS probe (frequency 2.7-8.0 MHz) was used for image acquisition.


Analysis method: After the image was collected and saved, the supporting analysis workstation EchoPAC Software was used to repeatedly detect various indicators of systolic and diastolic functions in 3 consecutive cardiac cycles.


Testing indicators and ultrasonic technology: see Table 9 below.


Testing equipment: GE Vivid S5 color Doppler ultrasound diagnostic system.


3.7 Blood Pressure Test


Test method: 15 mg/kg of ketamine hydrochloride was used for intramuscular injection to anesthetize the animal. After anesthesia, the animal was placed in a supine position. The left upper arm was shaved clean, and a suitable size cuff was tied according to the standard. The blood oxygen probe was clamped on the animal's finger or toe (except for the left hand), and the red light-sensitive surface was on the side of the finger pad. The blood pressure of the animal was continuously measured for 3 times in the automatic mode with an interval of 1 min. If the difference of DBP, SBP and MBP of blood pressure measured more than three times was not significant (the difference between the highest value and the lowest value was less than 15 mmHg), the measurement was completed.


Testing indicators: testing indicators included systolic blood pressure (SBP), diastolic blood pressure (DBP), mean blood pressure (MBP) and heart rate (HR).


Testing equipment: GE B40i electrophysiological monitor.









TABLE 9







Cardiac ultrasound testing indicators









Testing indicators
Unit
Testing technology





Left atrium anteroposterior
mm
Two-dimensional


diameter LAAD (mm)

ultrasound


Left atrium transverse
mm



diameter LATD (mm)




Longitudinal diameter of
mm



left atrium LAVD (mm)




LA Volume-Ellipsoid (ml)
ml



Blood flow peak velocity
cm/s
Pulse Doppler


of mitral valve at early




diastole E




E peak deceleration time
ms



EDT




Blood flow peak velocity
cm/s



of mitral valve at late




diastole A




E/A
/



A duration A Dur
ms



Peak Velocity of Mitral
cm/s
Tissue Doppler


Annulus at early diastole




Ea




Peak Velocity of Mitral
cm/s



Annulus at late diastole




Aa




Ea/Aa
/



E/Ea
/



Isovolumic diastolic time
ms



IVRT




Isovolumic contraction time
ms



IVCT




Pulmonary vein S wave peak
cm/s
Pulse Doppler


velocity PV S




Pulmonary vein D wave peak
cm/s



velocity PV D




Pulmonary venous reflux Ar
cm/s



wave velocity PV Ar




Pulmonary venous reflux Ar
ms



wave duration Ar Dur




Ar-A Dur
ms



Blood flow propagation velocity
cm/s
Color M-mode


of left ventricle at early

ultrasound


diastole Vp




Four-chamber heart end-diastolic
ml
Two-dimensional


volume LVEDV MOD A4C

biplane


Four-chamber heart end-diastolic
mm
Simpson method


left ventricular long diameter




LVLd A4C




Four-chamber heart end-systolic
ml



volume LVESV MOD A4C




Four-chamber heart end-systolic
mm



left ventricular long diameter




LVLs A4C




Four-chamber heart stroke
ml



volume SV MOD A4C




Four-chamber cardiac ejection
%



fraction LVEF MOD A4C




Two-chamber heart end-diastolic
ml



volume LVEDV MOD A2C




Two-chamber heart end-diastolic
mm



left ventricular long diameter




LVLd A2C




Two-chamber heart end-systolic
ml



volume LVESV MOD A2C




Two-chamber heart end-systolic
mm



left ventricular long diameter




LVLs A2C




Two-chamber heart stroke volume
ml



SV MOD A2C




Two-chamber cardiac ejection
%



fraction LVEF MOD A2C




Biplane end-systolic volume
ml



LVESV MOD BP




Biplane end-diastolic volume
ml



LVEDV MOD BP




Biplane ejection fraction
%



EF Biplane




Biplane output per minute
L/min



CO Biplane




Heart rate HR
BPM



RR interval R-R
ms









3.8 Clinical Observation


Number of observations: Observation was performed once a day.


Observation method: observation in cage.


Observation content: injection site, skin, hair, eyes, ears, nose, oral cavity, chest, abdomen, urogenital, limbs and other parts, as well as breathing, exercise, urinary, defecation and behavior changes.


3.9 Determination of Food Intake


Feeding method: Food was supplied once at 8:009:00 in the morning, 10:00 in the morning, 2:00 in the afternoon, and 4:00 in the afternoon, respectively, to ensure that there was food in the food box and animals ate freely. The remaining feed was removed at 07:40-8:00 in the next day.


Feeding amount: about 250-500 g/animal/day. In the experiment, it was ensured that there was feed in the food box for 24 hours, and the animals can eat freely.


Method for measuring food intake: The feed was estimated by a semi-quantitative method, and the amount of food given, the amount discarded and the remaining amount in food boxes were recorded every day. Food intake=food given−amount discarded−remaining amount in food box.


3.10 Weight Determination


Weighing time: before feeding on the day.


Determination method: The animal was fasted for 14-16 h before weighing, and the animal was awake, after transferred to the transfer cage, it was weighed with a large animal scale.


Measuring instrument: METTLER TOLEDO electronic platform scale.


4.0 Criteria for Termination of the Test

    • a. Serious adverse events occurred.
    • b. A serious infection occurred.


5. Results and Analysis


5.1 Effects on UACR


The effects of administration of each group on proteinuria (UACR) were shown in Table 10 below. In each group, the CKD rhesus monkeys who had moderate to severe increase in proteinuria, manifested by baseline urine albumin/creatinine ratio (UACR) ≥15 mg/g to 350 mg/g (4-6 hours urine collection), were chosen, and the change of albumin/creatinine ratio from baseline to 8-week administration (W8) was analyzed.


Placebo group (n=4): Compared with the baseline, the UACR of W8 measured at the end of the trial period was increased by 30.64±34.50% on average. The UACR of the proteinuria from 4 cases of animals was fluctuated steadily within a certain range, which did not show rapid progress.


Valsartan group (n=3, moderate to severe proteinuria increase was included in the statistics): Compared with the baseline, the UACR from 2 animals (No. 2091, No. 4861) of W8 was decreased by 40%-70%, and both showed benefit. For one animal, the baseline UACR was 15 mg/g, and the W8 UACR after administration was 20.4%, which showed the benefit was not obvious. Due to the small number of animals in the group, there was no statistical difference compared with the placebo group.


The group of the malate of compound of Formula I (5 mg/kg once a day (qd)) (n=6, 4/6 moderate-to-severe proteinuria increase was included in statistics): Compared with the baseline, the W8 UACR after administration was decreased by 46.87±36.93% on average, which was significantly lower than that in the placebo group (p<0.05). Among them, the UACR from 3 animals of W8 on was decreased by 60%-70%, as compared with the baseline, and all showed benefits. One animal showed no benefit.


The group of the malate of compound of formula I (2 mg/kg qd) (n=5, 3/5 moderate to severe proteinuria increase was included in the statistics): Compared with the baseline, the W8 UACR after administration was decreased by 66.00±17.42% on average, which was significantly lower than that in the placebo group (p<0.01). Among them, the UACR from three animals of W8 was decreased by about 50%-80% as compared with the baseline, and they all showed benefits.


The group of the malate of compound of formula I (1 mg/kg qd) (n=5, 2/5 moderate-to-severe proteinuria increase was included in statistics): Compared with the baseline, the UACR from one animal of W8 was decreased by 39%. The benefit was not obvious in one case.


In short, the proteinuria (UACR) can be significantly improved by administering 2-5 mg/kg qd of the malate of compound of formula I for 8 weeks.









TABLE 10





Effect of the malate of compound of formula I administered for 8 weeks on the UACR of rhesus monkey with spontaneous chronic


kidney disease (CKD)




































UACR













change
UACR















Animal
MALB (mg/L)
Cr-U (mg/dL)
Urine-Vol (ml)
UACR (mg/g)
rate
Benefit


















Group
No.
Baseline
8 weeks
Baseline
8 weeks
Baseline
8 weeks
Baseline
8 weeks
(%)
assessment





Placebo
6653
38.3
4.7
137.74
15.66
60
35
27.8
30.0
7.94
No


group










benefit


(n = 4)
231
10.6
16.6
73.27
68.31
115
210
14.5
24.3
67.97
No













benefit



1029
4.4
2.2
42.00
81.64
265
55
10.5
10.0
−4.55
No













benefit



4713
4.2
13.6
49.45
105.90
315
50
8.5
12.8
51.20
No













benefit



Mean ± SD
14.4 ± 16.2
9.3 ± 6.9
75.62 ± 43.51
67.88 ± 38.13
189 ± 121
88 ± 82
15.3 ± 8.7 
19.3 ± 9.5 
30.64 ± 34.50
/


Valsartan
2091
7.1
6.2
19.02
28.20
185
240
37.3
22.0
-41.10
Benefit


group
4861
60.0
39.9
94.34
206.27
20
80
63.6
19.3
-69.59
Benefit


(n = 3)
6645
25.8
19.5
171.64
95.47
20
37
15.0
20.4
35.88
/



Mean ± SD
31.0 ± 26.8
21.9 ± 17.0
95.00 ± 76.31
109.98 ± 89.92 
75 ± 95
119 ± 107
38.7 ± 24.3
20.6 ± 1.3 
−24.93 ± 54.56 
/


The group
144
93.2
72.4
78.63
52.32
30
60
118.5
138.4
16.75
No


with










benefit


5 mg/kg of
5073
4.3
1.2
40.84
21.19
235
400
10.5
5.7
NA
/


the malate
6651
46.5
3.4
182.87
37.25
30
90
25.4
9.1
−64.10
Benefit


of
257
59.3
112.3
21.23
128.11
208
25
279.3
87.7
−68.62
Benefit


compound
287
2.1
0.3
89.17
16.98
165
380
2.4
1.8
NA
/


of
4721
26.0
21.6
20.66
61.60
405
115
125.8
35.1
−72.14
Benefit


Formula I
Mean ± SD
38.6 ± 35.0
35.2 ± 46.7
72.23 ± 61.34
52.91 ± 40.68
179 ± 141
178 ± 167
 93.7 ± 105.9
46.3 ± 55.4

−46.87 ± 36.93#

/


(n = 6)







































UACR

















CHANGE
UACR















Animal
MALB (mg/L)
Cr-U (mg/dL)
Urine-Vol (ml)
UACR (mg/g)
RATE
Benefit






















Group
No.
Baseline
4 weeks
8 weeks
Baseline
4 weeks
8 weeks
Baseline
4 weeks
8 weeks
Baseline
4 weeks
8 weeks
(%)
assessment





The
4829
1.2
1.0
3.0
33.98
32.18
53.10
290
330
77
3.5
3.1
5.6
NA
/


group
6501
0.6
12.5
0.4
24.39
157.85
33.60
315
20
260
2.5
7.9
1.2
NA
/


with
287
5.3
5.3
0.6
34.58
41.80
26.33
53
320
420
15.3
12.7
2.3
−85.13
Benefit


2 mg/kg
4721
145.4
103.7
69.5
119.07
111.58
116.29
55
60
60
122.1
92.9
59.8
−51.06
Benefit


of the
2091
6.8
2.4
3.6
23.24
66.89
32.22
370
85
265
29.3
3.6
11.2
−61.81
Benefit


malate of
Mean ± SD
31.9 ± 63.5
25.0 ± 44.2
15.4 ± 30.3
47.05 ± 40.60
82.06 ± 52.33
52.31 ±
217 ± 151
163 ±
216 ±
34.5 ±
24.0 ±
16.0 ± 24.8
−66.00 ±
/


compound






37.16

150
150
50.1
38.7

17.42##



of

















Formula

















I (n = 5)

















The
1567
9.5
4.6
5.2
28.93
30.74
26.02
393
170
375
32.8
15.0
20.0
−39.14
Benefit


group
4861
26.8
34.1
38.4
89.48
121.39
118.23
55
5
25
30.0
28.1
32.5
8.44
No


with














benefit


1 mg/kg
5453
0.6
1.4
0.8
20.69
19.36
25.52
440
380
470
2.9
7.2
3.1
NA
/


of the
6653
0.9
5.1
7.6
56.97
108.29
290.17
120
310
25
1.6
4.7
2.6
NA
/


malate of
5073
0.9
1.3
1.1
21.90
16.09
17.93
443
470
520
4.1
8.1
6.1
NA
/


compound
Mean ± SD
 7.7 ± 11.3
 9.3 ± 14.0
10.6 ± 15.8
43.59 ± 29.56
59.17 ± 51.32
95.57 ±
290 ± 187
273 ±
283 ±
14.3 ±
12.6 ±
12.9 ± 13.0
−15.35 ±
/


of






116.36

172
241
15.7
9.4

33.65



Formula

















I

















(n = 5)





Note:


1. UACR=MALB/Cr-U


2. Change rate = (time point after administration − baseline value)/baseline value * 100%, negative value represented decrease; #compared with placebo group p < 0.05.


3. NA, UACR normal animals were not included in the statistics.


Note:


1. UACR=MALB/Cr-U


2. Change rate = (time point after administration − baseline value)/baseline value*100%, negative value represented decrease; #compared with placebo group p < 0.05, ##compared with placebo group p < 0.01.


3. NA, UACR normal animals were not included in the statistics.






5.2 Effect on Glomerular Filtration Rate eGFRcreat-cys


The effects on eGFR in each group were shown in Table 11 and Table 12 below. Selected eGFRcreat-cys in each group: 30˜59 ml/min/1.73 m2. The changes in the glomerular filtration rate of W8 and baseline were analyzed.


Placebo group (n=4): Compared with the baseline, the glomerular filtration rate eGFRcreat-cys from 4 cases of animal was fluctuated stably within a certain range and no rapid progress was shown.


Valsartan group (n=3): Compared with the baseline, the glomerular filtration rate eGFRcreat-cys of W8 in the Valsartan group was increased by 6±2 ml/min/1.73 m2 on average, which was a very significant increase compared with the change value of the placebo group (p<0.01). The glomerular deterioration was significantly improved, and the curative effect was time-dependent.


The group of the malate of compound of Formula I (5 mg/kg qd) (n=6): Compared with the baseline, the glomerular filtration rate eGFRcreat-cys of W8 was increased by 6±6 ml/min/1.73 m2 on average, which was a significant increase compared with the change value of the placebo group (p<0.05). The curative effect was time-dependent.


The group of the malate of compound of Formula I (2 mg/kg qd) (n=5): Compared with the baseline, the glomerular filtration rate eGFRcreat-cys of W8 was increased by 3±3 ml/min/1.73 m2 on average, which was a significant increase compared with the change value of the placebo group (p<0.05).


The group of the malate of compound of Formula I (1 mg/kg qd) (n=5): Compared with the baseline, the glomerular filtration rate eGFRcreat-cys of W8 was increased by 3±4 ml/min/1.73 m2 on average. Compared with the change value of the placebo group, no statistical change was shown.


In short, the glomerular filtration rate eGFRcreat-cys can be significantly improved by administering 2-5 mg/kg qd of the malate of compound of formula I for 8 weeks.









TABLE 11







Effect of the malate of compound of formula I on eGFRcreat-cys


for 8-week administration in rhesus monkeys with spontaneous chronic kidney disease





















eGFRcreat-cys









Change











Animal
eGFRcreat-cys (ml/min/1.73 m2)
value














Group
No.
Baseline
2 weeks
4 weeks
6 weeks
8 weeks
(8 weeks)

















Placebo
6653
47
49
43
47
44
−3


group
231
58
62
57
56
58
1


(n = 4)
1029
53
57
55
50
56
2



4713
47
49
43
46
45
−2



Mean ± SD
51 ± 5
54 ± 6
49 ± 8
50 ± 4
51 ± 8
1 ± 3  


Valsartan
2091
55
58
55
55
60
5


group
4861
50
54
57
55
59
8


(n = 3)
6645
40
46
46
47
46
5



Mean ± SD
48 ± 7
53 ± 6
53 ± 6
53 ± 5
55 ± 8
6 ± 2##


The group
144
55
51
52
53
57
3


with
5073
48
57
5
54
53
6


5 mg/kg of
6651
51
62
57
49
55
4


the malate
257
41
37
39
40
43
2


of
287
53
54
65
64
61
8


compound
4721
43
61
55
54
60
17


of Formula
Mean ± SD
49 ± 5
54 ± 9
53 ± 8
53 ± 8
55 ± 7
6 ± 6# 


I









(n = 6)









The group
4829
54
58
61
58
59
5


with
6501
50
47
45
48
50
0


2 mg/kg of
287
48
46
50
48
5
2


the malate
4721
46
43
56
51
52
6


of
2091
58
57
59
62
59
1


compound
Mean ± SD
51 ± 5
50 ± 7
54 ± 7
54 ± 6
54 ± 4
3 ± 3# 


of Formula









I









(n = 5)









The group
1567
51
53
60
58
58
7


with
4861
54
56
64
57
59
6


1 mg/kg of
5453
53
50
55
56
53
0


the malate
6653
50
48
52
53
48
−2


of
5073
52
56
56
57
57
5


compound
Mean ± SD
52 ± 1
52 ± 3
57 ± 5
56 ± 2
55 ± 5
3 ± 4  


of Formula









I









(n = 5)





Note:


Change value = 8 weeks − Baseline. Negative value represented decrease.



#change value compared with placebo group p < 0.05,




##change value compared with placebo group p < 0.01.














TABLE 12







Changes of eGFRcreat-cys in rhesus monkeys with spontaneous


chronic kidney disease after 8-week administration of the malate of compound of formula I











eGFRcreat-cy, Change value (ml/min/1.73 m2)













Group
Animal No.
Baseline
2 weeks
4 weeks
6 weeks
8 weeks
















Placebo
6653
0
2
−4
0
−3


group
231
0
4
−1
−2
1


(n = 4)
1029
0
4
2
−4
2



4713
0
2
−4
−1
−2



Mean ± SD
0 ± 0
3 ± 1
−2 ± 3 
−2 ± 2 
−1 ± 3 


Valsartan
2091
0
4
1
1
5


group
4861
0
4
7
5
8


(n = 3)
6645
0
6
6
7
5



Mean ± SD
0 ± 0
5 ± 1
5 ± 3
4 ± 3
6 ± 2


The group
144
0
4
−3
−1
3


with
5073
0
9
4
6
6


5 mg/kg of
6651
0
11
6
−2
4


the malate
257
0
−4
2
−1
2


of
287
0
1
12
11
8


compound
4721
0
17
11
11
17


of Formula
Mean ± SD
0 ± 0
5 ± 9
5 ± 6
4 ± 6
6 ± 6


I








(n = 6)








The group
4829
0
5
7
4
5


with
6501
0
−3
−5
−1
0


2 mg/kg of
287
0
−3
1
0
2


the malate
4721
0
−3
10
5
6


of
2091
0
0
1
5
1


compound
Mean ± SD
0 ± 0
−1 ± 3 
3 ± 6
3 ± 3
3 ± 3


of Formula








(n = 5)








The group
1567
0
1
8
7
7


with
4861
0
2
10
3
6


1 mg/kg of
5453
0
−3
3
3
0


the malate
6653
0
−2
1
3
2


of
5073
0
4
5
6
5


compound
Mean ± SD
0 ± 0
0 ± 3
5 ± 4
4 ± 2
3 ± 4


of Formula








I








(n = 5)





Note:


Change value = current value − Baseline, negative value represented decrease.






5.3 Effects on CysC, Cr—P and BUN


The effects on CysC, Cr—P and BUN after administration in each group were shown in Tables 13 to 15.









TABLE 13







Effect of the malate of compound of formula I on Cr-P in rhesus monkeys


with spontaneous chronic kidney disease for 8-week administration










Animal
Cr-P (mg/dL)













Group
No.
Baseline
2 weeks
4 weeks
6 weeks
8 weeks





Placebo
6653
1.33
1.23
1.19
1.28
1.16


group
 231
1.20
1.19
1.11
1.18
1.20


(n = 4)
1029
0.98
1.00
0.92
1.00
1.04



4713
1.33
1.42
1.25
1.47
1.33



Mean ± SD
1.21 ± 0.17
1.21 ± 0.17
1.12 ± 0.14
1.23 ± 0.20
1.18 ± 0.12


Valsartan
2091
1.28
1.12
1.12
1.11
1.09


group
4861
1.31
1.12
1.08
1.11
1.03


(n = 3)
6645
1.50
1.22
1.26
1.26
1.26



Mean ± SD
1.36 ± 0.12
 1.15 ± 0.06*
  1.15 ± 0.09**
  1.16 ± 0.09**
  1.13 ± 0.12**


The group
144
0.90
0.92
0.85
0.87
0.85


with 5 mg/kg
5073
1.53
1.14
1.26
1.42
1.22


of the malate
6651
1.10
0.88
0.89
1.02
0.95


of compound
 257
0.99
1.12
1.15
1.06
1.04


of Formula I
 287
1.43
1.40
1.13
1.12
1.11


(n = 6)
4721
1.79
0.96
1.33
1.32
1.18



Mean ± SD
1.29 ± 0.35
1.07 ± 0.19
1.10 ± 0.19
1.14 ± 0.20
 1.06 ± 0.14*


The group
4829
1.15
1.1 
1.15
1.17
1.17


with 2 mg/kg
6501
1.14
1.19
1.44
1.27
1.29


of the malate
 287
1.36
1.52
1.47
1.43
1.4 


of compound
4721
1.42
1.54
1.26
1.34
1.3 


of Formula I
2091
1.06
1.11
1.09
1.04
1.11


(n = 5)
Mean ± SD
1.23 ± 0.16
1.29 ± 0.22
1.28 ± 0.17
1.25 ± 0.15
1.25 ± 0.11


The group
1567
1.25
1.21
1.11
1.16
1.14


with 1 mg/kg
4861
1.03
1.07
1.01
1.07
1.01


of the malate
5453
1.09
1.12
1.12
1.07
1.17


of compound
6653
1.05
1.13
1.06
1.07
1.18


of Formula I
5073
1.21
1.24
1.26
1.22
1.22


(n = 5)
Mean ± SD
1.13 ± 0.10
1.15 ± 0.07
1.11 ± 0.09
1.12 ± 0.07
1.14 ± 0.08





Note:


*p < 0.05 compared with the baseline value,


**p < 0.01 compared with the baseline value.













TABLE 14







Effect of the malate of compound of formula I on CysC in rhesus monkeys


with spontaneous chronic kidney disease for 8-week administration










Animal
CysC (mg/L)













Group
No.
Baseline
2 weeks
4 weeks
6 weeks
8 weeks





Placebo group
6653
2.03
1.80
1.74
1.99
1.88


(n = 4)
 231
1.42
1.42
1.37
1.47
1.49



1029
1.59
1.71
1.66
1.63
1.83



4713
1.44
1.55
1.61
1.69
1.67



Mean ± SD
1.62 ± 0.28
1.62 ± 0.17
1.60 ± 0.16
1.70 ± 0.22
1.72 ± 0.18


Valsartan group
2091
1.66
1.44
1.47
1.58
1.59


(n = 3)
4861
1.68
1.62
1.65
1.58
1.64



6645
1.73
1.76
1.74
1.69
1.63



Mean ± SD
1.69 ± 0.04
1.61 ± 0.16
1.62 ± 0.14
1.62 ± 0.06
1.62 ± 0.03


The group with
 144
1.49
1.63
1.68
1.59
1.46


5 mg/kg of the
5073
1.63
1.63
1.73
1.47
1.69


malate of
6651
1.61
1.45
1.64
1.80
1.66


compound of
 257
2.52
2.63
2.41
2.45
2.29


Formula I
 287
1.47
1.46
1.36
1.40
1.50


(n = 6)
4721
1.55
1.64
1.44
1.46
1.39



Mean ± SD
1.71 ± 0.40
1.74 ± 0.44
1.71 ± 0.37
1.70 ± 0.40
1.67 ± 0.33


The group with
4829
1.57
1.44
1.31
1.39
1.36


2 mg/kg of the
6501
1.87
1.97
1.77
1.76
1.68


malate of
 287
1.72
1.71
1.56
1.65
1.58


compound of
4721
1.69
1.73
1.43
1.54
1.55


Formula I
2091
1.56
1.51
1.47
1.42
1.47


(n = 5)
Mean ± SD
1.68 ± 0.13
1.67 ± 0.21
 1.51 ± 0.17**
 1.55 ± 0.16**
1.53 ± 0.12


The group with
1567
1.67
1.66
1.49
1.48
1.51


1 mg/kg of the
4861
1.80
1.64
1.43
1.61
1.59


malate of
5453
1.72
1.83
1.57
1.62
1.62


compound of
6653
1.84
1.83
1.76
1.68
1.77


Formula I
5073
1.75
1.55
1.50
1.50
1.51


(n = 5)
Mean ± SD
1.76 ± 0.07
  1.70 ± 0.12**
 1.55 ± 0.13**
 1.58 ± 0.08**
 1.60 ± 0.11**





Note:


**p < 0.01 compared with the baseline value.













TABLE 15







Effect of the malate of compound of formula I


on BUN of rhesus monkeys with spontaneous chronic


kidney disease for 8-week administration









BUN (mg/dL)











Group
Animal No.
Baseline2
4 weeks
8 weeks














Placebo
6653
34.8
30.0
26.6


group
231
34.0
29.3
34.5


(n = 4)
1029
27.5
33.3
27.9



4713
36.7
37.4
38.6



Mean ± SD
33.3 ± 4.0
32.5 ± 3.7
31.9 ± 5.6


Valsartan
2091
19.0
22.2
19.5


group
4861
25.4
29.1
33.4


(n = 3)
6645
31.2
33.4
26.0



Mean ± SD
25.2 ± 6.1
28.2 ± 5.7
26.3 ± 7.0


The group
144
18.6
23.6
18.6


with 5 mg/kg
5073
37.3
32.5
30.4


of the malate
6651
41.3
32.6
30.1


of compound
257
77.3
62.5
62.5


of Formula I
287
18.1
27.2
27.9


(n = 6)
4721
81.2
53.8
41.9



Mean ± SD
 45.6 ± 27.7
 38.7 ± 15.7
 35.2 ± 15.3


The group
4829
37.5
42.7
36.4


with 2 mg/kg
6501
24.2
37.1
29.4


of the malate
287
18.7
22.9
23.6


of compound
4721
42.1
41.6
49.5


of Formula I
2091
25.4
30.4
22.6


(n = 5)
Mean ± SD
29.6 ± 9.8
34.9 ± 8.3
 32.3 ± 11.1


The group
1567
35.1
38.0
29.5


with 1 mg/kg
4861
32.4
24.9
30.5


of the malate
5453
19.3
15.4
16.2


of compound
6653
39.8
36.3
38.5


of Formula I
5073
40.8
32.7
28.1


(n = 5)
Mean ± SD
33.5 ± 8.6
29.5 ± 9.3
28.6 ± 8.0









5.4 Effect on Blood Pressure


See Table 16 for the effects of administration of each administration group on blood pressure. The blood pressure test of primates required anesthesia, so it was necessary to control the variability of heart rate at various time points to make statistics.


Placebo group (n=4, 2/4 hypertension): Compared with the baseline, the blood pressure of 4 cases of animals was fluctuated steadily within a certain range.


Valsartan group (n=3, 1/3 hypertension): Compared with the baseline, SBP and DBP were decreased by 14±4 mmHg and 7±0 mmHg on average respectively after W8 administration, which was extremely significantly lower than the change value of the placebo group (p<0.01).


The group of the malate of compound of Formula I (5 mg/kg qd) (n=6, 4/6 cases of hypertension): Compared with the baseline, SBP and DBP were reduced by 21±8 mmHg and 10±7 mmHg on average respectively after W8 administration, which was extremely significantly lower than the change value of the placebo group (p<0.01). The curative effect was time-dependent.


The group of the malate of compound of Formula I (2 mg/kg qd) (n=5, 2/5 hypertension): Compared with the baseline, SBP and DBP were reduced by 19±9 mmHg and 9±8 mmHg on average respectively after W8 administration, which was extremely significantly lower than the change value of the placebo group (P<0.01). The curative effect was time-dependent.


The group of the malate of compound of Formula I (1 mg/kg qd) (n=5): Compared with the baseline, SBP and DBP were reduced by 11±4 mmHg and 5±4 mmHg on average respectively after W8 administration, which was significantly lower than the change value of the placebo group (p<0.05).


In a word, the malate of compound of formula I showed the activity of lowering the blood pressure when administering 1-5 mg/kg qd for 8 weeks.









TABLE 16







Effect of the malate of compound of formula I on blood pressure in rhesus


monkeys with spontaneous chronic kidney disease for 8-week administration





















SBP Change



DBP Change







value



value













Animal
SBP (mmHg)
(mmHg)
DBP (mmHg)
(mmHg)


















Group
No.
Baseline
4 weeks
8 weeks
4 weeks
8 weeks
Baseline
4 weeks
8 weeks
4 weeks
8 weeks





















Placebo
6653
103
110
121
6
18
53
58
65
5
13


group
231
134
122
139
−11
6
63
61
66
−1
3


(n = 4)
1029
162
169
156
7
−6
72
73
87
4
13



4713
143
164
154
21
11
65
72
68
7
2



Mean ± SD
135 ± 24
141 ± 30
143 ± 16 
  6 ± 13
  7 ± 10  
63 ± 8 
67 ± 9 
72 ± 11
 −4 ± 4
  8 ± 7  


Valsartan
2091
131
130
121
−1
−10
67
62
61
−5
−6


group
4861
125
106
107
−19
−18
60
53
53
−7
−7


(n = 3)
6645
148
138
133
−10
−15
82
62
75
−20
−7



Mean ± SD
135 ± 12
125 ± 17
120 ± 13*
−10 ± 9 
−14 ± 4##
70 ± 11
59 ± 5 
  63 ± 11**
−10 ± 8#
 −7 ± 0##


The group
144
132
136
NA
3
NA
63
61
NA
−2
NA


with
5073
159
124
NA
−35
NA
105
70
NA
−35
NA


5 mg/kg of
6651
127
NA
114
NA
−12
74
NA
57
NA
−18


the malate
257
180
159
161
−21
−19
97
8
97
−16
0


of
287
154
144
121
−9
−32
70
68
60
−3
−11


compound
4721
149
139
128
−10
−21
71
68
59
−3
−13


of
Mean ± SD
150 ± 19
138 ± 12
 129 ± 16**
−14 ± 14#
−21 ± 8##
80 ± 17
71 ± 7 
 66 ± 15*
−12 ± 14#
−10 ± 7##


Formula I













(n = 6)













The group
4829
126
110
102
−16
−24
58
55
52
−2
−6


with
6501
111
NA
NA
NA
NA
56
NA
NA
NA
NA


2 mg/kg of
287
184
163
160
−21
−24
78
83
70
−5
−8


the malate
4721
143
NA
121
NA
−22
83
NA
62
NA
2


of
2091
137
138
133
1
−5
74
66
73
−8
−1


compound
Mean ± SD
140 ± 27
140 ± 20
128 ± 21 
−12 ± 11
−19 ± 9##
70 ± 12
71 ± 11
64 ± 8 
 −2 ± 7
 −9 ± 8##


of













Formula I













(n = 5)













The group
1567
101
104
94
4
−7
54
51
53
−3
0


with
4861
105
109
89
4
−15
56
54
49
−2
−8


1 mg/kg of
5453
125
124
114
−1
−12
60
65
53
5
−7


the malate
6653
121
NA
NA
NA
NA
59
NA
NA
NA
NA


of
5073
129
NA
NA
NA
NA
70
NA
NA
NA
NA


compound
Mean ± SD
116 ± 13
118 ± 17
111 ± 20 
  2 ± 3 
−11 ± 4# 
60 ± 6 
61 ± 12
60 ± 12
  0 ± 5
 −5 ± 4# 


of













Formula I













(n = 5)





Note:


*p < 0.05 compared with the baseline value,


**p < 0.01 compared with the baseline value.



#change value compared with placebo group p < 0.05,




##change value compared with placebo group p < 0.01.







5.5 Effect on Heart Function


See Table 17 for the effect on echocardiographic indicators in each group.


There was no obvious adverse reaction in the systolic and diastolic function in each dose group of the malate of the compound of formula I, Valsartan group and placebo group.


In addition, 2 cases of rhesus monkey animals (No. 5073 and No. 287) with low ejection fraction and cardiac insufficiency (SD) in the group with 5 mg/kg of the malate of compound of formula I for 8-week administration had increased LVEF % as compared with the baseline. However, whether this product can treat chronic heart failure needs to be supported by more follow-up research cohort data.









TABLE 17







Effect of the malate of compound of formula I on the main indexes of heart


function in rhesus monkeys with spontaneous chronic kidney disease and evaluation


of the therapeutic effect after 8-week administration












Animal
LVEF (%)
Ea (cm/s)
E/Ea














Group
No.
Baseline
8 weeks
Baseline
8 weeks
Baseline
8 weeks

















Placebo
6653
68.36
64.37
6.03
4.65
11.43
13.30


group
231
54.05
59.26
5.10
4.94
10.87
11.59


(n = 4)
1029
50.79
50.71
4.02
5.16
10.63
10.13



4713
58.81
55.63
4.26
4.60
26.16
22.38



Mean ± SD
58.00 ± 7.65 
57.49 ± 5.77 
4.85 ± 0.91
4.84 ± 0.26
14.77 ± 7.60 
14.35 ± 5.51 


Valsartan
2091
64.02
64.09
4.16
7.01
8.40
6.90


group
4861
58.75
57.98
5.14
7.17
13.80
10.43


(n = 3)
6645
59.26
61.97
3.44
4.61
18.35
16.40



Mean ± SD
60.68 ± 2.91 
61.35 ± 3.10 
4.25 ± 0.85
6.26 ± 1.43
13.52 ± 4.98 
11.24 ± 4.80 


The group
144
63.71
62.99
6.57
8.53
7.09
7.38


with 5 mg/kg
5073
42.11
45.89
4.92
8.26
11.46
9.25


of the malate
6651
71.95
73.88
3.29
6.47
9.21
8.22


of compound
257#
1
1
/
1
1
/


of Formula I
287
49.98
54.16
8.75
8.40
5.91
6.12


(n = 6)
4721
59.46
55.90
5.22
6.62
14.04
11.07



Mean ± SD
57.44 ± 11.67
58.56 ± 10.50
5.75 ± 2.04
7.66 ± 1.02
9.54 ± 3.29
8.41 ± 1.88


The group
4829
54.41
58.54
4.36
5.00
17.11
10.63


with 2 mg/kg
6501
70.41
67.80
6.37
6.66
7.73
6.35


of the malate
287
57.45
57.13
8.94
8.01
6.69
7.84


of compound
4721
57.90
58.03
4.46
5.91
17.43
9.88


of Formula I
2091
62.76
62.85
5.82
6.82
8.89
6.22


(n = 5)
Mean ± SD
60.59 ± 6.25 
60.87 ± 4.46 
5.99 ± 1.86
6.48 ± 1.12
11.57 ± 5.26 
8.18 ± 2.01


The group
1567
65.25
63.95
5.90
5.35
10.50
10.68


with 1 mg/kg
4861
55.89
55.81
6.31
5.65
9.53
11.36


of the malate
5453
54.86
54.03
4.81
6.99
9.22
8.16


of compound
6653
69.87
61.01
5.18
5.37
8.68
8.95


of Formula I
5073
47.92
49.37
6.37
5.06
10.36
10.64


(n = 5)
Mean ± SD
58.76 ± 8.75 
56.83 ± 5.76 
5.71 ± 0.69
5.68 ± 0.76
9.66 ± 0.77
9.96 ± 1.34





Remarks:


The level of abnormal diastolic function was determined after comprehensive consideration of Ea, E/Ea, Ea/Aa and other functional indicators; when the abnormal level of diastolic function of the test animal was improved from the baseline, it was regarded as a “benefit”.


Otherwise, it was regarded as “invalid”. For No. 257, the animal is with thoracic deformity, and cardiac ultrasound images cannot be collected.






5.6 Effect on Blood Biomarker of Heart Function Impairment


5.6.1 Effect on NT-proBNP


The effect of the malate of the compound of formula I on NT-proBNP was shown in Table 18.


Compared with the baseline period, there was no significant change in NT-proBNP levels in each administration group after 8-week administration.









TABLE 18







Effect of the malate of compound of formula I on


NT-proBNP in Rhesus Monkeys with Spontaneous Chronic


Kidney Disease for 8-week administration









NT-proBNP (pg/ml)











Group
Animal No.
Baseline
4 weeks
8 weeks














Placebo
6653
342
143
172


group
231
146
187
130


(n = 4)
1029
155
170
201



4713
169
177
230



Mean ± SD
203 ± 93 
169 ± 19 
183 ± 42


Valsartan
2091
211
179
217


group
4861
500
260
288


(n = 3)
6645
394
285
291



Mean ± SD
368 ± 146
241 ± 56 
266 ± 42


The group
144
312
254
261


with 5 mg/kg
5073
88
108
146


of the malate
6651
211
245
250


of compound
257
432
418
351


of Formula I
287
177
210
197


(n = 6)
4721
162
178
180



Mean ± SD
230 ± 123
236 ± 104
231 ± 73


The group
4829
43
46
54


with 2 mg/kg
6501
155
59
178


of the malate
287
335
304
253


of compound
4721
46
46
54


of Formula I
2091
250
107
174


(n = 5)
Mean ± SD
166 ± 127
113 ± 110
143 ± 87


The group
1567
88
44
70


with 1 mg/kg
4861
299
239
157


of the malate
5453
127
37
136


of compound
6653
203
304
141


of Formula I
5073
44
40
39


(n = 5)
Mean ± SD
152 ± 101
133 ± 129
109 ± 51









5.6.2 Effect on K+


The effect of the malate of compound of formula I on K+ was shown in Table 19.


Compared with the baseline period, there was no significant change in serum K+ levels of animals in each administration group for 8-week administration.









TABLE 19







Effect of the malate of compound of formula I on K+ in rhesus monkeys


with spontaneous chronic kidney disease for 8-week administration











K+ (mmol/L)















Animal
Baseline
Baseline






Group
No.
1
2
2 weeks
4 weeks
6 weeks
8 weeks





Placebo
6653
4.47
4.70
3.98
4.85
4.45
4.15


group
 231
4.79
5.37
5.78
4.76
5.02
5.32


(n = 4)
1029
4.43
3.92
3.68
3.50
4.12
3.68



4713
4.95
4.38
3.64
4.01
3.98
3.96



Mean ± SD
4.66 ± 0.25
4.59 ± 0.61
4.27 ± 1.02
4.28 ± 0.64
4.39 ± 0.46
4.28 ± 0.72


Valsartan
2091
4.94
4.63
5.38
5.68
5.28
5.94


group
4861
3.86
4.27
4.47
4.55
4.81
4.50


(n = 3)
6645
4.31
5.07
5.43
4.88
5.09
4.95



Mean ± SD
4.37 ± 0.54
4.66 ± 0.40
5.09 ± 0.54
5.04 ± 0.58
5.06 ± 0.24
5.13 ± 0.74


The group
 144
4.78
5.43
4.91
4.33
4.53
4.26


with
5073
4.54
4.74
4.55
4.55
4.16
4.64


5mg/kg of
6651
4.93
4.03
6.07
5.46
4.77
4.49


the malate
 257
5.41
5.20
5.37
4.50
4.76
5.28


of
 287
5.16
4.75
5.05
4.61
5.58
5.00


compound
4721
4.52
4.72
3.93
3.51
3.60
3.26


of Formula
Mean ± SD
4.89 ± 0.35
4.81 ± 0.48
4.98 ± 0.73
4.49 ± 0.62
4.57 ± 0.66
4.49 ± 0.70


1









(n = 6)









The group
4829
4.53
4.69
4.02
4.74
5.48
4.50


with
6501
4.42
5.80
4.83
4.76
5.72
4.36


2 mg/kg of
 287
4.92
4.77
4.73
4.55
4.90
4.47


the malate
4721
3.43
4.07
4.43
4.31
4.26
3.93


of
2091
6.19
5.40
5.08
4.76
4.99
4.77


compound
Mean ± SD
4.70 ± 1.00
4.95 ± 0.67
4.62 ± 0.41
4.62 ± 0.20
5.07 ± 0.57
4.41 ± 0.31


of Formula









I









(n = 5)









The group
1567
3.93
4.50
4.17
4.30
4.67
4.29


with
4861
4.30
4.51
4.14
3.94
4.99
4.17


1 mg/kg of
5453
4.15
4.40
4.15
4.26
4.08
4.15


the malate
6653
4.74
4.78
4.95
5.23
5.57
4.49


of
5073
4.98
4.79
5.04
4.79
5.30
4.65


compound
Mean ± SD
4.42 ± 0.43
4.60 ± 0.18
4.49 ± 0.46
4.50 ± 0.51
4.92 ± 0.58
4.35 ± 0.22


of Formula









I









(n = 5)









5.7 Safety Tolerance Study


During the administration period, there were no adverse events related to administration in the groups administered with malate of the compound of formula I, and no significant changes were observed in liver function, renal function, food intake, body weight, hematological indicators, etc.


6 Conclusion


The proteinuria (UACR) and the glomerular filtration rate eGFRcreat-cys can be significantly improved by administering the malate of compound of formula I in an amount of 2-5 mg/kg qd for 8 weeks. The effect thereof was equivalent to the Valsartan group.


After 8-week administration of the malate of compound of formula I in an amount of 1-5 mg/kg qd, the activity of lowering blood pressure was shown. At the same time, there was no risk of hyperkalemia under the test conditions.


Example 2: Evaluation of Preclinical Safety Profile of the Compound of Formula I

1. Materials and Methods


1.1 Test Substance


The compound of Formula I was prepared and confirmed according to the method in CN103562191B. The 0.5% of sodium carboxymethyl cellulose (CMC-Na) was used as a vehicle. The compound of Formula I was stored at room temperature, protected from light and wetness. For cynomolgus monkey safety studies, the test substance was suspended in 0.5% CMC-Na at the relevant concentrations. The homogeneity, concentration and one week stability of prepared suspension was analyzed to ensure proper administration of the reported doses. The six month accelerated stability of active pharmaceutical ingredient (API) was tested by the inventor.


1.2 Animal Husbandry


Healthy male and female cynomolgus monkeys aged 3-4 years were supplied by Hainan New Source Biotech Co. Ltd. (Hainan, China). The body weight range at the start of treatment was 3.2-5.8 kg for male and 2.5-3.7 kg for female. They were housed for a 36-day acclimation period before study. Once a day in the morning, animals eat and drink freely.


This nonclinical laboratory study has been carried out in compliance with the guidelines from the China Food and Drug Administration (CFDA). This experiment was conducted in facilities approved by the Association for Assessment and Accreditation of Laboratory Animal Care International (AAA LAC), and animals were maintained in accordance with the Guide for the Care and Use of Laboratory Animals (Nussberger et al., 2008).


1.3 Doses and Treatment Schedule


Cynomolgus monkeys (5/sex/group) were selected using a computerized randomization procedure based on body weight, and the compound of Formula I was administered to the cynomolgus monkeys by nasogastric feeding at dose levels of 0 (control), 20, 100, 450 mg/kg/day in a volume of 5 ml/kg. Individual dose volumes were adjusted weekly based on body weight of animals. For all the groups, 2/3 of the animals were randomly selected and euthanized on day 28. The remaining animals were euthanized after 28-day drug free period.


1.4 Clinical Observations


Mortality and clinical signs were evaluated daily from the beginning of the quarantine period. Each animal was examined at least twice daily for any change in behavior, reaction to treatment or illness after administration.


Observations included, but were not limited to the following: changes in skin and fur; eyes and mucous membranes; respiratory, circulatory, autonomic, central nervous systems and behavior patterns. Rectal temperature was measured before administration (twice), at 1 and 24 h after first administration, and before d26 administration and at 1 and 24 h after d26 administration. Body weight was measured every week. Food consumption was estimated for monkey every day. Ophthalmoscopy was conducted for the monkey study using a portable slit lamp (YZ2) and a direct ophthalmoscope (GFJY-01B).


1.5 Laboratory Test


For the monkey study, at quarantine period (twice, d01 and d02), day 14, day 28 and day 56, the following parameters were measured as shown in Table 20. (1) Hematological and coagulation index examination were performed using a Bayer ADVIA2120 (Germany) and a Sysmex CA-1500 (Japan). (2) The serum biochemical examination was assessed using a HITACHI 7080 automated analyzer (Japan) and an Easylyte PLUS electrolyte analyzer (MEDICA, America). (3) The 24-h pooled urine of each animal was collected by a tray under each cage. Urinalysis was carried out using Uritest-300 (China). (4) The 24-h pooled faeces of each monkey were collected by a tray under each cage. Fecal occult blood was detected. (5) ACCESS 2 Chemiluminescence immunoassay was used for myocardial troponin (TropI, non-GLP) test.









TABLE 20







Parameters evaluated in hematology, serum chemistry and urinanalysis.










Hematology
Serum chemistry
Urinanalysis
feces





Erythrocyte count (RBC)
Alanine aminotransferase
Glucose
Occult



(ALT)

blood


Hemoglobin concentration
Aspartate aminotransferase
pH



(HB)
(AST)




Hematocrit (HCT)
Alkaline phosphatase
Specific




(ALP)
gravity



Mean corpuscular volume
Glucose (GLU)
Protein



(MCV)





Mean corpuscular
Total protein (TP)
Bilirubin



hemoglobin (MCH)





Mean corpuscular
Albumin (ALB)
Ketone



hemoglobin concentration





(MCHC)





Platelet (PLT)
Urea nitrogen (BUN)
Occult





blood



Total leukocyte
Creatinine (CREA)
Nitrite



count (WBC)





Lymphocytes (LYMPH)
Triglyceride (TG)
Urobilinogen



Monocytes (MONO)
Total cholesterol





(CHOL)




Eosinophils (EOS)
Total bilirubin





(TBIL)




Basophils (BASO)
Gamma-glutamyl





transpeptidase (GGT)




Neutrophils (NEUT)
Calcium (Ca)




Reticulocyte count
Phosphorus (P)




(RETIC)





Prothrombin time (PT)
Creatine





phosphokinase (CPK)




Activated partial
K+, Na+, Cl




thromboplastin time





(APTT)






TropI









1.6 Routine ECG Analysis for the Monkeys Study


Routine ECG analysis was performed at each scheduled time (day 1 after administration, d01 and d02 after administration, 1 and 24 h after administration on day 1, before administration, and 1 and 24 h after administration on day 26 and day 56). P-wave, R-wave, T-wave, P-R intervals, Q-T intervals, QRS duration and heart rate were recorded by an ECG system using derivation DII (ECG-6951E, Shanghai, China).


1.7 Necropsy and Histopathology


A complete gross necropsy was conducted on all animals by visual inspection at the end of the exposure period (day 28) and the recovery phase (day 56). The following selected organs were trimmed, weighed and evaluated in terms of absolute weight and as a percentage of final body weight or brain weight: brain, heart, kidney, liver, spleen, thymus, testes, epididymides, uterus, ovaries, adrenals and thyroid (including the parathyroid). The following tissues were preserved in 10% neutral buffered formalin: brain, pituitary gland, thyroid (including the parathyroid), trachea, heart, pancreas, spleen, adrenal glands, prostate, ovaries, uterus (including cervix uteri and oviduct), vagina, testes, epididymides, seminal vesicle, esophagus, duodenum, jejunum, ileum, cecum, colon, rectum, mesenteric lymph nodes, Peyer's patches, submaxillary lymph nodes, aorta, eyes, skeletal muscle, sciatic nerve, femur (including metaphysis), mammary gland, sternum, salivary glands, spinal cord, urinary bladder, lung (including the bronchi), liver, kidneys, stomach, bone marrow (sternal), thymus, sternum, gallbladder, any gross lesions or masses. The lung tissue was inflated with fixative at the time of necropsy. All the preserved tissues were paraffin embedded, sectioned, stained with hematoxylin and eosin (HE) and examined microscopically. Bone marrow cellular morphology examination was conducted with both smear (sternum) and paraffin-embedded sternum section.


1.8 Toxicokinetics


Blood samples were obtained on day1 and day26 at 0, 0.5, 1.5, 3, 5, 7 and 24 h after administration. On each occasion, approximately 1.0 mL of blood was drawn from the vein and collected into EDTA-K2 anticoagulant tubes. The blood samples were put on ice and then centrifuged (3500 rpm, 5 min) at 4° C. to obtain plasma samples, which were kept frozen at about −80° C. until analysis. Toxicokinetics determination of the compound of Formula I was conducted at the Institute of Materia Medica, Chinese Academy of Sciences (Shanghai, China).


1.9 Statistical Analysis


For each sex, body weight, rectal temperature, ECG, organ weight, hematological parameters and serum biochemical data were analyzed with a statistical software SAS 9.3. Firstly homogeneity of variance was analyzed using Levene's test. If P>0.05, the one-way ANOVA analysis was employed. If P<0.05, the Kruskal-Wallis test was used. If the resulting ANOVA p-value was <0.05, a comparison of each group using the Dunnett T test was performed. If the resulting Kruskal-Wallis test p-value was <0.05, then the Dunnett T test was performed after data rank conversion.


2. Results


2.1 Clinical Observations


Slight declining body weight gain was observed in high dose compared with controls (2.7±0.4 versus 3.2±0.2 for females, 4.1±0.5 versus 4.6±0.5 for males) with no statistical significance.


Treatment related early death occurred in one high dose female on day23, with clinical signs including hypoactivity, hunch back, low body temperature, diarrhea, mild intestinal tympanites and thymus atrophy. There were no apparent clinical signs of toxicity related to treatment observed in other animals.


2.2 Hematology and Coagulation


Increased NEUT % and decreased LYMPH % were seen in all treated females and in high-dose males. The increases of Fbg in mid- and high-dose animals and APTT in high-dose females were also observed (Table 21).









TABLE 21







Hematology data for monkeys treated with the compound of Formula I


for 4 weeks.













Group
Time
WBC (×109 · L−1)
NEUT (%)
LYMPH (%)
APTT(sec)
Fbg(g · L−1)





Female








  0
d0
12.1 ± 1.9
34.6 ± 9.4
60.9 ± 8.8
16.2 ± 1.9
1.9 ± 0.4


mg/kg
d14
14.3 ± 4.6
38.8 ± 12.0
55.6 ± 11.3
16.4 ± 1.7
2.2 ± 1.0



d26
12.8 ± 2.7
36.7 ± 12.9
55.5 ± 11.8
19.1 ± 1.6
2.1 ± 1.1



d56
13.6 ± 0.3
36.7 ± 10.0
55.2 ± 13.4
18.0 ± 0.6
2.7 ± 1.6


 20
d0
15.3 ± 3.6
42.5 ± 18.8
51.3 ± 18.0
16.7 ± 0.8
2.0 ± 0.3


mg/kg
d14
16.0 ± 3.9
52.5 ± 10.4
40.8 ± 11.3
15.9 ± 1.0
2.9 ± 0.9



d26
19.1 ± 4.0
61.4 ± 13.2**
33.1 ± 12.8*
18.3 ± 1.4
2.8 ± 0.9



d56
12.4 ± 1.6
38.1 ± 7.1
53.1 ± 7.4
16.4 ± 1.1
1.7 ± 0.5


100
d0
10.9 ± 2.9
50.5 ± 17.3
44.0 ± 16.5
15.0 ± 1.6
1.9 ± 0.4


mg/kg
d14
14.0 ± 4.0
59.5 ± 5.3*
33.2 ± 4.4*
14.5 ± 0.9*
2.5 ± 0.4



d26
15.9 ± 6.2
63.2 ± 6.4**
30.0 ± 6.8**
19.7 ± 2.2
3.5 ± 0.8



d56
 9.9 ± 1.0
45.2 ± 2.3
48.7 ± 0.6
15.8 ± 1.5
2.1 ± 0.3


450
d0
14.4 ± 1.7
43.3 ± 12.3
51.8 ± 12.2
16.9 ± 1.4
1.8 ± 0.3


mg/kg
d14
16.9 ± 7.8
63.5 ± 15.2**
30.1 ± 14.3**
18.6 ± 0.8*
3.9 ± 0.8*



d26
18.1 ± 3.7
67.5 ± 9.2**
26.9 ± 8.9**
20.1 ± 3.1
4.2 ± 0.7**



d56
14.0 ± 4.9
42.3 ± 0.6
53.3 ± 0.3
18.0 ± 3.5
1.4 ± 0.0


Male








  0
d0
13.8 ± 4.2
48.3 ± 19.7
44.7 ± 19.6
17.2 ± 1.9
3.1 ± 1.7


mg/kg
d14
15.3 ± 5.6
49.7 ± 9.0
43.2 ± 9.8
17.7 ± 1.6
3.0 ± 1.0



d26
15.3 ± 5.3
48.5 ± 8.4
45.8 ± 7.5
19.5 ± 2.2
2.0 ± 0.3



d56
12.1 ± 2.3
30.3 ± 12.3
62.1 ± 9.2
17.3 ± 1.2
1.8 ± 0.2


 20
d0
13.4 ± 2.5
39.9 ± 14.6
55.3 ± 14.6
16.4 ± 1.1
2.1 ± 0.6


mg/kg
d14
17.7 ± 9.3
43.1 ± 20.4
51.1 ± 19.6
17.8 ± 2.7
3.2 ± 0.8



d26
13.8 ± 6.5
35.1 ± 12.4
59.5 ± 11.9
19.5 ± 2.4
2.2 ± 0.4



d56
20.1 ± 4.2
38.5 ± 12.6
53.3 ± 14.3
21.3 ± 4.0
3.1 ± 1.4


100
d0
12.6 ± 2.3
32.6 ± 18.2
61.5 ± 16.2
16.6 ± 1.8
2.2 ± 0.3


mg/kg
d14
15.6 ± 6.6
45.2 ± 12.2
48.0 ± 12.2
15.5 ± 0.5
2.8 ± 0.7



d26
20.3 ± 2.9
60.8 ± 10.5
32.1 ± 9.4
22.5 ± 2.5
4.1 ± 1.2**



d56
14.1 ± 0.2
42.5 ± 11.2
52.7 ± 9.0
17.7 ± 1.0
1.7 ± 0.3


450
d0
12.0 ± 3.8
46.9 ± 17.3
48.6 ± 16.8
17.0 ± 2.1
2.2 ± 0.7


mg/kg
d14
16.7 ± 6.4
59.2 ± 16.0
35.6 ± 15.5
16.1 ± 0.8
3.1 ± 1.0



d26
18.6 ± 5.9
68.6 ± 12.3*
26.5 ± 12.0*
20.0 ± 3.3
4.1 ± 0.6**



d56
14.6 ± 0.8
36.7 ± 23.2
59.8 ± 22.8
19.9 ± 0.3
1.8 ± 0.0





*p < 0.05, compared with control group,


**p < 0.01, compared with control group Values are mean ± S.D.






2.3 Serum Biochemistry


Serum chemistry parameters CPK, BUN and CREA were significantly increased in high-dose cynomolgus monkeys of both sexes, and especially in the monkey that died early on d23 (d14 data, BUN: 34.99; CREA: 526). CHOL in high-dose males was significantly decreased. Serum Na level of high-dose males decreased slightly, but statistically significantly on d26. Other statistically significant findings were isolated occurrences and considered of no toxicological significance (Table 22).









TABLE 22







Serum chemistry for monkeys treated with the compound of Formula I for 4


weeks.

















BUN
CREA
CHOL
Na
K




CPK
(mmol ·
(μmol ·
(mmol ·
(mmol ·
(mmol ·


Group
Days
(U · L−1)
L−1)
s−1 · L−1)
L−1)
L−1)
L−1)





Female









  0
d0
188.4 ± 83.4
 6.2 ± 1.3
 57.4 ± 4.9
3.3 ± 1.1
150.4 ± 2.3
4.7 ± 0.3


mg/kg
d14
180.0 ± 91.3
 5.5 ± 1.0
 57.8 ± 4.8
3.2 ± 1.2
149.3 ± 3.3
5.4 ± 0.4



d26
147.4 ± 42.2
 5.6 ± 0.6
 51.0 ± 5.5
3.1 ± 0.8
148.3 ± 1.8
5.0 ± 0.5



d56
135.5 ± 34.6
 6.3 ± 0.6
 49.0 ± 4.2
2.4 ± 0.4
147.5 ± 1.6
4.3 ± 0.1


 20
d0
158.4 ± 63.5
 6.1 ± 1.1
 59.0 ± 4.0
3.0 ± 0.5
149.4 ± 3.5
5.0 ± 0.3


mg/kg
d14
151.6 ± 45.2
 5.4 ± 0.9
 52.4 ± 3.8
2.8 ± 0.2
149.6 ± 1.7
5.2 ± 0.2



d26
149.4 ± 58.7
 6.3 ± 1.1
 51.2 ± 3.6
2.7 ± 0.3
149.2 ± 4.3
4.9 ± 0.5



d56
129.0 ± 5.7
 6.0 ± 0.6
 51.5 ± 4.9
3.0 ± 0.1
147.2 ± 4.2
5.0 ± 0.5


100
d0
187.6 ± 75.0
 5.9 ± 1.1
 55.2 ± 4.7
3.8 ± 1.3
149.4 ± 1.9
4.5 ± 0.4


mg/kg
d14
247.2 ± 89.5
 6.4 ± 0.8
 55.2 ± 3.8
2.9 ± 0.6
149.8 ± 3.7
4.7 ± 0.5*



d26
195.0 ± 96.8
 6.4 ± 1.6
 51.2 ± 3.6
3.1 ± 0.5
149.6 ± 3.8
4.8 ± 0.6



d56
112.0 ± 12.7
 5.2 ± 0.2
 54.5 ± 4.9
3.6 ± 0.2
147.9 ± 1.3
4.2 ± 0.2


450
d0
163.2 ± 56.2
 6.1 ± 0.7
 54.4 ± 7.1
3.3 ± 0.4
150.1 ± 2.2
5.1 ± 0.5


mg/kg
d14
815.2 ± 1316.0
14.3 ± 11.8
162.0 ± 204.0
2.4 ± 0.9
144.6 ± 3.5
5.5 ± 0.2



d26
328.5 ± 259.1
 9.7 ± 1.0**
 64.3 ± 7.1*
2.0 ± 0.7
145.0 ± 2.6
5.3 ± 0.4



d56
290.0 ± 176.8
 5.6 ± 0.6
 57.0 ± 2.8
3.1 ± 0.3
145.7 ± 0.9
4.8 ± 0.2


Male









  0
d0
137.2 ± 56.1
 5.6 ± 0.9
 63.6 ± 4.3
3.1 ± 0.4
150.3 ± 3.2
4.9 ± 0.5


mg/kg
d14
152.0 ± 54.9
 5.2 ± 1.0
 59.4 ± 5.4
3.2 ± 0.3
150.5 ± 3.2
5.1 ± 0.3



d26
176.4 ± 91.1
 5.7 ± 0.7
 59.4 ± 3.0
2.9 ± 0.4
149.0 ± 2.0
4.7 ± 0.2



d56
343.5 ± 352.8
 5.8 ± 0.1
 60.0 ± 1.4
2.9 ± 0.2
147.3 ± 3.2
4.6 ± 0.0


 20
d0
156.6 ± 76.3
 5.1 ± 0.9
 64.2 ± 7.2
2.9 ± 0.5
148.7 ± 2.0
5.1 ± 0.3


mg/kg
d14
128.2 ± 50.8
 4.8 ± 0.5
 65.0 ± 4.3
2.6 ± 0.4
148.7 ± 1.6
4.6 ± 0.3



d26
154.4 ± 21.5
 5.6 ± 0.3
 64.0 ± 5.5
2.7 ± 0.5
147.9 ± 1.9
5.4 ± 0.9



d56
161.0 ± 87.7
 5.2 ± 0.4
 64.0 ± 5.7
2.6 ± 0.1
144.6 ± 1.6
5.3 ± 1.1


100
d0
161.4 ± 12.3
 5.7 ± 0.5
 66.6 ± 8.6
3.6 ± 0.4
149.1 ± 2.9
4.8 ± 0.4


mg/kg
d14
213.4 ± 67.1
 6.1 ± 1.5
 63.0 ± 4.1
3.0 ± 0.6
148.3 ± 2.5
4.8 ± 0.4



d26
179.8 ± 42.1
 5.9 ± 1.0
 63.0 ± 8.5
2.8 ± 0.3
147.1 ± 2.4
5.2 ± 0.2



d56
109.0 ± 9.9
 5.7 ± 1.5
 58.5 ± 3.5
2.8 ± 0.6
146.5 ± 0.7
4.2 ± 0.0


450
d0
140.4 ± 44.6
 5.3 ± 0.6
 68.6 ± 5.1
3.4 ± 0.5
148.4 ± 2.5
5.2 ± 0.8


mg/kg
d14
290.2 ± 120.0*
11.3 ± 6.7*
115.8 ± 74.4
1.8 ± 0.3**
146.4 ± 3.3
5.0 ± 0.2



d26
133.6 ± 44.6
10.4 ± 3.1**
 79.0 ± 2.8**
1.7 ± 0.3**
144.3 ± 2.2**
5.2 ± 0.2



d56
 99.5 ± 7.8
 5.1 ± 0.0
 66.5 ± 3.5
2.8 ± 0.3
145.3 ± 1.1
4.8 ± 0.8





*p < 0.05, compared with control group,


**p < 0.01, compared with control group Values are mean ± S.D.






2.4 Urinanalysis


No significant changes were reported in any of the urinalysis parameters examined in either male or female monkeys.


2.5 ECG Parameters


Prolonged QTc intervals were seen in one mid-dose female at 1 h after administration on d26 (296 ms for animal No. 303). Sinus arrhythmia was observed in one high-dose male at 1 h after administration on d26 with prolonged QRS intervals (60 ms for animal No. 411). Prolonged QRS intervals were also seen in high-dose females at 1 h after administration on d1 and d26. (Table 23).









TABLE 23







ECG data for monkeys treated with the compound of Formula I for 4 weeks.














HR
QRS
QT
QTc


Group
Days
(bmp)
(ms)
(ms)
(ms)
















Female


















 0 mg/kg
d 0
187.2 ± 39.6
32.0 ± 4.5
 168.0 ± 19.2
243.1 ± 15.5














1 h
(d 1)
244.4 ± 55.0
32.0 ± 4.5
 154.0 ± 11.4
243.6 ± 12.1



24 h
(d 1)
216.7 ± 18.3
32.0 ± 4.5
 166.0 ± 15.2
254.4 ± 22.8



1 h
(d 26)
259.2 ± 13.4
30.0 ± 0.0
 142.0 ± 17.9
230.8 ± 25.7



24 h
(d 26
256.3 ± 10.7
30.0 ± 0.0
148.0 ± 8.4
240.0 ± 11.0













 d 56
222.8 ± 16.3
25.0 ± 7.1
 170.0 ± 14.1
262.9 ± 15.5


 20 mg/kg
d 0
185.7 ± 48.0
34.0 ± 5.5
 170.0 ± 20.0
244.6 ± 14.3














1 h
(d 1)
230.1 ± 20.5
38.0 ± 4.5
160.0 ± 7.1
250.2 ± 10.4



24 h
(d 1)
209.8 ± 9.3 
34.0 ± 8.9
158.0 ± 4.5
239.7 ± 5.4 



1 h
(d 26)
262.8 ± 13.5
36.0 ± 5.5
140.0 ± 7.1
228.9 ± 8.0 



24 h
(d 26
245.4 ± 43.9
36.0 ± 5.5
 150.0 ± 10.0
238.6 ± 14.1













 d 56
238.0 ± 29.2
40.0 ± 0.0
160.0 ± 0.0
253.1 ± 10.4


100 mg/kg
d 0
182.3 ± 28.9
32.0 ± 4.5
 170.0 ± 12.2
245.0 ± 9.2 














1 h
(d 1)
228.5 ± 12.8
36.0 ± 5.5
160.0 ± 7.1
249.6 ± 6.7 



24 h
(d 1)
217.6 ± 22.7
34.0 ± 5.5
 162.0 ± 11.0
248.2 ± 7.7 



1 h
(d 26)
246.4 ± 40.7
38.0 ± 4.5
 170.0 ± 10.0*
 271.1 ± 17.2**



24 h
(d 26
256.1 ± 17.1
38.0 ± 4.5
148.0 ± 8.4
239.8 ± 10.5













 d 56
230.8 ± 5.0 
30.0 ± 0.0
160.0 ± 0.0
250.7 ± 1.8 


450 mg/kg
d 0
174.1 ± 28.8
32.0 ± 4.5
 168.0 ± 16.4
238.1 ± 12.7














1 h
(d 1)
211.3 ± 20.5
  50.0 ± 14.1**
 175.0 ± 17.3
265.5 ± 19.9



24 h
(d 1)
211.3 ± 2.4 
35.0 ± 5.8
162.5 ± 9.6
247.2 ± 13.8



1 h
(d 26)
262.7 ± 15.4
 40.0 ± 0.2**
140.0 ± 8.2
229.0 ± 14.6



24 h
(d 26
257.8 ± 9.0 
32.5 ± 5.0
152.5 ± 9.6
247.8 ± 14.4













 d 56
207.4 ± 28.1
25.0 ± 7.1
 170.0 ± 14.1
256.3 ± 9.7 













Male


















 0 mg/kg
d 0
214.3 ± 12.6
36.0 ± 8.9
166.0 ± 8.9
253.7 ± 15.6














1 h
(d 1)
248.4 ± 16.7
36.0 ± 5.5
152.0 ± 4.5
243.9 ± 6.6 



24 h
(d 1)
208.5 ± 18.5
34.0 ± 5.5
 170.0 ± 14.1
256.9 ± 15.8



1 h
(d 26)
253.9 ± 12.1
32.0 ± 4.5
148.0 ± 8.4
239.2 ± 10.5



24 h
(d 26
232.1 ± 43.2
38.0 ± 8.4
156.0 ± 8.9
243.3 ± 8.7 













 d 56
194.5 ± 19.5
40.0 ± 0.0
180.0 ± 0.0
266.3 ± 8.9 


 20 mg/kg
d 0
198.4 ± 16.5
34.0 ± 8.9
 164.0 ± 11.4
243.9 ± 11.7














1 h
(d 1)
216.0 ± 27.0
32.0 ± 4.5
 158.0 ± 11.0
241.4 ± 12.3



24 h
(d 1)
206.1 ± 20.7
28.0 ± 4.5
166.0 ± 8.9
250.0 ± 10.4



1 h
(d 26)
260.0 ± 12.4
36.0 ± 5.5
 146.0 ± 11.4
237.8 ± 16.3



24 h
(d 26
264.9 ± 16.1
34.0 ± 5.5
 146.0 ± 11.4
239.2 ± 14.8













 d 56
225.6 ± 2.4 
30.0 ± 0.0
145.0 ± 7.1
225.4 ± 10.2


100 mg/kg
d 0
215.4 ± 11.2
32.0 ± 8.4
 154.0 ± 16.7
235.6 ± 24.5














1 h
(d 1)
230.0 ± 19.9
38.0 ± 4.5
 156.0 ± 11.4
243.8 ± 16.5



24 h
(d 1)
208.2 ± 10.4
36.0 ± 8.9
 162.0 ± 11.0
245.3 ± 18.6



1 h
(d 26)
256.9 ± 16.7
36.0 ± 5.5
144.0 ± 5.5
233.8 ± 12.2



24 h
(d 26
260.7 ± 9.1 
36.0 ± 5.5
144.0 ± 8.9
234.9 ± 13.9













 d 56
225.6 ± 2.4 
35.0 ± 7.1
165.0 ± 7.1
256.6 ± 11.9


450 mg/kg
d 0
214.3 ± 17.7
34.0 ± 5.5
156.0 ± 5.5
238.2 ± 7.2 














1 h
(d 1)
217.4 ± 3.2 
36.7 ± 5.8
 170.0 ± 10.0
261.1 ± 16.0



24 h
(d 1)
211.0 ± 15.9
32.0 ± 4.5
166.0 ± 8.9
252.1 ± 9.4 



1 h
(d 26)
247.6 ± 22.6
 36.0 ± 13.4
 150.0 ± 17.3
240.0 ± 23.7



24 h
(d 26
210.4 ± 49.9
38.0 ± 4.5
 158.0 ± 13.0
237.7 ± 12.2













 d 56
203.3 ± 15.5
35.0 ± 7.1
160.0 ± 0.0
240.2 ± 6.1 





*p < 0.05, compared with control group,


**p < 0.01, compared with control group.


Values are mean ± S.D.






2.6 Necropsy


2.6.1 Gross Pathology and Organ Weights


There were no macroscopic findings for monkeys deemed related to administration of the test article except mild intestinal tympanites and thymus atrophy for the monkey that died early. Higher relative weight of liver and kidney, lower relative weight of thymus and lower absolute or relative weight of uterus occurred in high-dose monkeys (Table 24).









TABLE 24







Organ weight for cynomolgus monkeys treated


with the compound of Formula I for 4 weeks.












Group
Time
Liver
kidney
Thymus
Uterus












Female

Absolute weight(g)












 0 mg/kg
d 28
58.43 ± 5.40 
12.17 ± 1.46
1.60 ± 0.46
8.33 ± 0.51



d 56
69.80 ± 6.51 
14.55 ± 0.64
1.95 ± 2.05
6.05 ± 2.76


 20 mg/kg
d 28
58.37 ± 7.38 
12.20 ± 2.23
1.23 ± 0.40
5.97 ± 1.20



d 56
68.35 ± 3.32 
13.75 ± 0.78
2.00 ± 0.71
11.30 ± 3.25 


100 mg/kg
d 28
57.60 ± 4.25 
11.67 ± 0.71
0.73 ± 0.50
7.03 ± 0.94



d 56
68.45 ± 20.01
14.15 ± 2.33
2.35 ± 0.49
8.30 ± 3.39


450 mg/kg
d 28
69.40 ± 3.68 
12.95 ± 0.07
0.50 ± 0.14
 3.50 ± 0.14**



d 56
72.40 ± 11.74
14.75 ± 1.91
1.40 ± 0.14
9.60 ± 2.40


Male







 0 mg/kg
d 28
74.10 ± 3.60 
15.80 ± 2.55
1.47 ± 0.83




d 56
82.90 ± 16.40
19.15 ± 0.49
1.75 ± 0.92



 20 mg/kg
d 28
69.03 ± 2.87 
14.30 ± 1.42
1.10 ± 0.20




d 56
71.25 ± 1.77 
16.40 ± 3.11
2.95 ± 0.07



100 mg/kg
d 28
72.93 ± 14.69
15.13 ± 3.62
1.57 ± 0.90




d 56
84.75 ± 5.59 
17.85 ± 1.91
2.45 ± 0.35



450 mg/kg
d 28
94.37 ± 12.72
16.77 ± 2.31
0.90 ± 0.10




d 56
80.90 ± 4.95 
18.75 ± 0.21
2.20 ± 0.42










Female

Relative weight (/body weight)












 0 mg/kg
d 28
1.83 ± 0.22
 0.38 ± 0.023
0.05 ± 0.01
0.26 ± 0.03



d 56
2.12 ± 0.08
 0.45 ± 0.08
0.06 ± 0.05
0.18 ± 0.06


 20 mg/kg
d 28
2.06 ± 0.22
 0.43 ± 0.07
0.04 ± 0.01
0.21 ± 0.04



d 56
2.01 ± 0.07
 0.41 ± 0.06
0.06 ± 0.02
0.33 ± 0.07


100 mg/kg
d 28
1.96 ± 0.13
 0.40 ± 0.02
0.02 ± 0.02
0.24 ± 0.03



d 56
1.86 ± 0.37
 0.39 ± 0.03
0.07 ± 0.02
0.23 ± 0.12


450 mg/kg
d 28
 2.78 ± 0.31**
 0.52 ± 0.03*
0.02 ± 0.01
 0.14 ± 0.01*



d 56
2.04 ± 0.05
 0.42 ± 0.00
0.04 ± 0.01
0.27 ± 0.03


Male







 0 mg/kg
d 28
1.77 ± 0.15
 0.38 ± 0.04
0.03 ± 0.02




d 56
1.60 ± 0.25
 0.37 ± 0.01
0.03 ± 0.02



 20 mg/kg
d 28
1.82 ± 0.21
 0.37 ± 0.02
0.03 ± 0.01




d 56
1.44 ± 0.02
 0.33 ± 0.06
0.06 ± 0.00



100 mg/kg
d 28
1.83 ± 0.25
 0.37 ± 0.06
0.04 ± 0.03




d 56
1.76 ± 0.06
 0.37 ± 0.03
0.05 ± 0.01



450 mg/kg
d 28
 2.42 ± 0.20*
 0.43 ± 0.03
0.02 ± 0.01




d 56
1.64 ± 0.22
 0.38 ± 0.02
0.04 ± 0.01










Female

Relative weight (/brain weight)












 0 mg/kg
d 28
97.71 ± 14.77
20.21 ± 1.89
2.66 ± 0.74
13.93 ± 1.90 



d 56
108.79 ± 9.78 
22.68 ± 1.07
3.03 ± 3.19
9.42 ± 4.27


 20 mg/kg
d 28
90.10 ± 7.72 
18.73 ± 1.74
1.87 ± 0.46
9.19 ± 1.57



d 56
104.00 ± 10.62 
20.86 ± 0.06
3.07 ± 1.24
17.30 ± 5.86 


100 mg/kg
d 28
96.55 ± 7.51 
19.61 ± 1.86
1.18 ± 0.73
11.84 ± 2.02 



d 56
104.90 ± 37.06 
21.60 ± 4.92
3.54 ± 0.52
12.44 ± 4.34 


450 mg/kg
d 28
112.03 ± 12.42 
20.87 ± 1.10
 0.81 ± 0.27*
 5.63 ± 0.10**



d 56
109.03 ± 13.64 
22.23 ± 2.05
2.12 ± 0.29
14.43 ± 3.09 


Male







 0 mg/kg
d 28
101.35 ± 5.42 
21.65 ± 3.87
2.02 ± 1.18




d 56
111.94 ± 22.78 
25.85 ± 0.82
2.36 ± 1.23



 20 mg/kg
d 28
103.68 ± 11.79 
21.46 ± 2.99
1.67 ± 0.43




d 56
100.96 ± 1.74 
23.16 ± 3.43
4.18 ± 0.08



100 mg/kg
d 28
100.31 ± 9.48 
20.85 ± 3.95
2.27 ± 1.44




d 56
115.98 ± 8.59 
24.36 ± 0.80
3.33 ± 0.02



450 mg/kg
d 28
135.54 ± 14.14*
24.07 ± 2.45
1.30 ± 0.20




d 56
119.80 ± 2.56 
27.81 ± 1.42
3.25 ± 0.50





*p < 0.05, compared with control group,


**p < 0.01, compared with control group.


Values are mean ± S.D.






2.7 Toxicokinetics


Systemic exposure, which was indicated by the plasma AUC0-24h and Cmax of parent compound and main metabolite, were proportionately greater than the magnitude of the dose increments in the dose range of 20 to 450 mg/kg. After consecutive 28 days of administration, the accumulation was not apparent either.


Example 3: Nonclinical Pharmacokinetics

1. Materials and Methods


1.1 Materials


The compound of Formula I was prepared and confirmed according to the method in CN103562191B.


1.2 Animals


Cynomolgus monkeys (3/sex/group: weight 3-5 kg, age 5-6 years old) were obtained from SuZhou XiShan ZhongKe Laboratory Animals (Shanghai, China). Animals were housed for a 7-day acclimation period before study. The monkeys were individually caged in a controlled temperature (18-26° C.) and humidity (50±20%) animal room with an air flow of 10 plus air changes per hour with fresh air. The lighting cycle was 12 h on and 12 h off, and the monkeys were fed once a day in the morning. All animal experiments were performed in accordance with the Animal Care and Use Guidelines set by the AMMS Animal Care and Use Committee.


2. Intravenous and Oral Administration of the Compound of Formula I


The compound of Formula I was intravenously or orally administered to cynomolgus monkeys fasted overnight. The doses were 1 mg/kg (intravenous), 1 mg/kg, 3 mg/kg, and 9 mg/kg (oral). Blood samples were collected from a limb vein at 0, 5, and 15 min and at 0.25, 0.5, 1, 2, 3, 4, 6, 8, 12, and 24 h (0.2 mL for each time point) after administration. All blood samples were centrifuged immediately after collection at 10,000×g for 5 min and the plasmas were stored at −75±10° C. until analysis. Plasma samples were prepared by protein precipitation. The concentrations of the compound of Formula I in plasma were determined by the validated liquid chromatography-tandem mass spectrometry method.


The pharmacokinetic parameters such as the area under the plasma concentration-time curve (AUC), maximum plasma concentration (Cmax), time to reach Cmax (Tmax), half-life (T1/2), total body plasma clearance (CL), mean residence time (MRT) and volume of distribution at steady state (Vss) were calculated by non-compartmental analysis with WinNonlin v1.3 (Pharsight, Mountain View, CA, USA). For allometric scaling, animal pharmacokinetic data were simulated by a two-compartment model with the Kinetica 5.1 software package (v.3.0; InnaPhase, Philadelphia, PA, USA).


3. Results


Plasma Pharmacokinetics and Bioavailability


Table 25 and FIG. 2 showed the pharmacokinetic data after intravenous and oral doses in cynomolgus monkeys. Drug concentration declined biexponentially after intravenous administration with a clearance of 1.96 L/h/kg and distribution volume of 1.86 L/kg for monkeys. After oral administration, the compound of Formula I reached a Cmax at 1.0-1.30 h, and disappeared with a T1/2 of 2.73-5.99 h in monkey plasma. The oral bioavailability of the compound of Formula I was about 3.3-11.3% in monkeys calculated from the AUC0-24h after iv and poadministration, which indicated that efflux transporters were involved.





















TABLE 25






Dose

CL
Vss
MRT
t1/2
Cmax
Tmax
AUCtot.
CL/F
Vss/F
F


Species
mg/kg
Route
L/h/kg
L/kg
h
h
ng/ml
h
ng*h/ml
L/h/kg
L/kg
(%)



























Monkey
1
iv
1.96
1.86
0.97
2.73
466

421






1
po


4.32
2.90
3.99
1.0
11.8
84.8
366
3.3



3
po


3.46
3.68
42.2
1.3
99.4
30.2
104
8.0



9
po


2.62
5.99
263
1.0
426
21.2
55.5
11.3





CL clearance after iv, Vss volume of distribution at steady state after iv, MRT mean residence time, T1/2 half-life, Cmax maximum plasma concentration, Tmax time to reach Cmax, AUCtot the total area under the plasma concentration-time curve, CL/F clearance after po, Vss/F volume of distribution at steady state after po, F absolute oral bioavailability, iv intrravenous, po oral.





Claims
  • 1. A method of treating or preventing chronic kidney disease comprising administering a nitrogen-containing saturated heterocyclic compound represented by the following formula I or a pharmaceutically acceptable salt thereof:
  • 2. The method according to claim 1, wherein, the pharmaceutically acceptable salt is hydrochloride, sulfate, phosphate, hydrobromide, acetate, fumarate, oxalate, citrate, methanesulfonate, benzenesulfonate, p-toluenesulfonate, maleate, malate, tartrate, acetate or naphthalene disulfonate.
  • 3. The method according to claim 1, wherein, the pharmaceutically acceptable salt is the malate of the compound of formula I, which is a compound formed by the compound of formula I and malic acid at a molar ratio of 1:1, and the structural formula thereof is as follows:
  • 4. The method according to claim 3, wherein, the malate is a crystal form, wherein the X-ray powder diffraction pattern thereof has characteristic peaks at 2θ of 7.767°±0.2°, 13.897°±0.2°, 14.775°±0.2°, 17.098°±0.2°, 18.999°±0.2°, 20.153±0.2°, 20.960°±0.2°, 21.423°±0.2°, 26.348°±0.2° and 27.892°±0.2°.
  • 5. (canceled)
  • 6. The method according to claim 1, wherein, the chronic kidney disease is hypertension with nephropathy, hypertension with nephropathy with abnormal glucose metabolism, chronic renal insufficiency with chronic heart failure or chronic kidney disease with abnormal glucose metabolism.
  • 7. The method according to claim 6, wherein, the hypertension in hypertension with nephropathy is grade 1 hypertension, grade 2 hypertension or grade 3 hypertension.
  • 8. The method according to claim 1, wherein, the chronic kidney disease refers to the G1, G2, G3a, G3b, or G4 stages of chronic kidney disease.
  • 9. The method according to claim 1, wherein, the compound is administered as a medicine comprising dosage forms selected from a tablet, a capsule, an intravenous injection, an inhalant, a nebulizer, a lyophilized agent, a patch, a gel, a spray, and a suppository.
  • 10. The method according to claim 9, wherein, the medicine is a unit dose.
  • 11. The method according to claim 4, wherein the X-ray powder diffraction pattern of the malate crystal further has characteristic peaks at 2θ of 5.598°±0.2°, 7.357°±0.2°, 10.395°±0.2°, 11.108°±0.2°, 16.037°±0.2°, 16.523°±0.2°, 19.410°±0.2°, 22.645°±0.2°, 26.630°±0.2° 26.891°±0.2°, 27.380°±0.2°, 31.056°±0.2°, 33.306°±0.2°, 33.775°±0.2° and 39.231°±0.2°.
  • 12. The method of claim 10, wherein the unit dose contains the nitrogen-containing saturated heterocyclic compound represented by formula I or the pharmaceutically acceptable salt thereof in a range of 25 mg to 200 mg.
  • 13. The method of claim 10, wherein the unit dose contains 25 mg, 50 mg, 100 mg, 150 mg, 200 mg of the nitrogen-containing saturated heterocyclic compound represented by formula I or the pharmaceutically acceptable salt thereof.
Priority Claims (1)
Number Date Country Kind
202010921155.4 Sep 2020 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2021/116338 9/3/2021 WO