loader
Please Wait
Applying Filters...

AbbVie Contract Manufacturing AbbVie Contract Manufacturing

X

Technical details about MolPort-001-662-635, learn more about the structure, uses, toxicity, action, side effects and more

Client Email Product
Menu
2D Structure
1. Also known as: 2-aminoethanethiol, 60-23-1, Mercaptamine, Thioethanolamine, Becaptan, Mercamine
Molecular Formula
C2H7NS
Molecular Weight
77.15  g/mol
InChI Key
UFULAYFCSOUIOV-UHFFFAOYSA-N
FDA UNII
5UX2SD1KE2

A mercaptoethylamine compound that is endogenously derived from the COENZYME A degradative pathway. The fact that cysteamine is readily transported into LYSOSOMES where it reacts with CYSTINE to form cysteine-cysteamine disulfide and CYSTEINE has led to its use in CYSTINE DEPLETING AGENTS for the treatment of CYSTINOSIS.
Cysteamine is a Cystine Depleting Agent. The mechanism of action of cysteamine is as a Cystine Disulfide Reduction.
1 2D Structure

2D Structure

2 Identification
2.1 Computed Descriptors
2.1.1 IUPAC Name
2-aminoethanethiol
2.1.2 InChI
InChI=1S/C2H7NS/c3-1-2-4/h4H,1-3H2
2.1.3 InChI Key
UFULAYFCSOUIOV-UHFFFAOYSA-N
2.1.4 Canonical SMILES
C(CS)N
2.2 Other Identifiers
2.2.1 UNII
5UX2SD1KE2
2.3 Synonyms
2.3.1 MeSH Synonyms

1. 2 Aminoethanethiol

2. 2-aminoethanethiol

3. 35s-labeled Cysteamine

4. Becaptan

5. Beta Mercaptoethylamine

6. Beta-mercaptoethylamine

7. Bitartrate, Cysteamine

8. Cystagon

9. Cysteamine Bitartrate

10. Cysteamine Dihydrochloride

11. Cysteamine Hydrobromide

12. Cysteamine Hydrochloride

13. Cysteamine Maleate (1:1)

14. Cysteamine Tartrate

15. Cysteamine Tartrate (1:1)

16. Cysteamine Tosylate

17. Cysteamine, 35s Labeled

18. Cysteamine, 35s-labeled

19. Cysteinamine

20. Dihydrochloride, Cysteamine

21. Hydrobromide, Cysteamine

22. Hydrochloride, Cysteamine

23. Mercamine

24. Mercaptamine

25. Mercaptoethylamine

26. Tartrate, Cysteamine

27. Tosylate, Cysteamine

2.3.2 Depositor-Supplied Synonyms

1. 2-aminoethanethiol

2. 60-23-1

3. Mercaptamine

4. Thioethanolamine

5. Becaptan

6. Mercamine

7. Beta-mercaptoethylamine

8. 2-mercaptoethylamine

9. Cysteinamine

10. Cysteamin

11. Lambraten

12. Lambratene

13. Beta-aminoethanethiol

14. Cystagon

15. Riacon

16. Decarboxycysteine

17. Mercaptoethylamine

18. 2-amino-1-ethanethiol

19. Ethanethiol, 2-amino-

20. Cisteamina

21. 2-aminoethyl Mercaptan

22. Beta-aminoethylthiol

23. 2-mercaptoethanamine

24. Mercaptamin

25. (2-mercaptoethyl)amine

26. Beta-mea

27. Mea (mercaptan)

28. Ethanethiolamine

29. 2-amino-ethanethiol

30. Aminoethyl Mercaptan

31. Mercaptamina

32. Mercaptaminum

33. Cysteamide

34. Mecramine

35. Mercamin

36. Merkamin

37. Cisteamina [italian]

38. Cystaran

39. Mercaptaminum [inn-latin]

40. Mercaptamina [inn-spanish]

41. 1-amino-2-mercaptoethylamine

42. L-1573

43. Cysteamine (usan)

44. Cysteamine [usan]

45. Wr 347

46. 2-aminoethane-1-thiol

47. B-mercaptoethylamine

48. Nsc 647528

49. Mercaptamine (inn)

50. Mercaptamine [inn]

51. .beta.-mercaptoethylamine

52. Nsc-647528

53. 5ux2sd1ke2

54. Chebi:17141

55. 2-aminoethane-1-thiol; Cysteamine

56. Nsc647528

57. Ncgc00015691-03

58. Cysteamine [usan:ban]

59. Beta-mercaptoethylamine;2-aminoethanethiol;2-mercaptoethylamine

60. Cysteamine Bitartate

61. Ccris 3083

62. Hsdb 7353

63. Einecs 200-463-0

64. L 1573

65. Unii-5ux2sd1ke2

66. Cystagone

67. B-aminoethylthiol

68. B-mea

69. B-aminoethanethiol

70. Mercapto Ethylamine

71. Dhl

72. Mfcd00008196

73. 2-amino-ethyl Thiol

74. Cash

75. .beta.-mea

76. Cysteamine, ~95%

77. Cysteamine, Free Base

78. .beta.-aminoethylthiol

79. Spectrum_001755

80. .beta.-aminoethanethiol

81. Cysteamine [mi]

82. Specplus_000654

83. Cysteamine [hsdb]

84. Cysteamine [inci]

85. Lopac-m-6500

86. Dsstox_cid_2875

87. Cysteamine [vandf]

88. Bmse000388

89. Chembl602

90. C-9500

91. Dsstox_rid_76770

92. Dsstox_gsid_22875

93. Kbioss_002235

94. Mercaptamine [mart.]

95. Divk1c_006750

96. Mercaptamine [who-dd]

97. Bdbm7968

98. Gtpl7440

99. 2,3-deshydrolanthionine

100. Dtxsid3022875

101. Kbio1_001694

102. Kbio2_002235

103. Kbio2_004803

104. Kbio2_007371

105. Cysteamine, >=98.0% (rt)

106. Bcp15015

107. Zinc8034121

108. Tox21_113092

109. Hy-77591a

110. S6402

111. Stk315355

112. Akos003793343

113. Ccg-204834

114. Db00847

115. Sb75360

116. Sdccgsbi-0050727.p004

117. Cas-60-23-1

118. Ncgc00015691-01

119. Ncgc00015691-02

120. Ncgc00015691-04

121. Ncgc00015691-14

122. Ncgc00162236-01

123. Ncgc00162236-02

124. Amy202100105

125. As-56218

126. Bp-13401

127. Nci60_002000

128. Sbi-0050727.p003

129. Db-053562

130. A0648

131. Ft-0611243

132. C01678

133. D03634

134. D88299

135. D88360

136. Ab00053754_09

137. Ab00053754_10

138. A832648

139. Q617563

140. F0001-1576

141. 2dfda1f8-7010-4225-8280-ab1c4c43f546

142. 2-aminoethanethiol;cysteamine, Beta-mercaptoethylamine, 2-mercaptoethylamine, Decarboxycysteine, Thioethanolamine, Mercaptamine

143. 2-mercaptoethylamine, Polymer-bound, 70-90 Mesh, Extent Of Labeling: 0.6-1.1 Mmol/g Loading, 1 % Cross-linked With Divinylbenzene

2.4 Create Date
2004-09-16
3 Chemical and Physical Properties
Molecular Weight 77.15 g/mol
Molecular Formula C2H7NS
XLogP3-0.4
Hydrogen Bond Donor Count2
Hydrogen Bond Acceptor Count2
Rotatable Bond Count1
Exact Mass77.02992040 g/mol
Monoisotopic Mass77.02992040 g/mol
Topological Polar Surface Area27 Ų
Heavy Atom Count4
Formal Charge0
Complexity10
Isotope Atom Count0
Defined Atom Stereocenter Count0
Undefined Atom Stereocenter Count0
Defined Bond Stereocenter Count0
Undefined Bond Stereocenter Count0
Covalently Bonded Unit Count1
4 Drug and Medication Information
4.1 Drug Information
1 of 2  
Drug NameCYSTAGON
Active IngredientCYSTEAMINE BITARTRATE
CompanyMYLAN (Application Number: N020392)

2 of 2  
Drug NameCYSTARAN
Active IngredientCYSTEAMINE HYDROCHLORIDE
CompanyLEADIANT BIOSCI INC (Application Number: N200740)

4.2 Therapeutic Uses

Cysteamine is indicated for the management of nephropathic cystinosis in children and adults. /Included in US product labeling/

Thomson.Micromedex. Drug Information for the Health Care Professional. 25th ed. Volume 1. Plus Updates. Content Reviewed by the United States Pharmacopeial Convention, Inc. Greenwood Village, CO. 2005.


Nephropathic cystinosis is a rare, autosomal recessive lysosomal storage disorder caused by mutations in the CTNS gene that codes for a cystine transporter in the lysosomal membrane. Affected patients store 50-100 times the normal amounts of cystine in their cells, and suffer renal tubular and glomerular disease, growth retardation, photophobia, and other systemic complications, including a myopathy and swallowing dysfunction. Using videofluoroscopy and ultrasound examinations, we assessed the swallowing function of 101 patients with nephropathic cystinosis on their most recent admission to the National Institutes of Health Clinical Center between 1987 and 2004. These patients ranged in age from 6 to 45 years; more than half had significant complaints of swallowing difficulty. On examination of barium swallow, the oral, pharyngeal, and esophageal phases of swallowing were abnormal in 24%, 51%, and 73% of patients, respectively. The frequency of dysfunction increased with age for each phase of swallowing. Both the Swallowing Severity Score (a measure of dysfunction on barium swallow) and the Oral Muscle Composite Score (a reflection of vocal strength, oral-facial movement, and tongue and lip function) increased (that is, worsened) with the number of years that a patient was not receiving treatment with cysteamine, the cystine-depleting agent of choice in cystinosis. The severity scores decreased with the number of years on cysteamine therapy. The Swallowing Severity Score varied directly with the severity of muscle disease, but was not correlated with the presence or absence of the 57-kb CTNS deletion that commonly occurs in nephropathic cystinosis patients. We conclude that swallowing dysfunction in cystinosis presents a risk of fatal aspiration, correlates with the presence of muscle atrophy, and, based on cross-sectional data, increases in frequency with age and number of years without cysteamine treatment. Cystine-depleting therapy with cysteamine should be considered the treatment of choice for both pre- and posttransplant cystinosis patients.

PMID:15879904 Sonies BC et al; Medicine (Baltimore) 84 (3): 137-46 (2005)


/EXPL THER/ N-Acetylcysteine (NAC) is protective against acetaminophen-induced hepatotoxicity primarily by providing precursor for the glutathione synthetase pathway, while cysteamine has been demonstrated to alter the cytochrome P-450 dependent formation of toxic acetaminophen metabolite. Mice administered acetaminophen (500 mg/kg) had elevations of serum alanine aminotransferase (ALT) to 273.0 +/- 37.5 and 555.8 +/- 193.4 U/mL at 12 and 24 h, respectively, after injection. Administration of cysteamine (100 mg/kg) or NAC (500 mg/kg) significantly reduced serum ALT activity (p less than 0.001). Reducing the dose of NAC or cysteamine by 50% greatly reduced their hepatoprotective effect while the co-administration of the reduced doses of NAC (250 mg/kg) and cysteamine (50 mg/kg) following acetaminophen overdose prevented elevation of serum ALT activity (39.2 +/- 1.17 and 32.5 +/- 5.63 U/mL at 12 and 24 h post-injection, p less than 0.001) and preserved normal mouse hepatic histology. Neither NAC (500 mg/kg), cysteamine (100 mg/kg), or the lower doses in combination of both agents were found to alter the half-life or peak levels of acetaminophen. Liver microsomal aryl hydrocarbon hydroxylase activity measured 24 h after drug administration was not significantly different between treatment groups and controls receiving only saline. These results indicate a possible role for the concomitant use of NAC and cysteamine in the prevention of hepatic necrosis following toxic doses of acetaminophen. Neither decrease in plasma acetaminophen levels nor depression of cytochrome P-450 enzyme activity appears to be the mechanism of protection when these doses of NAC, cysteamine, or both drugs together are administered with a toxic dose of acetaminophen in mice.

PMID:1581851 Peterson TC, Brown IR; Can J Physiol Pharmacol 70 (1): 20-8 (1992)


Although renal disease is the most prominent feature of the lysosomal storage disease cystinosis, corneal cystine crystal formation remains a major complication, leading to photophobia, corneal erosions, and keratopathies. Moreover, the extent of corneal crystal accumulation reflects the course and severity of the disease itself, and the cornea is accessible to direct examination. Therefore, we employed a scoring system, based on a library of slit-lamp photographs of corneas with increasing crystal densities (0.00-3.00), to assess the degree of crystal accumulation in 170 patients with nephropathic cystinosis examined at the National Institutes of Health between 1976 and 2000. None of the patients had received topical cystine-depleting therapy at the time of the evaluation. In this natural history study, infants in the first year of life had absent or minimal corneal crystals, i.e., a corneal cystine crystal score (CCCS) of 0 or 0.25. However, the CCCS increased linearly with age, such that every patient had visible crystals by 16 months of age, and plateaued at approximately 3.00 by early adolescence. Longitudinal studies in representative patients support the cross-sectional results. Individuals homozygous for the common 57-kb deletion involving the cystinosis gene (CTNS) displayed the same course of corneal crystal accumulation as did individuals not bearing the large deletion. Patients with ocular or nonnephropathic cystinosis had CCCSs that were, in general, half those expected for patients with nephropathic cystinosis of the same age. Administration of 0.55% cysteamine eyedrops, given 6 to 12 times per day, dissolved corneal cystine crystals in 10 representative patients with nephropathic cystinosis aged 1 to 32 years within 8 to 41 months.

PMID:11001803 Gahl WA et al; Mol Genet Metab 71 (1-2): 100-20 (2000)


The necessity to apply near-toxic amounts of radioprotective drugs to achieve adequate protection during radiation treatments represents a major problem in human medicine. One of the promising strategies to suppress the toxicity of these drugs involves their incorporation into biocompatible polymers. In this study cysteamine (Cy) was attached to poly(oxyethylene phosphate), POEP, via an ionic bond. Radioprotection of E. coli B cells by this substance and its acute toxicity on male C57 BL mice were measured. The toxicity of Cy immobilized within the poly(oxyethylene phosphate) was significantly lower in comparison to pure Cy while its radioprotective efficiency remained high at half the maximum tolerable dose. The high radioprotective efficiency of the Cy/POEP complexes was further confirmed on mice at different polymer molecular weight characteristics, drug immobilization degrees, application times, and doses. It was found that POEP with molecular weight 4700 Da and containing 24% repeating units with attached Cy has the highest protection potential combined with a depot effect.

PMID:12477362 Georgieva R et al; J Med Chem 45 (26): 5797-801 (2002)


4.3 Drug Warning

The most frequent adverse reactions seen involve the GI and central nervous systems. These are especially prominent at the initiation of therapy. Temporarily suspending treatment, then gradual reintroduction may be effective in improving tolerance. The most common events (> 5%) were vomiting (35%), anorexia (31%), fever (22%), diarrhea (16%), lethargy 11%) and rash (7%). Other adverse reactions are as follows: CNS: Somnolence; encephalopathy; headache; seizures; ataxia; confusion; tremor; hyperkinesia; decreased hearing; dizziness; jitteriness. GI: Nausea; bad breath; abdominal pain; dyspepsia; constipation; gastroenteritis; duodenitis; duodenal ulceration. Psychiatric: Nervousness; abnormal thinking; depression; emotional lability; hallucinations; nightmares. Miscellaneous: Abnormal liver function; anemia; leukopenia; dehydration; hypertension; urticaria.

Novak, K.M. (ed.). Drug Facts and Comparisons 59th Edition 2005. Wolters Kluwer Health. St. Louis, Missouri 2005., p. 738


Cysteamine has occasionally been associated with reversible leukopenia and abnormal liver function studies. Therefore, monitor blood counts and liver function studies.

Novak, K.M. (ed.). Drug Facts and Comparisons 59th Edition 2005. Wolters Kluwer Health. St. Louis, Missouri 2005., p. 738


Patients sensitive to penicillamine may be sensitive to this medication also.

Thomson.Micromedex. Drug Information for the Health Care Professional. 25th ed. Volume 1. Plus Updates. Content Reviewed by the United States Pharmacopeial Convention, Inc. Greenwood Village, CO. 2005.


FDA Pregnancy category C: Adequate, well controlled human studies are lacking, and animal studies have shown risk to the fetus or are lacking as well. There is a chance of fetal harm if the drug is given during pregnancy; but the potential benefits may outweigh the potential risk.

Thomson.Micromedex. Drug Information for the Health Care Professional. 25th ed. Volume 1. Plus Updates. Content Reviewed by the United States Pharmacopeial Convention, Inc. Greenwood Village, CO. 2005.


For more Drug Warnings (Complete) data for CYSTEAMINE (11 total), please visit the HSDB record page.


4.4 Drug Indication

The bitartrate salt of cysteamine is used for the oral treatment of nephropathic cystinosis and cystinuria in children 6 years old and above, and adults. The hydrochloride salt, used in eye drop preparations, is indicated for the treatment of corneal cystine crystal accumulation in patients with cystinosis.


FDA Label


Cystadrops is indicated for the treatment of corneal cystine crystal deposits in adults and children from 2 years of age with cystinosis.


Treatment of corneal cystine deposits,


5 Pharmacology and Biochemistry
5.1 Pharmacology

Cystine accumulation is the cause of organ damage in cystinosis. Cysteamine prevents the accumulation of cystine crystals in the body and is specifically prescribed to prevent kidney and eye damage. Cysteamine converts cystine into a form that may easily exit cells, preventing harmful accumulation.


5.2 MeSH Pharmacological Classification

Cystine Depleting Agents

Compounds and drugs that react with CYSTINE and convert it into a compound that can be more easily metabolized or intracellularly transported. Drugs in this class have been used to treat CYSTINOSIS. (See all compounds classified as Cystine Depleting Agents.)


5.3 FDA Pharmacological Classification
5.3.1 Active Moiety
CYSTEAMINE
5.3.2 FDA UNII
5UX2SD1KE2
5.3.3 Pharmacological Classes
Mechanisms of Action [MoA] - Cystine Disulfide Reduction
5.4 ATC Code

S01XA21


S01XA21


A16AA04

S76 | LUXPHARMA | Pharmaceuticals Marketed in Luxembourg | Pharmaceuticals marketed in Luxembourg, as published by d'Gesondheetskeess (CNS, la caisse nationale de sante, www.cns.lu), mapped by name to structures using CompTox by R. Singh et al. (in prep.). List downloaded from https://cns.public.lu/en/legislations/textes-coordonnes/liste-med-comm.html. Dataset DOI:10.5281/zenodo.4587355


A - Alimentary tract and metabolism

A16 - Other alimentary tract and metabolism products

A16A - Other alimentary tract and metabolism products

A16AA - Amino acids and derivatives

A16AA04 - Mercaptamine


S - Sensory organs

S01 - Ophthalmologicals

S01X - Other ophthalmologicals

S01XA - Other ophthalmologicals

S01XA21 - Mercaptamine


5.5 Absorption, Distribution and Excretion

Absorption

Orally administered cysteamine is absorbed in the gastrointestinal tract and reaches its maximum plasma concentration in about 1.4 hours, with some variation according to the type of formulation (delayed versus immediate-release). One pharmacokinetic study of adults with Cystic Fibrosis revealed a Cmax of 2.86 mg/L.The maximum plasma concentration after administration of cysteamine eye drops is unknown, however, it is likely to be considerably lower than oral administration. According to prescribing information, the AUC 0-12 h for the delayed-release oral tablets is 99.26 44.2 mol*h/L with a Cmax of 27.70 14.99 mol/L. The AUC 0-12 for the immediate-release tablets is 192.00 75.62 mol*h/L with a Cmax of 37.72 12.10 mol/L.


Volume of Distribution

Cysteamine has a volume of distribution of about 129 L, according to one pharmacokinetic study. Prescribing information indicates a volume of distribution of 382 L for the delayed-release formulation and 198 L for the immediate-release preparation. It is known to cross the blood-brain barrier.


Clearance

The plasma clearance of cysteamine is about 1.2 - 1.4 L/min. One reference mentions a clearance of 89.9 L/h in patients with Cystic Fibrosis.


/Cysteamine is/ poorly bound to plasma proteins.

Thomson.Micromedex. Drug Information for the Health Care Professional. 25th ed. Volume 1. Plus Updates. Content Reviewed by the United States Pharmacopeial Convention, Inc. Greenwood Village, CO. 2005.


It is not known whether cysteamine is distributed into breast milk.

Thomson.Micromedex. Drug Information for the Health Care Professional. 25th ed. Volume 1. Plus Updates. Content Reviewed by the United States Pharmacopeial Convention, Inc. Greenwood Village, CO. 2005.


In a patient with cystinosis, an oral dose of cysteamine was absorbed rapidly, with plasma cysteamine reaching a maximum of 56 microM 1 h after the dose. By 1.8 h the plasma cysteamine concentration had decreased to one-half the maximum value.

PMID:3364734 Smolin LA et al; Anal Biochem.168 (2): 374-9 (1988)


Cysteamine bitartrate was administered to 11 cystinosis patients at their regular dose level in a single-dose, open-label, steady-state study. Blood samples were collected and analysed for plasma cysteamine and white blood cell cystine content and pharmacokinetic and pharmacodynamic parameters estimated by NONMEM analysis using a linked pharmacokinetic-pharmacodynamic model. Cysteamine was rapidly cleared from the plasma (mean CL/F = 32.3 mL min(-1) kg(-1), range = 17.3-52.2), appeared to be extensively distributed (mean Vss/F = 15.1 l, range 2.7-32.3) and exhibited a mean Tmax of 1.4 hr. White blood cell cystine content post-dosing was significantly decreased compared with pre- and post-dose values (average decrement approximately 47%). A counter-clockwise hysteresis was noted in all patients, suggestive of a lag time (mean Tlag = 0.44 hr, range 0.22-0.92) between drug concentration and effect. The results of this study establish that cysteamine is rapidly cleared from the plasma but that an every 6 hr dosing interval adequately maintains white blood cell cystine content below the target of 1 nmol cystine per mg protein. /Cysteamine bitartrate/

PMID:14651726 Full text: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1884393 Belldina EB et al; Br J Clin Pharmacol 56 (5): 520-5 (2003)


Cysteamine (beta-mercaptoethylamine, MEA) is currently used to treat children with nephropathic cystinosis. In this study MEA was compared to phosphocysteamine (MEAP), a phosphorothioester that tastes and smells better than MEA, with respect to its ability to elevate plasma MEA and deplete leukocytes of cystine. Studies were performed in six children with nephropathic cystinosis ranging in age from 2 to 10 yr. After equimolar oral doses of either MEA or MEAP plasma cysteamine was determined at various times for 6 h. MEA was determined by sodium borohydride reduction followed by high-performance liquid chromatography separation and electrochemical detection. Leukocyte cystine was measured before and 1 and 6 h after drug administration. Peak plasma MEA was obtained 30 min to 1 hr after a dose and was not significantly different when MEA (48.6 +/- 10.7, mean +/- SD) or MEAP (54.1 +/- 20.2) was given. Significant plasma MEA concentrations were seen as early as 15 min after an oral dose, indicating rapid absorption. Analysis of vomitus indicated that hydrolysis of the phosphate group of MEAP occurs in the stomach. The percent decrease in leukocyte cystine content obtained with MEA administration (61.9%) was not significantly different from the decrease observed when MEAP was administered (65.3%). MEA and MEAP appear to be equally effective in their cystine-depleting properties.

PMID:3393396 Smolin LA et al; Pediatr Res 23 (6): 616-20 (1988)


5.6 Metabolism/Metabolites

There is limited information in the literature regarding the metabolism of cysteamine. This drug undergoes significant first-pass metabolism.


5.7 Biological Half-Life

The half-life of cysteamine is about 3.7 hours.


5.8 Mechanism of Action

Individuals born without the ability to metabolize cystine suffer from cystinosis, a rare genetic disorder characterized by the widespread accumulation of cystine crystals throughout the body and eye tissues. The cystine crystals may cause considerable damage, particularly in the renal tissues and corneal tissues. In some cases, renal failure can occur during childhood if the condition is left untreated. Other organs that may be affected by cystinosis include the CNS, thyroid, pancreas, muscle tissues, and gonads. Cysteamine converts cystine to cysteine and cysteine-cysteamine mixed disulfides, reducing the buildup of corneal cystine crystals. This drug participates in a thiol-disulfide interchange reaction with lysosomes, leading to cysteine exit from the lysosome in patients diagnosed with cystinosis.


Cysteamine is an aminothiol that converts cystine to cysteine and cysteine-cysteamine mixed disulfide, both of which can pass through the lysosomal membrane of patients with cystinosis. In the nephropathic form of cystinosis, the accumulation of cystine and the formation of crystals damage various organs, especially the kidney. Cysteamine improves glomerular function without affecting tubular function.

Thomson.Micromedex. Drug Information for the Health Care Professional. 25th ed. Volume 1. Plus Updates. Content Reviewed by the United States Pharmacopeial Convention, Inc. Greenwood Village, CO. 2005.


Post Enquiry
POST ENQUIRY