Language selection

Search

Patent 2500836 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2500836
(54) English Title: METAL-CONTAINING MATERIALS, COMPOSITIONS AND METHODS
(54) French Title: MATERIAUX ET COMPOSITIONS METALLIFERES, ET PROCEDES ASSOCIES
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 33/38 (2006.01)
  • A61K 9/00 (2006.01)
  • A61K 9/12 (2006.01)
  • A61K 9/14 (2006.01)
  • A61K 9/51 (2006.01)
  • A61K 9/70 (2006.01)
  • A61K 31/28 (2006.01)
  • A61K 47/36 (2006.01)
  • A61K 47/38 (2006.01)
(72) Inventors :
  • BURRELL, ROBERT E. (Canada)
  • WRIGHT, JOHN B. (United States of America)
  • LAM, KAN (United States of America)
  • YIN, HUA QING (Canada)
  • NAYLOR, ANTONY G. (Canada)
  • MOXHAM, PETER H. (Canada)
  • GILLIS, SCOTT H. (United States of America)
  • SCHECHTER, PAUL (United States of America)
  • STILES, JAMES ALEXANDER ROBERT (Canada)
(73) Owners :
  • SMITH & NEPHEW (OVERSEAS)LIMITED (United Kingdom)
(71) Applicants :
  • NUCRYST PHARMACEUTICALS CORP. (Canada)
(74) Agent: MCKAY-CAREY & COMPANY
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2003-10-22
(87) Open to Public Inspection: 2004-05-06
Examination requested: 2008-10-15
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2003/033446
(87) International Publication Number: WO2004/037187
(85) National Entry: 2005-03-31

(30) Application Priority Data:
Application No. Country/Territory Date
10/277,673 United States of America 2002-10-22
10/277,358 United States of America 2002-10-22
10/277,356 United States of America 2002-10-22
10/277,298 United States of America 2002-10-22
10/277,362 United States of America 2002-10-22
10/277,320 United States of America 2002-10-22
10/364,983 United States of America 2003-02-12

Abstracts

English Abstract




Metal-containing materials, compositions and methods are disclosed. The metal-
containing material can be, for example, an antimicrobial material, an anti-
biofilm material, an antibacterial material, an anti-inflammatory material, an
anti-fungal material, an anti-viral material, an anti-cancer material, a pro-
apoptosis material, anti-proliferative, MMP modulating material, an atomically
disordered, crystalline material, and/or a nanocrystalline material. In
certain embodiments, the metal-containing material is an atomically
disordered, nanocrystalline silver-containing material.


French Abstract

L'invention porte sur des matériaux et compositions métallifères, et les procédés associés. Un tel matériau peut par exemple être un antimicrobien, un anti-film bio, un antibactérien, un anti-inflammatoire, un antifongique, un antiviral, un anticancéreux, un pro-apoptose, un anti-prolifération, un modulateur de la MMP, un corps cristallin à désordre atomique, et/ou un corps nanocristallin. Dans certaines exécutions, le matériau métallifère est un corps nanocristallin argentifère à désordre atomique.

Claims

Note: Claims are shown in the official language in which they were submitted.



WHAT IS CLAIMED IS:

1. A pharmaceutical composition, comprising:
a pharmaceutically acceptable carrier; and
a silver-containing material in the pharmaceutically acceptable carrier,
wherein the pharmaceutical composition comprises from about 0.001 weight
percent to about 50 weight percent of the silver-containing material.

2. The pharmaceutical composition of claim 1, wherein the pharmaceutical
composition comprises at least about 0.1 weight percent of the silver-
containing material.

3. The pharmaceutical composition of claim 1, wherein the pharmaceutical
composition comprises at least about 0.5 weight percent of the silver-
containing material.

4. The pharmaceutical composition of claim 1, wherein the pharmaceutical
composition comprises less than about 40 weight percent of the silver-
containing
material.

5. The pharmaceutical composition of claim 1, wherein the pharmaceutical
composition comprises less than about 30 weight percent of the silver-
containing
material.

6. The pharmaceutical composition of claim 1, wherein the silver-containing
material is selected from the group consisting of colloidal silver, silver
nitrate and silver
sulfadiazine, silver carbonate, silver acetate, silver lactate, silver
citrate, silver oxide,
silver hydroxide, silver succinate, silver chlorate, alkali silver
thiosulphates, silver
myristate, silver sorbate, silver stearate, silver oleate, silver glutonate,
silver adipate,
atomically disordered silver, nanocrystalline silver and combinations thereof.

7. A method of treating a subject having a condition, comprising:
contacting an area of the subject having the condition with a pharmaceutical
composition, the pharmaceutical composition comprising:

97



a pharmaceutically acceptable carrier; and
a silver-containing material in the pharmaceutically acceptable carrier,
wherein the pharmaceutical composition comprises from about 0.001 weight
percent to about 50 weight percent of the silver-containing material.

8. The method of claim 7, wherein the pharmaceutical composition
comprises at least about 0.1 weight percent of the silver-containing material.

9. The method of claim 7, wherein the pharmaceutical composition
comprises at least about 0.5 weight percent of the silver-containing material.

10. The method of claim 7, wherein the pharmaceutical composition
comprises less than about 40 weight percent of the silver-containing material.

11. The method of claim 7, wherein the pharmaceutical composition
comprises less than about 30 weight percent of the silver-containing material.

12. The method of claim 7, wherein the silver-containing material is selected
from the group consisting of colloidal silver, silver nitrate and silver
sulfadiazine, silver
carbonate, silver acetate, silver lactate, silver citrate, silver oxide,
silver hydroxide, silver
succinate, silver chlorate, alkali silver thiosulphates, silver myristate,
silver sorbate, silver
stearate, silver oleate, silver glutonate, silver adipate, atomically
disordered silver,
nanocrystalline silver and combinations thereof.

13. The method of claim 7, bacterial conditions, biofilm conditions, microbial
conditions, inflammatory conditions, fungal conditions, viral conditions,
autoimmune
conditions, idiopathic conditions, hyperproliferative conditions, noncancerous
growths
and cancerous conditions.

14. The method of claim 7, wherein the condition comprises a skin condition.

98



15. A method of treating a subject having a respiratory condition, comprising:
contacting an area of the subject having the respiratory condition with a
silver-
containing material.

16. The method of claim 15, wherein the respiratory condition is selected from
the group consisting of bacterial conditions, biofilm conditions, microbial
conditions,
inflammatory conditions, fungal conditions, viral conditions, autoimmune
conditions,
idiopathic conditions, noncancerous growths, hyperproliferative conditions,
cancerous
conditions and combinations thereof.

17. The method of claim 15, wherein the condition is selected from the group
consisting of asthma, emphysema, bronchitis, pulmonary edema, acute
respiratory
distress syndrome, bronchopulmonary dysplasia, fibrotic conditions, pulmonary
atelectasis, tuberculosis, pneumonia, sinusitis, allergic rhinitis,
pharyngitis, mucositis,
stomatitis, chronic obstructive pulmonary disease, bronchiectasis, cystic
fibrosis and
combinations thereof.

18. The method of claim 15, wherein the silver-containing material is selected
from the group consisting of colloidal silver, silver nitrate and silver
sulfadiazine, silver
carbonate, silver acetate, silver lactate, silver citrate, silver oxide,
silver hydroxide, silver
succinate, silver chlorate, alkali silver thiosulphates, silver myristate,
silver sorbate, silver
stearate, silver oleate, silver glutonate, silver adipate, atomically
disordered silver,
nanocrystalline silver and combinations thereof.

19. The method of claim 15, wherein, when contacted with the area of the
subject having the respiratory condition, the silver-containing material is in
a solution, in
an aerosol, in a pharmaceutically acceptable carrier, or in the form of a free-
standing
powder.

99



20. The method of claim 15, wherein the area of the subject is selected from
the group consisting of the subject's oral cavity, the subject's nasal cavity,
the subject's
lungs and combinations thereof.

21. The method of claim 15, wherein, when contacted with the area of the
subject having the respiratory condition, the silver-containing material is in
a solution,
and the solution contains at most about 0.5 weight percent of the silver-
containing
material.

22. The method of claim 15, wherein, when contacted with the area of the
subject having the respiratory condition, the silver-containing material is
the form of a
dry powder aerosol, and the dry powder aerosol contains at most about 99
weight percent
of the silver-containing material.

23. The method of claim 22, wherein the dry powder aerosol contains at least
about 10 weight percent of the silver-containing material.

24. A method of treating a subject having a condition, comprising:
contacting an area of the subject having the condition with a silver-
containing
material by injecting a free-standing powder of the silver-containing material
into the
subject, or by inhaling a free-standing powder of the silver-containing
material,
wherein the silver-containing material is selected from the group consisting
of
colloidal silver, silver nitrate and silver sulfadiazine, silver carbonate,
silver acetate,
silver lactate, silver citrate, silver oxide, silver hydroxide, silver
succinate, silver chlorate,
alkali silver thiosulphates, silver myristate, silver sorbate, silver
stearate, silver oleate,
silver glutonate, silver adipate, atomically disordered silver,
nanocrystalline silver and
combinations thereof.

25. The method of claim 24, wherein the condition is selected from the group
consisting of wherein the condition is selected from the group consisting of
bacterial
conditions, biofilm conditions, microbial conditions, inflammatory conditions,
fungal

100



conditions, viral conditions, autoimmune conditions, idiopathic conditions,
hyperproliferative conditions, noncancerous growths, cancerous conditions and
combinations thereof.

26. The method of claim 24, wherein the condition is selected from skin
conditions, integument conditions, respiratory conditions, musculo-skeletal
conditions,
circulatory conditions, mucosal conditions, serosal conditions and
combinations thereof.

27. The method of claim 24, wherein the free-standing powder has an average
particle size of about 10 microns or less.

28. A free-standing powder of a silver-containing material,
wherein the silver-containing material is selected from the group consisting
of
colloidal silver, silver nitrate and silver sulfadiazine, silver carbonate,
silver acetate,
silver lactate, silver citrate, silver oxide, silver hydroxide, silver
succinate, silver chlorate,
alkali silver thiosulphates, silver myristate, silver sorbate, silver
stearate, silver oleate,
silver glutonate, silver adipate, atomically disordered silver,
nanocrystalline silver and
combinations thereof.

29. The method of claim 28, wherein the free-standing powder has an average
particle size of about 10 microns or less.

30. An aerosol comprising a silver-containing material, wherein the silver-
containing material is selected from the group consisting of colloidal silver,
silver nitrate
and silver sulfadiazine, silver carbonate, silver acetate, silver lactate,
silver citrate, silver
oxide, silver hydroxide, silver succinate, silver chlorate, alkali silver
thiosulphates, silver
myristate, silver sorbate, silver stearate, silver oleate, silver glutonate,
silver adipate,
atomically disordered silver, nanocrystalline silver and combinations thereof.

31. The aerosol of claim 30, wherein the aerosol further comprises a solvent
for the silver-containing material.

101



32. The aerosol of claim 31, wherein the aerosol comprises at most about 99
weight percent of the silver-containing material.

33. The aerosol of claim 30, wherein the aerosol comprises at most about 99
weight percent of the silver-containing material.

34. A method of treating a subject having a condition, comprising:
contacting an area of the subject having the condition with a material that
comprises:
a metal; and
an element selected from the group consisting of oxygen, nitrogen,
carbon, boron, sulfur, a halogen, phosphorus, silicon, hydrogen and
combinations thereof,
wherein the nanocrystalline material contains at least about one atomic
percent of the element.

35. The method of claim 34, wherein the condition is a respiratory condition.

36. The method of claim 35, wherein the condition is selected from the group
consisting of bacterial conditions, biofilm conditions, microbial conditions,
inflammatory
conditions, fungal conditions, viral conditions, autoimmune conditions,
idiopathic
conditions, hyperproliferative conditions, noncancerous growths, cancerous
conditions
and combinations there.

37. The method of claim 35, wherein the condition is selected from the group
consisting of asthma, emphysema, bronchitis, pulmonary edema, acute
respiratory
distress syndrome, bronchopulmonary dysplasia, fibrotic conditions, pulmonary
atelectasis, tuberculosis, pneumonia, sinusitis, allergic rhinitis,
pharyngitis, mucositis,
stomatitis, chronic obstructive pulmonary disease, bronchiectasis, cystic
fibrosis and
combinations thereof.

102



38. The method of claim 34, wherein the condition comprises a musculo-
skeletal condition.

39. The method of claim 38, wherein the condition is selected from the group
consisting of bacterial conditions, microbial conditions, inflammatory
conditions, fungal
conditions, viral conditions, autoimmune conditions, idiopathic conditions,
hyperproliferative conditions, noncancerous growths, cancerous conditions and
combinations thereof.

40. The method of claim 38, wherein the musculo-skeletal condition is
selected from the group consisting of tendonitis, osteomyelitis, fibromyalgia,
bursitis,
arthritis and combinations thereof.

41. The method of claim 34, wherein the condition comprises a circulatory
condition.

42. The method of claim 41, wherein the condition is selected from the group
consisting of bacterial conditions, microbial conditions, inflammatory
conditions, fungal
conditions, viral conditions, autoimmune conditions, idiopathic conditions,
hyperproliferative conditions, noncancerous growths, cancerous conditions and
combinations thereof.

43. The method of claim 41, wherein the circulatory condition is selected from
the group consisting of arteriosclerosis, lymphoma, septicemia, leukemia,
ischemic
vascular disease, lymphangitis, atherosclerosis and combinations thereof.

44. The method of claim 34, wherein the condition is selected from mucosal
conditions, serosal conditions and combinations thereof.

45. The method of claim 44, wherein the condition is selected from the group
consisting of bacterial conditions, microbial conditions, inflammatory
conditions, fungal

103



conditions, viral conditions, autoimmune conditions, idiopathic conditions,
hyperproliferative conditions, noncancerous growths, cancerous conditions and
combinations thereof.
46. The method of claim 44, wherein the condition is selected from the group
consisting of pericarditis, Bowen's disease, stomatitis, prostatitis,
sinusitis, allergic
rhinitis, digestive disorders, peptic ulcers, esophageal ulcers, gastric
ulcers, duodenal
ulcer, espohagitis, gastritis, enteritis, enterogastric intestinal hemorrhage,
toxic epidermal
necrolysis syndrome, Stevens Johnson syndrome, fibrotic conditions,
bronchitis,
pneumonia, pharyngitis, common cold, ear infections, sore throat, sexually
transmitted
diseases, inflammatory bowel disease, colitis, hemorrhoids, thrush, dental
conditions, oral
conditions, conjunctivitis, periodontal conditions and combinations thereof.
47. The method of claim 34, wherein the condition comprises cancer.
48. The method of claim 47, wherein the cancer is selected from the group
consisting of tumors, hematologic malignancies and combinations thereof.
49. The method of claim 34, wherein the area of the subject is selected from
the group consisting of a hyperplastic tissue, a tumor tissue, a cancerous
lesion and
combinations thereof.
50. The method of claim 49, wherein the method induces apoptosis at the area
of the subject.
51. The method of claim 49, wherein the method modulates matrix
metalloproteinases at the area of the subject.
52. The method of claim 34, wherein the condition is selected from skin
conditions, integument conditions and combinations thereof.



104


53. The method of claim 52, wherein the condition is selected from the group
consisting of bacterial conditions, microbial conditions, inflammatory
conditions, fungal
conditions, viral conditions, autoimmune conditions, idiopathic conditions,
hyperproliferative conditions, noncancerous growths, cancerous conditions and
combinations thereof.
54. The method of claim 52, wherein the condition is selected from the group
consisting of a burn, eczema, erythroderma, an insect bite, mycosis fungoides,
pyoderma
gangrenosum, eythrema multiforme, rosacea, onychomyocosis, acne, psoriasis,
Reiter's
syndrome, pityriasis rubra pilaris, hyperpigmentation, vitiligo, hypertropic
scarring,
keloid, lichen plainus, age related skin disorders, hyperproliferative
variants of the
disorders of keratinization and combinations thereof.
55. A method of treating a subject having a condition, comprising:
contacting an area of the subject having the condition with a material that
comprises:
a metal; and
an element selected from the group consisting of oxygen, nitrogen,
carbon, boron, sulfur, a halogen, phosphorus, silicon, hydrogen and
combinations thereof,
wherein the atomically disordered, crystalline material contains at least
about one atomic percent of the element.
56. The method of claim 55, wherein the condition is a respiratory condition.
57. The method of claim 56, wherein the condition is selected from the group
consisting of bacterial conditions, biofilm conditions, microbial conditions,
inflammatory
conditions, fungal conditions, viral conditions, autoimmune conditions,
idiopathic
conditions, hyperproliferative conditions, noncancerous growths, cancerous
conditions
and combinations thereof.



105


58. The method of claim 56, wherein the condition is selected from the group
consisting of asthma, emphysema, bronchitis, pulmonary edema, acute
respiratory
distress syndrome, bronchopulmonary dysplasia, fibrotic conditions, pulmonary
atelectasis, tuberculosis, pneumonia, sinusitis, allergic rhinitis,
pharyngitis, mucositis,
stomatitis, chronic obstructive pulmonary disease, bronchiectasis, cystic
fibrosis and
combinations thereof.
59. The method of claim 55, wherein the condition comprises a musculo-
skeletal condition.
60. The method of claim 59, wherein the condition is selected from the group
consisting of bacterial conditions, microbial conditions, inflammatory
conditions, fungal
conditions, viral conditions, autoimmune conditions, idiopathic conditions,
hyperproliferative conditions, noncancerous growths, cancerous conditions and
combinations thereof.
61. The method of claim 59, wherein the musculo-skeletal condition is
selected from the group consisting of tendonitis, osteomyelitis, fibromyalgia,
bursitis,
arthritis and combinations thereof.
62. The method of claim 55, wherein the condition comprises a circulatory
condition.
63. The method of claim 62, wherein the condition is selected from the group
consisting of bacterial conditions, microbial conditions, inflammatory
conditions, fungal
conditions, viral conditions, autoimmune conditions, idiopathic conditions,
hyperproliferative conditions, noncancerous growths, cancerous conditions and
combinations thereof.



106


64. The method of claim 62, wherein the circulatory condition is selected from
the group consisting of arteriosclerosis, lymphoma, septicemia, leukemia,
ischemic
vascular disease, lymphangitis, atherosclerosis and combinations thereof.
65. The method of claim 55, wherein the condition is selected from mucosal
conditions, serosal conditions and combinations thereof.
66. The method of claim 65, wherein the condition is selected from the group
consisting of bacterial conditions, microbial conditions, inflammatory
conditions, fungal
conditions, viral conditions, autoimmune conditions, idiopathic conditions,
hyperproliferative conditions, noncancerous growths, cancerous conditions and
combinations thereof.
67. The method of claim 65, wherein the condition is selected from the group
consisting of pericarditis, Bowen's disease, stomatitis, prostatitis,
sinusitis, allergic
rhinitis, digestive disorders, peptic ulcers, esophageal ulcers, gastric
ulcers, duodenal
ulcer, espohagitis, gastritis, enteritis, enterogastric intestinal hemorrhage,
toxic epidermal
necrolysis syndrome, Stevens Johnson syndrome, fibrotic conditions,
bronchitis,
pneumonia, pharyngitis, common cold, ear infections, sore throat, sexually
transmitted
diseases, inflammatory bowel disease, colitis, hemorrhoids, thrush, dental
conditions, oral
conditions, conjunctivitis, periodontal conditions, combinations thereof and
combinations
thereof.

68. The method of claim 55, wherein the condition comprises cancer.

69. The method of claim 68, wherein the cancer is selected from the group
consisting of tumors, hematologic malignancies and combinations thereof.

70. The method of claim 55, wherein the area of the subject is selected from
the group consisting of a hyperplastic tissue, a tumor tissue, a cancerous
lesion and
combinations thereof.



107


71. The method of claim 70, wherein the method induces apoptosis at the area
of the subject.
72. The method of claim 70, wherein the method modulates matrix
metalloproteinases at the area of the subject.
73. A nanocrystalline material, comprising:
a metal; and
an element selected from the group consisting of oxygen, nitrogen, carbon,
boron, sulfur, phosphorus, silicon, a halogen, hydrogen and combinations
thereof,
wherein the nanocrystalline material contains at least about one atomic
percent of the element.
74. The nanocrystalline material of claim 73, wherein the nanocrystalline
material comprises at most about 90 weight percent of the element.
75. The nanocrystalline material of claim 73, wherein the nanocrystalline
material is in the form of a free-standing powder.
76. The nanocrystalline material of claim 73, wherein the nanocrystalline
material is contained within a composition that further comprises a
pharmaceutically
acceptable carrier.
77. The nanocrystalline material of claim 73, wherein the nanocrystalline
material is contained within a solution that further comprises a solvent.
78. The nanocrystalline material of claim 73, wherein the nanocrystalline
material is in the form of an aerosol.



108


79. The nanocrystalline material of claim 73, wherein the nanocrystalline
material is contained in a composition that further includes a hydrocolloid.
80. The nanocrystalline material of claim 73, wherein the nanocrystalline
material is contained in an article in the form of a tape, a pill, a capsule,
a tablet, a
lozenge or a suppository.
81. The nanocrystalline material of claim 73, wherein the nanocrystalline
material is in the form of an article including a substrate and a coating, and
the
nanocrystalline material is in the coating.
82. The nanocrystalline material of claim 73, wherein the nanocrystalline
material is in the form of an agglomerate of clusters of atoms.
83. The nanocrystalline material of claim 73, wherein the nanocrystalline
material comprises an atomically disordered, crystalline material.
84. The nanocrystalline material of claim 73, wherein the nanocrystalline
material comprises a material selected from the group consisting of
antimicrobial
materials, antibacterial materials, anti-inflammatory materials, antifungal
materials,
antiviral materials, anti-autoimmune materials, anti-cancer materials, pro-
apoptosis
materials, MMP modulating materials, anti-proliferative materials, and
combinations
thereof.
85. The nanocrystalline material of claim 73, wherein the metal comprises
silver.6
86. The nanocrystalline material of claim 73, wherein the nanocrystalline
material comprises at least two different metal elements.
87. An atomically disordered, crystalline material, comprising:



109


a metal; and
an element selected from the group consisting of oxygen, nitrogen, carbon,
boron, sulfur, a halogen, phosphorus, silicon, hydrogen and combinations
thereof,
wherein the atomically disordered, crystalline material contains at least
about one atomic percent of the element.
88. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material comprises at most about 90 weight
percent of
the element.
89. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material is in the form of a free-standing
powder.
90. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material is contained within a composition
that further
comprises a pharmaceutically acceptable carrier.
91. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material is contained within a solution
that further
comprises a solvent.
92. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material is in the form of an aerosol.
93. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material is contained in a composition that
further
includes a hydrocolloid.
94. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material is contained in an article in the
form of a tape,
a pill, a capsule, a tablet, a lozenge or a suppository.



110


95. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material is in the form of an article
including a
substrate and a coating, and the atomically disordered, crystalline material
is in the
coating.
96. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material is in the form of an agglomerate
of clusters of
atoms.
97. The atomically disordered, crystalline material of claim 87, wherein the
atomically disordered, crystalline material comprises a material selected from
the group
consisting of antimicrobial materials, antibacterial materials, anti-
inflammatory materials,.
antifungal materials, antiviral materials, anti-autoimmune materials, anti-
cancer
materials, pro-apoptosis materials, MMP modulating materials, anti-
proliferative, and
combinations thereof.
98. The atomically disordered, crystalline material of claim 87, wherein the
metal comprises silver.
99. The atomically disordered, crystalline material of claim 87, wherein the
nanocrystalline material comprises at least two different metal elements.



111

Description

Note: Descriptions are shown in the official language in which they were submitted.




CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Metal-Containing Materials, Compositions and Methods
TECHNICAL FIELD
The invention relates to metal-containing materials, compositions and methods.
BACKGROUND
It is generally desirable to treat a subject (e.g., a human) that has an
undesirable
condition. Many different compositions have been developed to treat
undesirable
conditions. For example, certain forms of silver have been reported to be
effective in
treating some undesirable skin conditions.
SUMMARY
The invention relates to metal-containing materials, compositions and methods.
In one aspect, the invention features a pharmaceutical composition that
includes a
pharmaceutically acceptable carrier and a silver-containing material in the
pharmaceutically acceptable carrier. The pharmaceutical composition includes
from
~5 about 0.001 weight percent to about 50 weight percent of the silver-
containing material.
In another aspect, the invention features a method of treating a subject
having a
condition. The method includes contacting an area of the subject having the
condition
with a pharmaceutical composition that includes a pharmaceutically acceptable
Garner
and a silver-containing material in the pharmaceutically acceptable Garner.
The
2o pharmaceutical composition includes from about 0.001 weight percent to
about SO weight
percent of the silver-containing material.
In a further aspect, the invention features a method of treating a subject
having a
respiratory condition that includes contacting an area of the subj ect having
the respiratory
condition with a silver-containing material.
25 In one aspect, the invention features a method of treating a subject having
a
condition. The method includes contacting an area of the subj ect having the
condition
with a silver-containing material by injecting a free-standing powder of the
silver-



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
containing material into the subject, or by inhaling a free-standing powder of
the silver-
containing material. The silver-containing material is colloidal silver,
silver nitrate and
silver sulfadiazine, silver carbonate, silver acetate, silver lactate, silver
citrate, silver
oxide, silver hydroxide, silver succinate, silver chlorate, alkali silver
thiosulphates, silver
s myristate, silver sorbate, silver stearate, silver oleate, silver glutonate,
silver adipate,
atomically disordered silver and/or nanocrystalline silver.
In another aspect, the invention features a free-standing powder of a silver-
containing material. The silver-containing material is colloidal silver,
silver nitrate and
silver sulfadiazine, silver carbonate, silver acetate, silver lactate, silver
citrate, silver
oxide, silver hydroxide, silver succinate, silver chlorate, alkali silver
thiosulphates, silver
myristate, silver sorbate, silver stearate, silver oleate, silver glutonate,
silver adipate,
atomically disordered silver and/or nanocrystalline silver.
In a further aspect, the invention features an aerosol that includes a silver
containing material. The silver-containing material is colloidal silver,
silver nitrate and
~ 5 silver sulfadiazine, silver carbonate, silver acetate, silver lactate,
silver citrate, silver
oxide, silver hydroxide, silver succinate, silver chlorate, alkali silver
thiosulphates, silver
myristate; silver sorbate, silver stearate, silver oleate, silver glutonate,
silver ad~pate,
atomically disordered silver and/or nanocrystalline silver.
In one aspect, the invention features a method of treating a subject having a
2o condition. The method includes contacting an area of the subject having the
condition
with a material. The material includes a metal and at least about one atomic
percent of a
different element. The different element can be, for example, oxygen,
nitrogen, carbon,
boron, sulfur, a halogen, phosphorus, silicon and/or hydrogen.
In another aspect, the invention features a nanocrystalline material that
includes a
25 metal and at least one atomic percent of a different element. The element
can be, for
example, oxygen, nitrogen, carbon, boron, sulfur, phosphorus, silicon, a
halogen and/or
hydrogen.
In a further aspect, the invention features a atomically disordered,
crystalline
material that includes a metal and at least one atomic percent of a different
element. The
so element can be, for example, oxygen, nitrogen, carbon, boron, sulfur,
phosphorus, silicon,
a halogen and/or hydrogen.



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Embodiments can include one or more of the following features.
In some embodiments, the material can include at most about 90 weight percent
of the element.
In certain embodiments, the material can in the form of a free-standing
powder.
In some embodiments, the material can be contained within a composition that
further includes a pharmaceutically acceptable carrier.
In certain embodiments, the material can be contained within a solution that
1 o further includes a solvent.
In some embodiments, the material can be in the form of an aerosol.
In certain embodiments, the material can be contained in a composition that
further includes a hydrocolloid.
In some embodiments, the material can be contained in an article in the form
of a
~ 5 tape, a pill, a capsule, a tablet, a lozenge or a suppository.
In certain embodiments, the material can be in the form of an article
including a
substrate and a coating, and the nanocrystalline material is in the coating.
In some embodiments, the material can be in the form of an agglomerate of
clusters of atoms.
2o In certain embodiments, the material can be an atomically disordered,
crystalline
material and/or a nanocrystalline material.
In some embodiments, the material can be an antimicrobial material, an
antibacterial material, an anti-inflammatory materials, an antifungal
material, an antiviral
material, an anti-autoimmune material, an anti-cancer material, a pro-
apoptosis material,
25 an MMP modulating material, and/or an anti-proliferative material.
In certain embodiments, the metal can be silver.
In some embodiments, the material can include at least two different metal
elements.
In some embodiments, the material can be, for example, atomically disordered,
so crystalline material and/or a nanocrystalline material.



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
In certain embodiments, the condition is a respiratory condition. The
respiratory
condition can be, for example, a bacterial condition, a biofilm condition, a
microbial
condition, an inflammatory condition, a fungal condition, a viral condition,
an
autoimmune condition, an idiopathic condition, a hyperproliferative condition,
a
noncancerous growth, and/or a cancerous condition. Examples of respiratory
conditions
include asthma, emphysema, bronchitis, pulmonary edema, acute respiratory
distress
syndrome, bronchopulmonary dysplasia, fibrotic conditions, pulmonary
atelectasis,
tuberculosis, pneumonia, sinusitis, allergic rhinitis, pharyngitis, mucositis,
stomatitis,
chronic obstructive pulmonary disease, bronchiectasis and cystic fibrosis.
In some embodiments, the condition includes a musculo-skeletal condition. The
musculo-skeletal condition can be, for example, a bacterial condition, a
biofilm
condition, a microbial condition, an inflammatory condition, a fungal
condition, a viral
condition, an autoimmune condition, an idiopathic condition, a
hyperproliferative
condition, a noncancerous growth, and/or a cancerous condition. Examples of
musculo-
~ 5 skeletal conditions include tendonitis, osteomyelitis, fibromyalgia,
bursitis and arthritis.
In certain embodiments, the condition is a circulatory condition. The
circulatory
condition can be, for example, a bacterial condition, a biofilm condition, a
microbial
condition, an inflammatory condition, a fungal condition, a viral condition,
an
autoimmune condition, an idiopathic condition, a hyperproliferative condition,
a
2o noncancerous growth, and/or a cancerous condition. Examples of circulatory
conditions
include arteriosclerosis, lymphoma, septicemia, leukemia, ischemic vascular
disease,
lymphangitis, atherosclerosis and combinations thereof.
In some embodiments, the condition is a mucosal condition and/or a serosal
condition. The mucosal and/or serosal condition can be, for example, a
bacterial
25 condition, a biofilin condition, a microbial condition, an inflammatory
condition, a fungal
condition, a viral condition, an autoimmune condition, an idiopathic
condition, a
hyperproliferative condition, a noncancerous growth, and/or a cancerous
condition.
Exemplary mucosal and/or serosal conditions include pericarditis, Bowen's
disease,
stomatitis, prostatitis, sinusitis, allergic rhinitis, digestive disorders,
peptic ulcers,
3o esophageal ulcers, gastric ulcers, duodenal ulcer, espohagitis, gastritis,
enteritis,
enterogastric intestinal hemorrhage, toxic epidermal necrolysis syndrome,
Stevens
4



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Johnson syndrome, fibrotic conditions, bronchitis, pneumonia, pharyngitis,
common cold,
ear infections, sore throat, sexually transmitted diseases, inflammatory bowel
disease,
colitis, hemorrhoids, thrush, dental conditions, oral conditions,
conjunctivitis, periodontal
conditions and combinations thereof.
In certain embodiments, the condition is cancer. The cancer can be, for
example,
in the form of tumors and/or hematologic malignancies.
In some embodiments, the area of the subject is a hyperplastic tissue, a tumor
tissue and/or a cancerous lesion. The method can, for example, induce
apoptosis at the
area of the subject. The method can, for example, modulate matrix
metalloproteinases at
the area of the subject.
In certain embodiments, the condition is a skin condition and/or an integument
condition. The skin and/or integument condition can be a bacterial condition,
a biofilm
condition, a microbial condition, an inflammatory condition, a fungal
condition, a viral'
condition, an autoimmune condition, an idiopathic condition, a
hyperproliferative
15 condition, a noncancerous growth, and/or a cancerous condition. Examples of
skin
and/or integument conditions include a burn, eczema, erythroderma, an insect
bite,
mycosis fungoides, pyoderma gangrenosum, eythrema multiforme, rosacea,
onychomyocosis, acne, psoriasis, Reiter's syndrome, pityriasis rubra pilaris,
hyperpigmentation, vitiligo, hypertropic scarring, keloid, lichen plainus, age
related skin
2o disorders, and hyperproliferative variants of the disorders of
keratinization.
In some embodiments, a pharmaceutical composition can include at least about
0.1 weight percent of the silver-containing material (e.g., at least about 0.5
weight percent
of the silver-containing material) andlor less than about 40 weight percent of
the silver-
containing material (e.g., less than about 30 weight percent of the silver-
containing
25 material).
Examples of silver-containing materials include colloidal silver, silver
nitrate and
silver sulfadiazine, silver carbonate, silver acetate, silver lactate, silver
citrate, silver
oxide, silver hydroxide, silver succinate, silver chlorate, alkali silver
thiosulphates, silver
myristate, silver sorbate, silver stearate, silver oleate, silver glutonate,
silver adipate,
3o atomically disordered silver and nanocrystalline silver.



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Examples of conditions include bacterial conditions, biofilm conditions,
microbial
conditions, inflammatory conditions, fungal conditions, viral conditions,
autoimmune '
conditions, idiopathic conditions, hyperproliferative conditions, noncancerous
growths
and cancerous conditions.
In some embodiments, when contacted with the area of the subject having the
respiratory condition, the silver-containing material is in a solution, in an
aerosol, in a
pharmaceutically acceptable carrier, or in the form of a free-standing powder.
In certain embodiments, the area of the subject having a respiratory condition
can
be the subject's oral cavity, the subject's nasal cavity and/or the subject's
lungs.
In some embodiments, when contacted with the area of the subject having the
respiratory condition, the silver-containing material is in a solution, and
the solution
contains at most about 0.5 weight percent of the silver-containing material.
In certain embodiments, when contacted with the area of the subject having the
respiratory condition, the silver-containing material is the form of a dry
powder aerosol,
~ 5 and the dry powder aerosol contains at most about 99 weight percent of the
silver-
containing material and/or at least about 10 weight percent of the silver-
containing
material.
In some embodiments, the condition is a skin condition, an integument
condition,
a respiratory condition, a musculo-skeletal condition, a circulatory
condition, a mucosal
2o condition and/or a serosal condition.
In certain embodiments, a free-standing powder has an average particle size of
about 10 microns or less.
In some embodiments, an aerosol can further include a solvent for the silver-
containing material. The aerosol can include at most about 99 weight percent
of the
25 silver-containing material.
Other features and advantages of the methods will be apparent from the
description and drawings, and from the claims.
DESCRIPTION OF DRAWINGS
Fig. 1 is a schematic view of a deposition system;
3o Fig. 2 is a graph showing the efficacy of different forms of silver on
erythema;



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
material is capable of releasing clusters of the metal (e.g., clusters of
Ag°, clusters of Ag+,
clusters containing both Ag+ and Ag°) that provide the observed
therapeutic properties.
In a further potential mechanism, it is believed that the concentration of
silver in a
solution can be raised above the saturation concentration of bare silver ions
(e.g., to
provide a relatively sustaining reservoir of silver as bare silver ions are
consumed). It is
believed that, as the bare silver ions are consumed, some of the other silver-
containing
species can decompose to create additional bare silver ions in accordance with
chemical
equilibria. It is also believed that the presence of silver in one or more
forms other than
bare silver ions may raise the level for the effective silver concentration
that is
nonharmful (e.g., non-toxic) to the cells of a subject (e.g., a human). In an
additional
potential mechanism, it is believed that one or more forms of silver complexes
may be
capable of penetrating cellular membranes (e.g., by mimicking species that are
normally
transported through the membranes), which may accelerate the permeation of
silver into
the cells. In general, it is believed that the form of the silver-containing
species contained
in an aqueous solution depends on the solution pH and/or the concentrations of
the
various silver-containing species in the solid form of the silver-containing
material. It is
believed that, in general, at low pH the dominant species is a bare silver
ion, but that at
higher pH, where the solubility of bare silver ions is believed to be limited
by the
solubility of silver hydroxide, other types of species including complexed
silver ions
2o and/or silver-containing clusters become increasingly stable provided that
the
concentration of bare silver ions remains at the saturation concentration . It
is also
believed that the nature and relative population of the silver-containing
species can
depend on the rate at which the species can dissolve from the solid silver-
bearing material
and the rate at which the species can react with one another in the solution.
It is believed
that combinations of potential mechanisms may result in the observed
therapeutic effect
of the metal-containing material.
In general, clusters refer to relatively small groups of atoms, ions or the
like. For
example, a cluster can contain at least two (e.g., at least three, at least
four, at least five,
at least six, at least seven, at least eight, at least nine, at least 10, at
least 11, at least 12, at
least 13, at least 14, at least 15, at least 20, at least 30, at least 40, at
least 50, at least 60,
at least 70, at least 80, at least 90) atoms, ions or the like, and/or at most
1,000 (e.g., at



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Fig. 3 is a graph showing the efficacy of different forms of silver on edema;
Fig. 4 is a graph showing MMP activity of incision fluids recovered from
incisions dressed with materials;
Fig. 5 is a graph showing total protease activity of incision fluids recovered
from
different dressings;
Fig. 6 is a graph showing the concentrations (ng/ml) of active MMP-9 in fluid
samples recovered from ulcers dressed with different materials;
Fig. 7 is a graph showing the concentrations (ng/ml) of active MMP-2 in fluid
samples recovered from ulcers dressed with different materials;
~ o Fig. 8 is a graph showing the concentrations (pg/ml) of active TNF-a in
fluid
samples recovered from ulcers dressed with different materials; and
Fig. 9 is a graph showing the concentrations (pg/ml) of active IL-1(3 in fluid
samples recovered from ulcers dressed with different materials.
DETAILED DESCRIPTION
~ 5 The inventors have discovered that certain metal-containing materials
(e.g.,
antimicrobial, atomically disordered, nanocrystalline silver-containing
materials) can be
used to treat a subj ect with a condition by contacting an area of the subj
ect having the
condition with the metal-containing material. As explained below, the metal-
containing
material can be in any of a variety of forms when delivered to a subject, and
the metal-
2o containing material can be delivered to a subject in a variety of ways. As
also explained
below, the metal-containing material can be used to treat various subjects,
conditions, and
condition locations.
Without wishing to be bound by theory, it is believed that the therapeutic
properties of the metal-containing materials may be explained by one or more
potential
2s mechanisms. W one potential mechanism (e.g., at relatively high pH), it is
believed that
the metal-containing material (e.g., antimicrobial, atomically disordered,
nanocrystalline
silver-containing materials) forms one or more metastable, relatively high
level metal
hydroxide species (e.g., Ag(OH)43-, Ag(OH)63-) that either directly or
indirectly (e.g., via
the formation of one or more biological mediators) provide the observed
therapeutic
3o properties. In another potential mechanism, it is believed that the metal-
containing



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
most 900, at most 800, at most 700, at most 600, at most 500, at most 400, at
most 300, at
most 200, at most 100) atoms, ions or the like. Clusters are described, for
example, in R.
P. Andres et al., "Research Opportunities on Cluster and Cluster-Assembled
Materials",
J. Mater. Res. Vol. 4, No 3, 1989, p. 704. In certain embodiments, a cluster
(e.g., a
cluster containing silver) can contain less than the 14 atoms and have a
normal face
centered cubic crystal lattice.
Materials
The metal-containing material can be an ionic material or a non-ionic
material.
The metal-containing material can be, for example, an atom, a molecule, or a
cluster.
In general, the metal-containing material is a metal or an alloy. Examples of
metal elements that can be contained in metal-containing materials include
Group I A
metal elements, Group II A metal elements, Group III A metal elements, Group
IV A
metal elements, Group V A metal elements, Group VI A metal elements, Group VII
A
~ 5 metal elements, Group VIII A metal elements, Group I B metal elements,
Group II B
metal elements, members of the lanthanide metal element series, and members of
the
actinide metal element series. In certain embodiments, metal-containing
materials
contain silver, gold, platinum, palladium, iridium, zinc, copper, tin,
antimony, and/or
bismuth. In some embodiments, a metal-containing material can include one or
more
2o transition metal elements (e.g., scandium, titanium, vanadium, chromium,
manganese,
iron, cobalt, nickel, copper and/or zinc). As an example, a metal-containing
material can
contain silver and platinum.
Examples of silver-containing materials include colloidal silver, silver
nitrate and
silver sulfadiazine, silver carbonate, silver acetate, silver lactate, silver
citrate, silver
25 oxide, silver hydroxide, silver succinate, silver chlorate, silver
stearate, silver sorbate,
silver oleate, silver glutonate, silver adipate, silver myristate, and alkali
silver
thiosulphate (e.g., sodium silver thiosulphate, potassium silver
thiosulphate).
In addition to one or more metal elements, a metal-containing material can
contain, for example, oxygen, nitrogen, carbon, boron, sulfur, phosphorus,
silicon, a
3o halogen (e.g., fluorine, chlorine, bromine, iodine) and/or hydrogen.
Examples of such
metal-containing materials include metal oxides, metal hydroxides, metal
nitrides, metal



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
carbides, metal phosphides, metal silicates, metal borides, metal sulfides,
metal halides
(e.g., metal fluorides, metal chlorides, metal bromides, metal iodides), metal
myristates,
metal sorbates, metal stearates, metal oleates, metal glutonates, metal
adipates, metal
silicates, metal phosphides, metal hydrides, metal nitrates, metal carbonates,
metal
sulfadiazines, metal hydrides, metal acetates, metal lactates, metal citrates,
alkali metal
thiosulphates (e.g., sodium metal thiosulphate, potassium metal thiosulphate).
In certain
embodiments, a metal-containing material contains at least about one atomic
percent
(e.g., at least about three atomic percent, at least about five atomic
percent, at least about
atomic percent, at least about 20 atomic percent, at least about 30 atomic
percent, at
least about 40 atomic percent, at least about 50 atomic percent) and/or at
most about 90
atomic percent (e.g., at most about ~0 atomic percent, at most about 70 atomic
percent, at
most about 60 atomic percent, at most about 50 atomic percent, at most about
40 atomic
percent, at most about 30 atomic percent, at most about 20 atomic percent, at
most about
atomic percent, at most about 12 atomic percent, at most about 10 atomic
percent) of
15 nonmetallic elements. For example, in some embodiments, a silver-
containing material
can contain oxygen in an amount from about five atomic percent to about 20
atomic
percent (e.g., from about five atomic percent to about 15 atomic percent, from
about eight
atomic percent to about 12 atomic percent).
In certain embodiments, the metal-containing materials are an antimicrobial
2o material, an anti-biofilm, an antibacterial material, an anti-inflammatory
material, an
antifungal material, an antiviral material, an anti-autoimmune material, an
anti-cancer
material, a pro-apoptosis material, an anti-proliferative material, an MMP
modulating
material, an atomically disordered crystalline material, and/or a
nanocrystalline material.
As used herein, an antimicrobial material herein refers to a material that has
2s sufficient antimicrobial activity to have a beneficial therapeutic effect.
In certain
embodiments, an antimicrobial material has a corrected zone of inhibition
("CZOI") of at
least about two millimeters (e.g., at least about three millimeters, at least
about four
millimeters, at lest about five millimeters, at least about six millimeters,
at least about
seven millimeters, at least about eight millimeters, at least about nine
millimeters, at least
3o about 10 millimeters). The CZOI of a material is determined as follows. The
material is
formed as a coating on a dressing (see discussion below). Basal medium Eagle
(BME)
to



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
with Earle's salts and L-glutamine is modified with calf/serum (10%) and 1.5%
agar prior
to being dispensed (15 ml) into Petri dishes. The agar containing Petri dishes
are allowed
to surface dry prior to being inoculated with a lawn of Staphylococcus aureus
ATCC
#25923. The inoculant is prepared from Bactrol Discs (Difco, M.) which are
reconstituted as per the manufacturer's directions. Immediately after
inoculation, the
coatings to be tested are placed on the surface of the agar. The dishes are
incubated for
24 hours at 37°C. After this incubation period, the zone of inhibition
("ZOI") is
measured and the CZOI is calculated as the ZOI minus the diameter of the test
material in
contact with the agar. It is to be noted that, while this test for
antimicrobial properties is
performed on materials that are in the form of a coating on a substrate (e.g.,
in the form
of a dressing), antimicrobial materials are not limited to materials that are
coated on a
substrate. Rather, a material in any form may be antimicrobial, but it is in
the form of a
coating on a substrate (e.g., in the form of a dressing) when its
antimicrobial properties
are tested according to the procedure described herein.
1s As referred to herein, an atomically disordered, crystalline material
(e.g., an
atomically disordered, nanocrystalline material) means a material that has
more long
range ordered, crystalline structure (a lesser degree of defects) than the
material has in a
fully amorphous state, but that also has less long range, ordered crystalline
structure (a
higher degree of defects) than the material has in a bulk crystalline state,
such as in the
2o form of a cast, wrought or plated material. Examples of defects include
point defects,
vacancies, line defects, grain boundaries, subgrain boundaries and amorphous
regions.
Point defects are defects on a size scale of no more than about four atomic
spacings. A
vacancy is the omission of an atom from its regular atomic site in the crystal
lattice. Line
defects are defective regions (e.g., edge dislocations, screw dislocations)
that result in
2s lattice distortions along a line (which may or may not be a straight line),
and generally
have a longer scale than point defects. In an edge dislocation, a lattice
displacement is
produced by a plane of atoms that forms a terminus of the lattice. In a screw
dislocation,
part of the lattice is displaced with respect to an adjacent part of the
lattice. Grain
boundaries separate regions having different crystallographic orientation or
3o misorientation (e.g., high angle grain boundaries, low angle grain
boundaries, including
tilt boundaries and twist boundaries). Subgrain boundaries refer to low angle
grain
11



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
boundaries. An amorphous region is a region that does not exhibit long range,
ordered
crystalline structure. In certain embodiments, an atomically disordered,
crystalline
material (e.g., an atomically disordered, nanocrystalline material) has a
degree of atomic
disorder that is about the same as the degree of atomic disorder of the
nanocrystalline
silver coating of a member of the Acticoat~' family of dressings (Smith &
Nephew, Hull,
UK) (e.g., an Acticoat~ dressing, an Acticoat7~ dressing, an Acticoat~
moisture coating
dressing, an Acticoat~ absorbent dressings). In some embodiments, an
atomically
disordered, crystalline material (e.g., an atomically disordered,
nanocrystalline material)
has a degree of atomic disorder that is about the same as the degree of atomic
disorder of
the nanocrystalline silver coatings having a CZOI of at least five millimeters
that are
disclosed in the examples of Burrell et al., U.S. Patent No. 5,958,440. In
certain
embodiments, an atomically disordered, crystalline material (e.g., an
atomically
disordered, nanocrystalline material), when contacted with an alcohol or water-
based
electrolyte, is released into the alcohol or water-based electrolyte (e.g., as
ions, atoms,
~ 5 molecules and/or clusters) over a time scale of at least about one hour
(e.g., at least about
two hours, at least about 10 hours, at least about a day). Examples of
alcohols and/or
water-based electrolytes include body fluids (e.g., blood, urine, saliva) and
body tissue
(e.g., skin, muscle, bone).
As referred to herein, a nanocrystalline material is a single-phase
polycrystal or a
2o multi-phase polycrystal having a maximum dimension of about 100 nanometers
or less
(e.g., about 90 nanometers or less, about 80 nanometers or less, about 70
nanometers or
less, about 60 manometers or less, about 50 manometers or less, about 40
manometers or
less, about 30 manometers or less, about 25 manometers or less) in at least
one dimension.
Examples of antimicrobial metal-containing materials (which may or may not
also
25 be an atomically disordered crystalline material or a nanocrystalline
material) include
antimicrobial silver-containing materials (e.g., antimicrobial silver,
antimicrobial silver
alloys, antimicrobial silver oxides, antimicrobial silver carbides,
antimicrobial silver
nitrides, antimicrobial silver borides, antimicrobial silver sulfides,
antimicrobial silver
myristates, antimicrobial silver stearates, antimicrobial silver oleates,
antimicrobial silver
3o glutonates, antimicrobial silver adipates, antimicrobial silver silicates,
antimicrobial
silver phosphides, antimicrobial silver halides, antimicrobial silver
hydrides,
12



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
antimicrobial silver nitrates, antimicrobial silver carbonates, antimicrobial
silver
sulfadiazines, antimicrobial silver acetates, antimicrobial silver lactates,
antimicrobial
silver citrates, antimicrobial alkali silver thiosulphates (e.g.,
antimicrobial sodium silver
thiosulphate, antimicrobial potassium silver thiosulphate)), antimicrobial
gold-containing
materials (e.g., antimicrobial gold, antimicrobial gold alloys, antimicrobial
gold oxides,
antimicrobial gold carbides, antimicrobial gold nitrides, antimicrobial gold
borides,
antimicrobial gold sulfides, antimicrobial gold myristates, antimicrobial gold
stearates,
antimicrobial gold oleates, antimicrobial gold glutonates, antimicrobial gold
glutonates,
antimicrobial gold adipates, antimicrobial gold silicates, antimicrobial gold
phosphides,
antimicrobial gold halides, antimicrobial gold hydrides, antimicrobial gold
nitrates,
antimicrobial gold carbonates, antimicrobial gold sulfadiazines, antimicrobial
gold
acetates, antimicrobial gold lactates, antimicrobial gold citrates,
antimicrobial alkali gold
thiosulphates (e.g., antimicrobial sodium gold thiosulphate, antimicrobial
potassium gold
thiosulphate)), antimicrobial platinum-containing materials (e.g.,
antimicrobial platinum,
~ 5 antimicrobial platinum alloys, antimicrobial platinum oxides,
antimicrobial platinum
carbides, antimicrobial platinum nitrides, antimicrobial platinum borides,
antimicrobial
platinum sulfides, antimicrobial platinum myristates, antimicrobial platinum
stearates,
antimicrobial platinum oleates, antimicrobial platinum glutonates,
antimicrobial platinum
glutonates, antimicrobial platinum adipates, antimicrobial platinum silicates,
2o antimicrobial platinum phosphides, antimicrobial platinum halides,
antimicrobial
platinum hydrides, antimicrobial platinum nitrates, antimicrobial platinum
carbonates,
antimicrobial platinum sulfadiazines, antimicrobial platinum acetates,
antimicrobial
platinum lactates, antimicrobial platinum citrates, antimicrobial alkali
platinum
thiosulphates (e.g., antimicrobial sodium platinum thiosulphate, antimicrobial
potassium
25 platinum thiosulphate)), antimicrobial palladium-containing materials
(e.g., antimicrobial
palladium, antimicrobial palladium alloys, antimicrobial palladium oxides,
antimicrobial
palladium carbides, antimicrobial palladium nitrides, antimicrobial palladium
borides,
antimicrobial palladium sulfides, antimicrobial palladium myristates,
antimicrobial
palladium stearates, antimicrobial palladium oleates, antimicrobial palladium
glutonates,
3o antimicrobial palladium glutonates, antimicrobial palladium adipates,
antimicrobial
palladium silicates, antimicrobial palladium phosphides, antimicrobial
palladium halides,
13



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
antimicrobial palladium hydrides, antimicrobial palladium nitrates,
antimicrobial
palladium carbonates, antimicrobial palladium sulfadiazines, antimicrobial
palladium
acetates, antimicrobial palladium lactates, antimicrobial palladium citrates,
antimicrobial
alkali palladium thiosulphates (e.g., antimicrobial sodium palladium
thiosulphate,
antimicrobial potassium palladium thiosulphate)), antimicrobial iridium-
containing
materials (e.g., antimicrobial iridium, antimicrobial iridium alloys,
antimicrobial iridium
oxides, antimicrobial iridium carbides, antimicrobial iridium nitrides,
antimicrobial
iridium borides, antimicrobial iridium sulfides, antimicrobial iridium
myristates,
antimicrobial iridium stearates, antimicrobial iridium oleates, antimicrobial
iridium
glutonates, antimicrobial iridium glutonates, antimicrobial iridium adipates,
antimicrobial
iridium silicates, antimicrobial iridium phosphides, antimicrobial iridium
halides,
antimicrobial iridium hydrides, antimicrobial iridium nitrates, antimicrobial
iridium
carbonates, antimicrobial iridium sulfides, antimicrobial iridium
sulfadiazines,
antimicrobial iridium acetates, antimicrobial iridium lactates, antimicrobial
iridium
~5 citrates, antimicrobial alkali iridium thiosulphates (e.g., antimicrobial
sodium iridium
thiosulphate, antimicrobial potassium iridium thiosulphate)), antimicrobial
zinc-
containing materials (e.g., antimicrobial zinc, antimicrobial zinc alloys,
antimicrobial
zinc oxides, antimicrobial zinc carbides, antimicrobial zinc nitrides,
antimicrobial zinc
borides, antimicrobial zinc sulfides, antimicrobial zinc myristates,
antimicrobial zinc
2o stearates, antimicrobial zinc oleates, antimicrobial zinc glutonates,
antimicrobial zinc
glutonates, antimicrobial zinc adipates, antimicrobial zinc silicates,
antimicrobial zinc
phosphides, antimicrobial zinc halides, antimicrobial zinc hydrides,
antimicrobial zinc
nitrates, antimicrobial zinc carbonates, antimicrobial zinc sulfides,
antimicrobial zinc
sulfadiazines, antimicrobial zinc acetates, antimicrobial zinc lactates,
antimicrobial zinc
25 citrates, antimicrobial alkali zinc thiosulphates (e.g., antimicrobial
sodium zinc
thiosulphate, antimicrobial potassium zinc thiosulphate)), antimicrobial
copper -
containing materials (e.g., antimicrobial copper, antimicrobial copper alloys,
antimicrobial copper oxides, antimicrobial copper carbides, antimicrobial
copper nitrides,
antimicrobial copper borides, antimicrobial copper sulfides, antimicrobial
copper
3o myristates, antimicrobial copper stearates, antimicrobial copper oleates,
antimicrobial
copper glutonates, antimicrobial copper glutonates, antimicrobial copper
adipates,
14



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
antimicrobial copper silicates, antimicrobial copper phosphides, antimicrobial
copper
halides, antimicrobial copper hydrides, antimicrobial copper nitrates,
antimicrobial
copper carbonates, antimicrobial copper sulfides, antimicrobial copper
sulfadiazines,
antimicrobial copper acetates, antimicrobial copper lactates, antimicrobial
copper citrates,
antimicrobial alkali copper thiosulphates (e.g., antimicrobial sodium copper
thiosulphate,
antimicrobial potassium copper thiosulphate)), antimicrobial tin-containing
materials
(e.g., antimicrobial tin, antimicrobial tin alloys, antimicrobial tin oxides,
antimicrobial tin
carbides, antimicrobial tin nitrides, antimicrobial tin borides, antimicrobial
tin sulfides,
antimicrobial tin myristates, antimicrobial tin stearates, antimicrobial tin
oleates,
antimicrobial tin glutonates, antimicrobial tin glutonates, antimicrobial tin
adipates,
antimicrobial tin silicates, antimicrobial tin phosphides, antimicrobial tin
halides,
antimicrobial tin hydrides, antimicrobial tin nitrates, antimicrobial tin
carbonates,
antimicrobial tin sulfides, antimicrobial tin sulfadiazines, antimicrobial tin
acetates,
antimicrobial tin lactates, antimicrobial tin citrates, antimicrobial alkali
tin thiosulphates
(e.g., antimicrobial sodium tin thiosulphate, antimicrobial potassium tin
thiosulphate)),
antimicrobial antimony-containing materials (e.g., antimicrobial antimony,
antimicrobial
antimony alloys, antimicrobial antimony oxides, antimicrobial antimony
carbides,
antimicrobial antimony nitrides, antimicrobial antimony borides, antimicrobial
antimony
sulfides, antimicrobial antimony myristates, antimicrobial antimony stearates,
2o antimicrobial antimony oleates, antimicrobial antimony glutonates,
antimicrobial
antimony glutonates, antimicrobial antimony adipates, antimicrobial antimony
silicates,
antimicrobial antimony phosphides, antimicrobial antimony halides,
antimicrobial
antimony hydrides, antimicrobial antimony nitrates, antimicrobial antimony
carbonates,
antimicrobial antimony sulfides, antimicrobial antimony sulfadiazines,
antimicrobial
antimony acetates, antimicrobial antimony lactates, antimicrobial antimony
citrates,
antimicrobial alkali antimony thiosulphates (e.g., antimicrobial sodium
antimony
thiosulphate, antimicrobial potassium antimony thiosulphate)), antimicrobial
bismuth
containing materials (e.g., antimicrobial bismuth, antimicrobial bismuth
alloys,
antimicrobial bismuth oxides, antimicrobial bismuth carbides, antimicrobial
bismuth
3o nitrides, antimicrobial bismuth borides, antimicrobial bismuth sulfides,
antimicrobial
bismuth myristates, antimicrobial bismuth stearates, antimicrobial bismuth
oleates,



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
antimicrobial bismuth glutonates, antimicrobial bismuth glutonates,
antimicrobial
bismuth adipates, antimicrobial bismuth silicates, antimicrobial bismuth
phosphides,
antimicrobial bismuth halides, antimicrobial bismuth hydrides, antimicrobial
bismuth
nitrates, antimicrobial bismuth carbonates, antimicrobial bismuth sulfides,
antimicrobial
bismuth sulfadiazines, antimicrobial bismuth acetates, antimicrobial bismuth
lactates,
antimicrobial bismuth citrates, antimicrobial allcali bismuth thiosulphates
(e.g.,
antimicrobial sodium bismuth thiosulphate, antimicrobial potassium bismuth
thiosulphate)).
While the preceding paragraph lists certain metal-containing materials that
are
anti-microbial, similar metal-containing materials (oxides, carbides,
nitrides, borides,
sulfides, myristates, stearates, oleates, glutonates, adipates, silicates,
phosphides, halides,
hydrides, nitrates, hydroxides, carbonates, sulfides, sulfadiazines, acetates,
lactates,
citrates and/or alkali metal thiosulphates of silver, gold, palladium,
platinum, tin, iridium,
antimony, bismuth, copper, zinc) can be anti-biofilm materials, antibacterial
materials,
~ 5 anti-inflammatory materials, antifungal materials, antiviral materials,
anti-autoimmune
materials, anti-cancer materials, pro-apoptosis materials, anti-
proliferatives, and/or MMP
modulating materials.
Examples of nanocrystalline metal-containing materials (which may or may not
also be an antimicrobial material or an atomically disordered crystalline
material) include
2o nanocrystalline silver-containing materials (e.g., nayocrystalline silver,
nanocrystalline
silver alloys, nanocrystalline silver oxides, nanocrystalline silver carbides,
nanocrystalline silver nitrides, nanocrystalline silver borides,
nanocrystalline silver
sulfides, nanocrystalline silver halides, nanocrystalline silver myristates,
nanocrystalline
silver stearates, nanocrystalline silver oleates, nanocrystalline silver
glutonates,
25 nanocrystalline silver glutonates, nanocrystalline silver adipates,
nanocrystalline silver
silicates, nanocrystalline silver phosphides, nanocrystalline silver hydrides,
nanocrystalline silver nitrates, nanocrystalline silver carbonates,
nanocrystalline silver
sulfides, nanocrystalline silver sulfadiazines, nanocrystalline silver
acetates,
nanocrystalline silver lactates, nanocrystalline silver citrates,
nanocrystalline alkali silver
3o thiosulphates (e.g., nanocrystalline sodium silver thiosulphate,
nanocrystalline potassium
silver thiosulphate)), nanocrystalline gold-containing materials (e.g.,
nanocrystalline
16



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
gold, nanocrystalline gold alloys, nanocrystalline gold oxides,
nanocrystalline gold
carbides, nanocrystalline gold nitrides, nanocrystalline gold borides,
nanocrystalline gold
sulfides, nanocrystalline gold halides, nanocrystalline gold hydrides,
nanocrystalline gold
nitrates, nanocrystalline gold myristates, nanocrystalline gold stearates,
nanocrystalline
gold oleates, nanocrystalline gold glutonates, nanocrystalline gold
glutonates,
nanocrystalline gold adipates, nanocrystalline gold silicates, nanocrystalline
gold
phosphides, nanocrystalline gold carbonates, nanocrystalline gold sulfides,
nanocrystalline gold sulfadiazines, nanocrystalline gold acetates,
nanocrystalline gold
lactates, nanocrystalline gold citrates, nanocrystalline alkali gold
thiosulphates (e.g.,
1 o nanocrystalline sodium gold thiosulphate, nanocrystalline potassium gold
thiosulphate)),
nanocrystalline platinum-containing materials (e.g., nanocrystalline platinum,
nanocrystalline platinum alloys, nanocrystalline platinum oxides,
nanocrystalline
platinum carbides, nanocrystalline platinum nitrides, nanocrystalline platinum
borides,
nanocrystalline platinum sulfides, nanocrystalline platinum myristates,
nanocrystalline
platinum stearates, nanocrystalline platinum oleates, nanocrystalline platinum
glutonates,
nanocrystalline platinum glutonates, nanocrystalline platinum adipates,
nanocrystalline
platinum silicates, nanocrystalline platinum phosphides, nanocrystalline
platinum halides,
nanocrystalline platinum hydrides, nanocrystalline platinum nitrates,
nanocrystalline
platinum carbonates, nanocrystalline platinum sulfides, nanocrystalline
platinum
2o sulfadiazines, nanocrystalline platinum acetates, nanocrystalline platinum
lactates,
nanocrystalline platinum citrates, nanocrystalline alkali platinum
thiosulphates (e.g.,
nanocrystalline sodium platinum thiosulphate, nanocrystalline potassium
platinum
thiosulphate)), nanocrystalline palladium-containing materials (e.g.,
nanocrystalline
palladium, nanocrystalline palladium alloys, nanocrystalline palladium oxides,
nanocrystalline palladium carbides, nanocrystalline palladium nitrides,
nanocrystalline
palladium borides, nanocrystalline palladium sulfides, nanocrystalline
palladium
myristates, nanocrystalline palladium stearates, nanocrystalline palladium
oleates,
nanocrystalline palladium glutonates, nanocrystalline palladium glutonates,
nanocrystalline palladium adipates, nanocrystalline palladium silicates,
nanocrystalline
3o palladium phosphides, nanocrystalline palladium halides, nanocrystalline
palladium
hydrides, nanocrystalline palladium nitrates, nanocrystalline palladium
carbonates,
17



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
nanocrystalline palladium sulfides, nanocrystalline palladium sulfadiazines,
nanocrystalline palladium acetates, nanocrystalline palladium lactates,
nanocrystalline
palladium citrates, nanocrystalline alkali palladium thiosulphates (e.g.,
nanocrystalline
sodium palladium thiosulphate, nanocrystalline potassium palladium
thiosulphate)),
nanocrystalline iridium-containing materials (e.g., nanocrystalline iridium,
nanocrystalline iridium alloys, nanocrystalline iridium oxides,
nanocrystalline iridium
carbides, nanocrystalline iridium nitrides, nanocrystalline iridium borides,
nanocrystalline
iridium sulfides, nanocrystalline iridium myristates, nanocrystalline iridium
stearates,
nanocrystalline iridium oleates, nanocrystalline iridium glutonates,
nanocrystalline
iridium glutonates, nanocrystalline iridium adipates, nanocrystalline iridium
silicates,
nanocrystalline iridium phosphides, nanocrystalline iridium halides,
nanocrystalline
iridium hydrides, nanocrystalline iridium nitrates, nanocrystalline iridium
carbonates,
nanocrystalline iridium sulfides, nanocrystalline iridium sulfadiazines,
nanocrystalline
iridium acetates, nanocrystalline iridium lactates, nanocrystalline iridium
citrates,
nanocrystalline alkali iridium thiosulphates (e.g., nanocrystalline sodium
iridium
thiosulphate, nanocrystalline potassium iridium thiosulphate)),
nanocrystalline zinc-
containing materials (e.g., nanocrystalline zinc, nanocrystalline zinc alloys,
nanocrystalline zinc oxides, nanocrystalline zinc carbides, nanocrystalline
zinc nitrides,
nanocrystalline zinc borides, nanocrystalline zinc sulfides, nanocrystalline
zinc
2o myristates, nanocrystalline zinc stearates, nanocrystalline zinc oleates,
nanocrystalline
zinc glutonates, nanocrystalline zinc glutonates, nanocrystalline zinc
adipates,
nanocrystalline zinc silicates, nanocrystalline zinc phosphides,
nanocrystalline zinc
halides, nanocrystalline zinc hydrides, nanocrystalline zinc nitrates,
nanocrystalline zinc
carbonates, nanocrystalline zinc sulfides, nanocrystalline zinc sulfadiazines,
nanocrystalline zinc acetates, nanocrystalline zinc lactates, nanocrystalline
zinc citrates,
nanocrystalline alkali zinc thiosulphates (e.g., nanocrystalline sodium zinc
thiosulphate,
nanocrystalline potassium zinc thiosulphate)), nanocrystalline copper -
containing
materials (e.g., nanocrystalline copper, nanocrystalline copper alloys,
nanocrystalline
copper oxides, nanocrystalline copper carbides, nanocrystalline copper
nitrides,
3o nanocrystalline copper borides, nanocrystalline copper sulfides,
nanocrystalline copper
myristates, nanocrystalline copper stearates, nanocrystalline copper oleates,
18



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
.. . .._. .... "", ."", , "",~, ,.",~r
nanocrystalline copper glutonates, nanocrystalline copper glutonates,
nanocrystalline
copper adipates, nanocrystalline copper silicates, nanocrystalline copper
phosphides,
nanocrystalline copper halides, nanocrystalline copper hydrides,
nanocrystalline copper
nitrates, nanocrystalline copper carbonates, nanocrystalline copper
sulfadiazines,
nanocrystalline copper acetates, nanocrystalline copper lactates,
nanocrystalline copper
citrates, nanocrystalline alkali copper thiosulphates (e.g., nanocrystalline
sodium copper
thiosulphate, nanocrystalline potassium copper thiosulphate)), nanocrystalline
tin-
containing materials (e.g., nanocrystalline tin, nanocrystalline tin alloys,
nanocrystalline
tin oxides, nanocrystalline tin carbides, nanocrystalline tin nitrides,
nanocrystalline tin
borides, nanocrystalline tin sulfides, nanocrystalline tin myristates,
nanocrystalline tin
stearates, nanocrystalline tin oleates, nanocrystalline tin glutonates,
nanocrystalline tin
glutonates, nanocrystalline tin adipates, nanocrystalline tin silicates,
nanocrystalline tin
phosphides, nanocrystalline tin halides, nanocrystalline tin hydrides,
nanocrystalline tin
nitrates, nanocrystalline tin carbonates, nanocrystalline tin sulfides,
nanocrystalline tin
sulfadiazines, nanocrystalline tin acetates, nanocrystalline tin lactates,
nanocrystalline tin
citrates, nanocrystalline alkali tin thiosulphates (e.g., nanocrystalline
sodium tin
thiosulphate, nanocrystalline potassium tin thiosulphate)), nanocrystalline
antimony-
containing materials (e.g., nanocrystalline antimony, nanocrystalline antimony
alloys,
nanocrystalline antimony oxides, nanocrystalline antimony carbides,
nanocrystalline
2o antimony nitrides, nanocrystalline antimony borides, nanocrystalline
antimony sulfides,
nanocrystalline antimony myristates, nanocrystalline antimony stearates,
nanocrystalline
antimony oleates, nanocrystalline antimony glutonates, nanocrystalline
antimony
glutonates, nanocrystalline antimony adipates, nanocrystalline antimony
silicates,
nanocrystalline antimony phosphides, nanocrystalline antimony halides,
nanocrystalline
antimony hydrides, nanocrystalline antimony nitrates, nanocrystalline antimony
carbonates, nanocrystalline antimony sulfides, nanocrystalline antimony
sulfadiazines,
nanocrystalline antimony acetates, nanocrystalline antimony lactates,
nanocrystalline
antimony citrates, nanocrystalline alkali antimony thiosulphates (e.g.,
nanocrystalline
sodium antimony thiosulphate, nanocrystalline potassium antimony
thiosulphate)),
3o nanocrystalline bismuth containing materials (e.g., nanocrystalline
bismuth,
nanocrystalline bismuth alloys, nanocrystalline bismuth oxides,
nanocrystalline bismuth
19



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
carbides, nanocrystalline bismuth nitrides, nanocrystalline bismuth borides,
nanocrystalline bismuth sulfides, nanocrystalline bismuth myristates,
nanocrystalline
bismuth stearates, nanocrystalline bismuth oleates, nanocrystalline bismuth
glutonates,
nanocrystalline bismuth glutonates, nanocrystalline bismuth adipates,
nanocrystalline
bismuth silicates, nanocrystalline bismuth phosphides, nanocrystalline bismuth
halides,
nanocrystalline bismuth hydrides, nanocrystalline bismuth nitrates,
nanocrystalline
bismuth carbonates, nanocrystalline bismuth sulfides, nanocrystalline anti
bismuth
sulfadiazines, nanocrystalline bismuth acetates, nanocrystalline bismuth
lactates,
nanocrystalline bismuth citrates, nanocrystalline alkali bismuth thiosulphates
(e.g.,
nanocrystalline sodium bismuth thiosulphate, nanocrystalline potassium bismuth
thiosulphate)).
Examples of atomically disordered, crystalline metal-containing material
(which
may or may not also be an antimicrobial material or a nanocrystalline
material) include
atomically disordered, crystalline silver-containing materials (e.g.,
atomically disordered,
~ 5 crystalline silver; atomically disordered, crystalline silver alloys;
atomically disordered,
crystalline silver oxides; atomically disordered, crystalline silver carbides;
atomically
disordered, crystalline silver nitrides; atomically disordered, crystalline
silver borides;
atomically disordered, crystalline silver sulfides; atomically disordered,
crystalline silver
myristates; atomically disordered, crystalline silver stearates; atomically
disordered,
2o crystalline silver oleates; atomically disordered, crystalline silver
glutonates; atomically
disordered, crystalline silver glutonates; atomically disordered, crystalline
silver adipates;
atomically disordered, crystalline silver silicates; atomically disordered,
crystalline silver
phosphides; atomically disordered, crystalline silver halides; atomically
disordered,
crystalline silver hydrides, atomically disordered, crystalline silver
nitrates; atomically
25 disordered, crystalline silver carbonates; atomically disordered,
crystalline silver sulfides;
atomically disordered, crystalline silver sulfadiazines; atomically
disordered, crystalline
silver acetates; atomically disordered, crystalline silver lactates;
atomically disordered,
crystalline silver citrates; atomically disordered, crystalline alkali silver
thiosulphates
(e.g., atomically disordered, crystalline sodium silver thiosulphate,
atomically disordered,
3o crystalline potassium silver thiosulphate)), atomically disordered,
crystalline gold-
containing materials (atomically disordered, crystalline gold; atomically
disordered,



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
crystalline gold alloys; atomically disordered, crystalline gold oxides;
atomically
disordered, crystalline gold carbides; atomically disordered, crystalline gold
nitrides;
atomically disordered, crystalline gold borides; atomically disordered,
crystalline gold
sulfides; atomically disordered, crystalline gold myristates; atomically
disordered,
crystalline gold stearates; atomically disordered, crystalline gold oleates;
atomically
disordered, crystalline gold glutonates; atomically disordered, crystalline
gold glutonates;
atomically disordered, crystalline gold adipates; atomically disordered,
crystalline gold
silicates; atomically disordered, crystalline gold phosphides; atomically
disordered,
crystalline gold halides; atomically disordered, crystalline gold hydrides,
atomically
disordered, crystalline gold nitrates; atomically disordered, crystalline gold
carbonates;
atomically disordered, crystalline gold sulfides; atomically disordered,
crystalline gold
sulfadiazines; atomically disordered, crystalline gold acetates; atomically
disordered,
crystalline gold lactates; atomically disordered, crystalline gold citrates;
atomically
disordered, crystalline alkali gold thiosulphates (e.g., atomically
disordered, crystalline
~ 5 sodium gold thiosulphate, atomically disordered, crystalline potassium
gold
thiosulphate)), atomically disordered, crystalline platinum-containing
materials (e.g.,
atomically disordered, crystalline platinum; atomically disordered,
crystalline platinum
alloys; atomically disordered, crystalline platinum oxides; atomically
disordered,
crystalline platinum carbides; atomically disordered, crystalline platinum
nitrides;
2o atomically disordered, crystalline platinum borides; atomically disordered,
crystalline
platinum sulfides; atomically disordered, crystalline platinum myristates;
atomically
disordered, crystalline platinum stearates; atomically disordered, crystalline
platinum
oleates; atomically disordered, crystalline platinum glutonates; atomically
disordered,
crystalline platinum glutonates; atomically disordered, crystalline platinum
adipates;
25 atomically disordered, crystalline platinum silicates; atomically
disordered, crystalline
platinum phosphides; atomically disordered, crystalline platinum halides;
atomically
disordered, crystalline platinum hydrides, atomically disordered, crystalline
platinum
nitrates; atomically disordered, crystalline platinum carbonates; atomically
disordered,
crystalline platinum sulfides; atomically disordered, crystalline platinum
sulfadiazines;
3o atomically disordered, crystalline platinum acetates; atomically
disordered, crystalline
platinum lactates; atomically disordered, crystalline platinum citrates;
atomically
21



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
disordered, crystalline allcali platinum thiosulphates (e.g., atomically
disordered,
crystalline sodium platinum thiosulphate, atomically disordered, crystalline
potassium
platinum thiosulphate), atomically disordered, crystalline palladium-
containing materials
(e.g., atomically disordered, crystalline palladium; atomically disordered,
crystalline
palladium alloys; atomically disordered, crystalline palladium oxides;
atomically
disordered, crystalline palladium carbides; atomically disordered, crystalline
palladium
nitrides; atomically disordered, crystalline palladium borides; atomically
disordered,
crystalline palladium sulfides; atomically disordered, crystalline palladium
myristates;
atomically disordered, crystalline palladium stearates; atomically disordered,
crystalline
1 o palladium oleates; atomically disordered, crystalline palladium
glutonates; atomically
disordered, crystalline palladium glutonates; atomically disordered,
crystalline palladium
adipates; atomically disordered, crystalline palladium silicates; atomically
disordered,
crystalline palladium phosphides; atomically disordered, crystalline palladium
halides;
atomically disordered, crystalline palladium hydrides, atomically disordered,
crystalline
~5 palladium nitrates; atomically disordered, crystalline palladium
carbonates; atomically
disordered, crystalline palladium sulfides; atomically disordered, crystalline
palladium
sulfadiazines; atomically disordered, crystalline palladium acetates;
atomically
disordered, crystalline palladium lactates; atomically disordered, crystalline
palladium
citrates; atomically disordered, crystalline alkali palladium thiosulphates
(e.g., atomically
2o disordered, crystalline sodium palladium thiosulphate, atomically
disordered, crystalline
potassium palladium thiosulphate)), atomically disordered, crystalline iridium-
containing
materials (e.g., atomically disordered, crystalline iridium; atomically
disordered,
crystalline iridium alloys; atomically disordered, crystalline iridium oxides;
atomically
disordered, crystalline iridium carbides; atomically disordered, crystalline
iridium
25 nitrides; atomically disordered, crystalline iridium borides; atomically
disordered,
crystalline iridium sulfides; atomically disordered, crystalline iridium
myristates;
atomically disordered, crystalline iridium stearates; atomically disordered,
crystalline
iridium oleates; atomically disordered, crystalline iridium glutonates;
atomically
disordered, crystalline iridium glutonates; atomically disordered, crystalline
iridium
3o adipates; atomically disordered, crystalline iridium silicates; atomically
disordered,
crystalline iridium phosphides; atomically disordered, crystalline iridium
halides;
22



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
atomically disordered, crystalline iridium hydrides, atomically disordered,
crystalline
iridium nitrates; atomically disordered, crystalline iridium carbonates;
atomically
disordered, crystalline iridium sulfides; atomically disordered, crystalline
iridium
sulfadiazines; atomically disordered, crystalline iridium acetates; atomically
disordered,
crystalline iridium lactates; atomically disordered, crystalline iridium
citrates; atomically
disordered, crystalline alkali iridium thiosulphates (e.g., atomically
disordered, crystalline
sodium iridium thiosulphate, atomically disordered, crystalline potassium
iridium
thiosulphate)), atomically disordered, crystalline zinc-containing materials
(e.g.,
atomically disordered, crystalline zinc; atomically disordered, crystalline
zinc alloys;
atomically disordered, crystalline zinc oxides; atomically disordered,
crystalline zinc
carbides; atomically disordered, crystalline zinc nitrides; atomically
disordered,
crystalline zinc borides; atomically disordered, crystalline zinc sulfides;
atomically
disordered, crystalline zinc myristates; atomically disordered, crystalline
zinc stearates;
atomically disordered, crystalline zinc oleates; atomically disordered,
crystalline zinc
~ 5 glutonates; atomically disordered, crystalline zinc glutonates; atomically
disordered,
crystalline zinc adipates; atomically disordered, crystalline zinc silicates;
atomically
disordered, crystalline zinc phosphides; atomically disordered, crystalline
zinc halides;
atomically disordered, crystalline zinc hydrides, atomically disordered,
crystalline zinc
nitrates; atomically disordered, crystalline zinc carbonates; atomically
disordered,
2o crystalline zinc sulfides; atomically disordered, crystalline zinc
sulfadiazines; atomically
disordered, crystalline zinc acetates; atomically disordered, crystalline zinc
lactates;
atomically disordered, crystalline zinc citrates; atomically disordered,
crystalline alkali
zinc thiosulphates (e.g., atomically disordered, crystalline sodium zinc
thiosulphate,
atomically disordered, crystalline potassium zinc thiosulphate)), atomically
disordered,
25 crystalline copper-containing materials (e.g., atomically disordered,
crystalline copper;
atomically disordered, crystalline copper alloys; atomically disordered,
crystalline copper
oxides; atomically disordered, crystalline copper carbides; atomically
disordered,
crystalline copper nitrides; atomically disordered, crystalline copper
borides; atomically
disordered, crystalline copper sulfides; atomically disordered, crystalline
copper
3o myristates; atomically disordered, crystalline copper stearates; atomically
disordered,
crystalline copper oleates; atomically disordered, crystalline copper
glutonates;
23



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
atomically disordered, crystalline copper glutonates; atomically disordered,
crystalline
copper adipates; atomically disordered, crystalline copper silicates;
atomically
disordered, crystalline copper phosphides; atomically disordered, crystalline
copper
halides; atomically disordered, crystalline copper hydrides, atomically
disordered,
crystalline copper nitrates; atomically disordered, crystalline copper
carbonates;
atomically disordered, crystalline copper sulfides; atomically disordered,
crystalline
copper sulfadiazines; atomically disordered, crystalline copper acetates;
atomically
disordered, crystalline copper lactates; atomically disordered, crystalline
copper citrates;
atomically disordered, crystalline alkali copper thiosulphates (e.g.,
atomically disordered,
crystalline sodium copper thiosulphate, atomically disordered, crystalline
potassium
copper thiosulphate)), atomically disordered, crystalline tin-containing
materials (e.g.,
atomically disordered, crystalline tin; atomically disordered, crystalline tin
alloys;
atomically disordered, crystalline tin oxides; atomically disordered,
crystalline tin
carbides; atomically disordered, crystalline tin nitrides; atomically
disordered, crystalline
~ 5 tin borides; atomically disordered, crystalline tin sulfides; atomically
disordered,
crystalline tin myristates; atomically disordered, crystalline tin stearates;
atomically
disordered, crystalline tin oleates; atomically disordered, crystalline tin
glutonates;
atomically disordered, crystalline tin glutonate's; atomically disordered,
crystalline tin
adipates; atomically disordered, crystalline tin silicates; atomically
disordered, crystalline
2o tin phosphides; atomically disordered, crystalline tin halides; atomically
disordered,
crystalline tin hydrides, atomically disordered, crystalline tin nitrates;
atomically
disordered, crystalline tin carbonates; atomically disordered, crystalline tin
sulfides;
atomically disordered, crystalline tin sulfadiazines; atomically disordered,
crystalline tin
acetates; atomically disordered, crystalline tin lactates; atomically
disordered, crystalline
2s tin citrates; atomically disordered, crystalline alkali tin thiosulphates
(e.g., atomically
disordered, crystalline sodium tin thiosulphate, atomically disordered,
crystalline
potassium tin thiosulphate)), atomically disordered, crystalline antimony-
containing
materials (e.g., atomically disordered, crystalline antimony; atomically
disordered,
crystalline antimony alloys; atomically disordered, crystalline antimony
oxides;
3o atomically disordered, crystalline antimony carbides; atomically
disordered, crystalline
antimony nitrides; atomically disordered, crystalline antimony borides;
atomically
24



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
disordered, crystalline antimony sulfides; atomically disordered, crystalline
antimony
myristates; atomically disordered, crystalline antimony stearates; atomically
disordered,
crystalline antimony oleates; atomically disordered, crystalline antimony
glutonates;
atomically disordered, crystalline antimony glutonates; atomically disordered,
crystalline
antimony adipates; atomically disordered, crystalline antimony silicates;
atomically
disordered, crystalline antimony phosphides; atomically disordered,
crystalline antimony
halides; atomically disordered, crystalline antimony hydrides, atomically
disordered,
crystalline antimony nitrates; atomically disordered, crystalline antimony
carbonates;
atomically disordered, crystalline antimony sulfides; atomically disordered,
crystalline
antimony sulfadiazines; atomically disordered, crystalline antimony acetates;
atomically
disordered, crystalline go antimony ld lactates; atomically disordered,
crystalline
antimony citrates; atomically disordered, crystalline alkali antimony
thiosulphates (e.g.,
atomically disordered, crystalline sodium antimony thiosulphate, atomically
disordered,
crystalline potassium antimony thiosulphate)), atomically disordered,
crystalline bismuth-
~5 containing materials (e.g., atomically disordered, crystalline bismuth;
atomically
disordered, crystalline bismuth alloys; atomically disordered, crystalline
bismuth oxides;
atomically disordered, crystalline bismuth carbides; atomically disordered,
crystalline
bismuth nitrides; atomically disordered, crystalline bismuth borides;
atomically
disordered, crystalline bismuth sulfides; atomically disordered, crystalline
bismuth
2o myristates; atomically disordered, crystalline bismuth stearates;
atomically disordered,
crystalline bismuth oleates; atomically disordered, crystalline bismuth
glutonates; 1
atomically disordered; crystalline bismuth glutonates; atomically disordered,
crystalline
bismuth adipates; atomically disordered, crystalline bismuth silicates;
atomically
disordered, crystalline bismuth phosphides; atomically disordered, crystalline
bismuth
25 halides; atomically disordered, crystalline bismuth hydrides, atomically
disordered,
crystalline bismuth nitrates; atomically disordered, crystalline bismuth
carbonates;
atomically disordered, crystalline bismuth sulfides; atomically disordered,
crystalline
bismuth sulfadiazines; atomically disordered, crystalline bismuth acetates;
atomically
disordered, crystalline bismuth lactates; atomically disordered, crystalline
bismuth
3o citrates; atomically disordered, crystalline alkali bismuth thiosulphates
(e.g., atomically



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
disordered, crystalline sodium bismuth thiosulphate, atomically disordered,
crystalline
potassium bismuth thiosulphate)).
Subi ects
The metal-containing material can be used to treat, for example a human or an
animal (e.g., a dog, a cat, a horse, a bird, a reptile, an amphibian, a fish,
a turtle, a guinea
pig, a hamster, a rodent, a cow, a pig, a goat, a primate, a monkey, a
chicken, a turkey, a
buffalo, an ostrich, a sheep, a llama).
Conditions and Condition Locations
The conditions that can be treated with the metal-containing material include,
for
example, bacterial conditions, microbial conditions, biofilm conditions,
inflammatory
conditions, fungal conditions, viral conditions, autoimmune conditions,
idiopathic
conditions, hyperproliferative conditions, noncancerous growths and/or
cancerous
~5 conditions (e.g., tumorous conditions, hematologic malignancies). Such
conditions can
be associated with, for example, one or more prions, parasites, fungi, viruses
and/or
bacteria. In general, the location of the condition to be treated corresponds
to the type of
condition to be treated.
In some embodiments, the condition can be a skin condition or a integument
20 condition (e.g., a bacterial skin condition, a microbial skin condition, a
biofilm skin
condition, an inflammatory skin condition, a hyperproliferative skin
condition, a fungal
skin condition, a viral skin condition, an autoimmune skin condition, an
idiopathic skin
condition, a hyperproliferative skin condition, a cancerous skin condition, a
microbial
integument condition, an inflammatory integument condition, a fungal
integument
2s condition, a viral integument condition, an autoimmune integument
condition, an
idiopathic integument condition, a hyperproliferative integument condition, a
cancerous
integument condition). Examples of skin conditions or integument conditions
include
burns, eczema (e.g., atopic eczema, acrodermatitis continua, contact allergic
dermatitis,
contact irritant dermatitis, dyshidrotic eczema, pompholyx, lichen simplex
chronicus,
so nummular eczema, seborrheic dermatitis, stasis eczema), erythroderma,
insect bites,
mycosis fungoides, pyoderma gangrenosum, eythrema multiforme, rosacea,
26



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
onychomycosis, acne (e.g., acne vulgaris, neonatal acne, infantile acne,
pomade acne),
psoriasis, Reiter's syndrome, pityriasis rubra pilaris, hyperpigmentation,
vitiligo, scarnng
conditions (e.g., hypertropic scarnng), keloids, lichen planus, age-related
skin disorders
(e.g., wrinkles, cellulite) and hyperproliferative skin disorders, such as,
for example,
hyperproliferative variants of the disorders of keratinization (e.g., actinic
keratosis, senile
keratosis). Generally, the treatment of skin or integument conditions involves
contacting
the metal-containing material with the area of the skin having the condition.
As an
example, a skin or integument condition can be treated by contacting the area
of skin
having the condition with a dressing having a coating of the metal-containing
material.
As another example, a skin or integument condition can be treated by
contacting the area
of skin having the condition with a solution containing the metal-containing
material. As
an additional example, a skin or integument condition can be treated by
contacting the
area of skin having the condition with a pharmaceutical carrier composition
containing
the metal-containing material. In the case of onychomycosis, the material may
be applied
~5 to the nail in an appropriate form (see below) such that the material
penetrates the hard
nail to contact the affected area.
In certain embodiments, the condition can be a respiratory condition (e.g., a
bacterial respiratory condition, a biofilm respiratory condition, a microbial
respiratory
condition, an inflammatory respiratory condition, a fungal respiratory
condition, a viral
2o respiratory condition, an autoimmune respiratory condition, an idiopathic
respiratory
condition, a hyperproliferative respiratory condition, a cancerous respiratory
condition).
Examples of respiratory conditions include asthma, emphysema, bronchitis,
pulmonary
edema, acute respiratory distress syndrome, bronchopulmonary dysplasia,
fibrotic
conditions (e.g., pulmonary fibrosis), pulmonary atelectasis, tuberculosis,
pneumonia,
25 sinusitis, allergic rhinitis, pharyngitis, mucositis, stomatitis, chronic
obstructive
pulmonary disease, bronchiectasis, lupus pneumonitis and cystic fibrosis. In
general, the
treatment of respiratory conditions involves contacting the metal-containing
material with
the area of the respiratory system having the condition. Areas of the
respiratory system
include, for example, the oral cavity, the nasal cavity, and the lungs. As an
example,
so certain respiratory conditions can be treated by inhaling a free standing
powder of the
metal-containing material (e.g., with a dry powder inhaler). As another
example, certain
27



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
respiratory conditions can be treated by inhaling an aerosol containing the
metal-
containing material (e.g., with an inhaler).
In some embodiments, the condition can be a musculo-skeletal condition (e.g.,
a
bacterial musculo-slceletal condition, a biofilm musculo-skeletal condition, a
microbial
musculo-skeletal condition, an inflammatory musculo-skeletal condition, a
fungal
musculo-skeletal condition, a viral musculo-skeletal condition, an autoimmune
musculo-
skeletal condition, an idiopathic musculo-skeletal condition, a
hyperproliferative
musculo-skeletal condition, a cancerous musculo-skeletal condition). A musculo-
skeletal
condition can be, for example, a degenerative musculo-skeletal condition
(e.g., arthritis)
or a traumatic musculo-skeletal condition (e.g., a torn or damaged muscle).
Examples of
musculo-skeletal conditions include tendonitis, osteomyelitis, fibromyalgia,
bursitis and
arthritis. Generally, the treatment of musculo-skeletal conditions involves
contacting the
metal-containing material material with the area of the musculo-skeletal
system having
the condition. Areas of the musculo-skeletal system include, for example, the
joints, the
~ 5 muscles, and the tendons. As an example, certain musculo-skeletal
conditions can be
treated by injecting (e.g., via a small needle injector) a solution containing
the metal-
containing material into the subj ect. As another example, certain musculo-
skeletal
conditions can be treated by injecting (e.g., via a needleless injector) a
free standing
powder of the metal-containing material into the subject. As an additional
example,
2o certain musculo-skeletal conditions can be treated by using a
pharmaceutical carrier
composition of the metal-containing material, such as a penetrating
pharmaceutical
Garner composition of the metal-containing material (e.g., a composition
containing
DMSO).
hl certain embodiments, the condition can be a circulatory condition (e.g., a
25 bacterial circulatory condition, a biofilm circulatory condition, a
microbial circulatory
condition, an inflammatory circulatory condition, a fungal circulatory
condition, a viral
circulatory condition, an autoimmune circulatory condition, an idiopathic
circulatory
condition, a hyperproliferative circulatory condition, a cancerous circulatory
condition).
As referred to herein, circulatory conditions include lymphatic conditions.
Examples of
3o circulatory conditions include arteriosclerosis, lymphoma, septicemia,
leukemia,
ischemic vascular disease, lymphangitis and atherosclerosis. In general, the
treatment of
28



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
circulatory conditions involves contacting the metal-containing material with
the area of
the circulatory system having the condition. Areas of the circulatory system
include, for
example, the heart, the lymphatic system, blood, blood vessels (e.g.,
arteries, veins). As
an example, certain circulatory conditions can be treated by injecting (e.g.,
via a small
needle injector) a solution containing the metal-containing material into the
subject. As
another example, certain circulatory conditions can be treated by injecting
(e.g., via a
needleless injector) a free standing powder of the metal-containing material
into the
subj ect.
In some embodiments, the condition can be a mucosal or serosal condition
(e.g., a
1 o bacterial mucosal or serosal condition, a biofihn mucosal or serosal
condition, a
microbial mucosal or serosal condition, an inflammatory mucosal or serosal
condition, a
fungal mucosal or serosal condition, a viral mucosal or serosal condition, an
autoimmune
mucosal or serosal condition, an idiopathic mucosal or serosal condition, a
hyperproliferative mucosal or serosal condition, a cancerous mucosal or
serosal
~ 5 condition). Examples of mucosal or serosal conditions include
pericarditis, Bowen's
disease, stomatitis, prostatitis, sinusitis, allergic rhinitis, digestive
disorders, peptic ulcers,
esophageal ulcers, gastric ulcers, duodenal ulcer, espohagitis, gastritis,
enteritis,
enterogastric intestinal hemorrhage, toxic epidermal necrolysis syndrome,
Stevens
Johnson syndrome, fibrotic condition (e.g., cystic fibrosis), bronchitis,
pneumonia (e.g.,
2o nosocomial pneumonia, ventilator-assisted pneumonia), pharyngitis, connnon
cold, ear
infections, sore throat, sexually transmitted diseases (e.g., syphilis,
gonorrhea, herpes,
genital warts, HIV, chlamydia), inflammatory bowel disease, colitis,
hemorrhoids, thrush,
dental conditions, oral conditions, conjunctivitis, and periodontal
conditions. Generally,
the treatment of mucosal or serosal conditions involves contacting the metal-
containing
25 material with the area of a mucosal or serosal region having the condition.
Mucosal or
serosal areas include, for example, the oral cavity, the nasal cavity, the
colon, the small
intestine, the large intestine, the stomach, and the esophagus. As an example,
certain
mucosal or serosal conditions can be treated by inhaling a free standing
powder of the
metal-containing material (e.g., with a dry powder inhaler). As another
example, certain
3o mucosal or serosal conditions can be treated by inhaling an aerosol
containing the metal-
containing material (e.g., with an inhaler). As an additional example, certain
mucosal or
29



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
serosal conditions can be treated by gargling or spraying a solution of the
metal-
containing material. As another example, certain mucosal or serosal conditions
can be
treated using a suppository. As a further example, certain mucosal or serosal
conditions
can be treated by an enema.
In embodiments in which the metal-containing material is used to treat
hyperproliferation of cell growth (e.g., cancerous conditions, such as
malignant tumors,
or non-cancerous conditions, such as benign tumors), the metal-containing
material can
be used to induce apoptosis (programmed cell death), modulate matrix
metalloproteinases
(MMPs) and/or modulates cytokines by contacting affected tissue (e.g., a
hyperplastic
tissue, a tumor tissue or a cancerous lesion) with the metal-containing
material. It has
been observed that the metal-containing material (e.g., an antimicrobial, anti-
biofilm,
antibacterial, anti-inflammatory, antifungal, antiviral, anti-autoimmune, anti-
cancer, pro-
apoptosis, anti-proliferative, and/or MMP modulating, nanocrystalline and/or
atomically
disordered, silver-containing material) can be effective in preventing
production of a high
~ 5 number of MMPs and/or cytokines by certain cells without necessarily
reducing MMP
and/or cytokine production by the same cells to about zero. It is believed,
however, that
in certain embodiments, the metal-containing material can be used to inhibit
MMP and/or
cytokine production (e.g., bring MMP and/or cytokine production to normal
levels,
desired levels, and/or about zero) in certain cells.
2o MMPs refer to any protease of the family of MMPs which are involved in the
degradation of connective tissues, such as collagen, elastins, fibronectin,
laminin, and
other components of the extracellular matrix, and associated with conditions
in which
excessive degradation of extracellular matrix occurs, such as tumor invasion
and
metastasis. Examples of MMPs include MMP-2 (secreted by fibroblasts and a wide
25 variety of other cell types) and MMP-9 (released by mononuclear phagocytes,
neutrophils, corneal epithelial cells, tumor cells, cytotrophoblasts and
keratinocytes).
Cytokine refers to a nonimmunoglobulin polypeptide secreted by monocytes and
lymphocytes in response to interaction with a specific antigen, a nonspecific
antigen, or a
nonspecific soluble stimulus (e.g., endotoxin, other cytokines). Cytokines
affect the
3o magnitude of inflammatory or immune responses. Cytokines can be divided
into several
groups, which include interferons, tumor necrosis factor (TNF), interleukins
(IL-1 to IL-



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
~), transforming growth factors, and the hematopoietic colony-stimulating
factors. An
example of a cytokine is TNF-a. A fibroblast is an area connective tissue cell
which is a
flat-elongated cell with cytoplasmic processes at each end having a flat, oval
vesicular
nucleus. Fibroblasts which differentiate into chondroblasts, collagenoblasts,
and
osteoblasts form the fibrous tissues in the body, tendons, aponeuroses,
supporting and
binding tissues of all sorts. Hyperplastic tissue refers to tissue in which
there is an
abnormal multiplication or increase in the number of cells in a normal
arrangement in
normal tissue or an organ. A tumor refers to spontaneous growth of tissue in
which
multiplication of cells is abnormal, uncontrolled and progressive. A tumor
generally
serves no useful function and grows at the expense of the healthy organism. A
cancerous
lesion is a tumor of epithelial tissue, or malignant, new growth made up of
epithelial cells
tending to infiltrate surrounding tissues and to give rise to metastases. As
used in
reference to the skin, a cancerous lesion means a lesion which may be a result
of a
primary cancer, or a metastasis to the site from a local tumor or from a tumor
in a distant
~ 5 site: It may take the form of a cavity, an open area on the surface of the
skin, skin
nodules, or a nodular growth extending from the surface of the skin.
Conditions characterized by undesirable MMP activity include ulcers, asthma,
acute respiratory distress syndrome, skin disorders, skin aging, keratoconus,
restenosis,
osteo- and rheumatoid arthritis, degenerative joint disease, bone disease,
wounds, cancer
2o including cell proliferation, invasiveness, metastasis (carcinoma,
fibrosarcoma,
osteosarcoma), hypovolemic shock, periodontal disease, epidennolysis bullosa,
scleritis,
atherosclerosis, multiple sclerosis, inflammatory diseases of the central
nervous system,
vascular leakage syndrome, collagenase induced disease, adhesions of the
peritoneum,
strictures of the esophagus or bowel, ureteral or urethral strictures, and
biliary strictures.
25 Excessive TNF production has been reported in diseases which are
characterized by
excessive MMP activity, such as autoimmune disease, cancer, cachexia, HIV
infection,
and cardiovascular conditions.
Forms of the Material and Methods of Applying' the Material
so In general, the metal-containing material can be in any desired form or
formulation. For example, the material can be a coating on a substrate (e.g.,
in the form
31



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
of a dressing, a coated medical implant), a free standing powder, a solution,
or disposed
within a pharmaceutically acceptable Garner.
In some embodiments, the metal-containing material can act as a preservative.
In
such embodiments, a form or formulation containing the metal-containing
material can be
prepared with or without additional preservatives. Moreover, in embodiments in
which
the metal-containing material acts as a preservative, the metal-containing
material may be
included in a therapeutic formulation containing other therapeutic agents
(e.g., the metal-
containing material may be included primarily in certain therapeutic
compositions to act
as a preservative).
Moreover, the material can be applied to the subject in any of a variety of
ways,
generally depending upon the form of the material as applied and/or the
location of the
condition to be treated. In general, the amount of material used is selected
so that the
desired therapeutic effect (e.g., reduction in the condition being treated) is
achieved while
the material introduces an acceptable level of toxicity (e.g., little or no
toxicity) to the
~ 5 subj ect. Generally, the amount of the material used will vary with the
conditions being
treated, the stage of advancement of the condition, the age and type of host,
and the type,
concentration and form of the material as applied. Appropriate amounts in any
given
instance will be readily apparent to those skilled in the art or capable of
determination by
routine experimentation. In some embodiments, a single application of the
material may
2o be sufficient. In certain embodiments, the material may be applied
repeatedly over a
period of time, such as several times a day for a period of days, weeks,
months or years.
Substrate Coatings
Examples of commercially available metal-containing materials include the
25 Acticoat~ family of dressings (Smith & Nephew, Hull, UI~), which are formed
of
antimicrobial, anti-inflammatory atomically disordered, nanocrystalline silver-
containing
material coated on one or more substrates. Such dressings include the
Acticoat~
dressings, the Acticoat7~ dressings, the Acticoat~ moisture coating dressings,
and the
Acticoat~ absorbent dressings.
3o A coating of a metal-containing material (e.g., an antimicrobial,
atomically
disordered, nanocrystalline silver-containing material) can be formed on a
substrate using
32



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
a desired technique. In certain embodiments, the coating is formed by
depositing the
material on the substrate surface using chemical vapor deposition, physical
vapor
deposition, and/or liquid phase deposition. Exemplary deposition methods
include
vacuum evaporation deposition, arc evaporation deposition, sputter deposition,
magnetron sputter deposition and ion plating.
In some embodiments, the coating is prepared using physical vapor deposition.
Fig. 1 shows a vapor deposition system 100 that includes a vacuum chamber 110,
an
energy source 120 (e.g., an electron beam source, an ion source, a laser beam,
a
magnetron source), a target 130 and a substrate 140. During operation, energy
source
120 directs a beam of energy 122 to target 130, causing material 132 to be
removed (e.g.,
by evaporation) from target 130 and directed to a surface 142 of substrate
140. At least a
portion of the removed material 132 is deposited on surface 142.
W general, the values of the system parameters (e.g., the temperature of
surface
142, the pressure of chamber 110, the angle of incidence of removed material
132 on
~5 surface 142, the distance between target 130 and surface 142) can be
selected as desired.
The temperature of surface 142 can be relatively low during the deposition
process. For
example, during the deposition process, the ratio of the temperature of
substrate 140 to
the melting point of the material forming target 130 (as determined in using
Kelvin) can
be about 0.5 or less (e.g., about 0.4 or less, about 0.35 or less, about 0.3
or less).
2o The pressure in chamber 110 can be relatively high. For example, vacuum
evaporation deposition, electron beam deposition or arc evaporation, the
pressure can be
about 0.01 milliTorr or greater. For gas scattering evaporation (pressure
plating) or
reactive arc evaporation, the pressure in chamber 110 can be about 20
milliTorr or
greater. For sputter deposition, the pressure in chamber 110 can be about 75
milliTorr or
25 greater. For magnetron sputter deposition, the pressure in chamber 110 can
be about 10
milliTorr or greater. For ion plating, the pressure in chamber 110 can be 200
milliTorr or
greater.
The angle of incidence of removed material 132 on surface 142 (8) can be
relatively low. For example, the angle of incidence of removed material 132 on
surface
30 142 can be about 75° or less (e.g., about 60° or less, about
45° or less, about 30° or less).
33



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
The distance between target 130 and surface 142 can be selected based upon the
values of the other system parameters. For example, the distance between
target 130 and
surface 142 can be about 250 millimeters or less (e.g., about 150 millimeters
or less, 125
millimeters or less, about 100 millimeters or less, about 90 millimeters or
less, about 80
millimeters or less, about 70 millimeters or less, about 60 millimeters or
less, about 50
millimeters or less, about 40 millimeters or less).
As noted above, it is believed that, the metal-containing material, when
contacted
with an alcohol or water-based electrolyte, can be released into the alcohol
or water-
based electrolyte (e.g., as ions, atoms, molecules and/or clusters). It is
also believed that
the ability to release the metal (e.g., as atoms, ions, molecules and/or
clusters) on a
sustainable basis from a coating is generally dependent upon a number of
factors,
including coating characteristics such as composition, structure, solubility
and thickness,
and the nature of the environment in which the device is used. As the level of
atomic
disorder is increased, it is believed that the amount of metal species
released per unit time
increases. For example, a silver metal film deposited by magnetron sputtering
at a ratio
of substrate temperature to the target melting point of less than about 0.5
and a working
gas pressure of about 0.93 Pascals (about seven milliTorr) releases
approximately 1/3 of
the silver ions that a film deposited under similar conditions, but at four
Pascals (about 30
milliTorr), will release over 10 days. Coatings formed with an intermediate
structure
20 (e.g., lower pressure, lower angle of incidence etc.) have been observed to
have metal
(e.g., silver) release values intermediate to these values as determined by
bioassays. In
general, to obtain relatively slow release of the metal, the coating should
have a relatively
low degree of atomic disorder, and, to obtain relatively fast release of the
metal, the
coating should have a relatively high degree of atomic disorder.
25 For continuous, uniform coatings, the time for total dissolution is
generally a
function of coating thickness and the nature of the environment to which the
coating is
exposed. The release of metal is believed to increase approximately linearly
as the
thickness of the coating is increased. For example, it has been observed that
a two fold
increase in coating thickness can result in about a two fold increase in
longevity.
3o In certain embodiments, it is possible to manipulate the degree of atomic
disorder,
and therefore the metal release from a coating, by forming a thin film coating
with a
34



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
modulated structure. For example, a coating deposited by maytrqn shuttering
such that
the working gas pressure was relatively low (e.g., about two Pascals or about
15
milliTorr) for about 50% of the deposition time and relatively high (e.g.,
about four
Pascals or 30 milliTorr) for the remaining time, can result in a relatively
rapid initial
release of metal (e.g., ions, clusters, atoms, molecules), followed by a
longer period of
slow release. This type of coating is can be particularly effective on devices
such as
urinary catheters for which an initial rapid release is advantageous to
achieve quick
antimicrobial concentrations followed by a lower release rate to sustain the
concentration
of metal (e.g., ions, clusters, atoms, molecules) over a period of weeks.
It is further believed that the degree of atomic disorder of a coating can be
manipulated by introducing one or more dissimilar materials into the coating.
For
example, one or more gases can be present in chamber 110 during the deposition
process.
Examples of such gases include oxygen-containing gases (e.g., oxygen, air,
water),
nitrogen-containing gases (e.g., nitrogen), hydrogen-containing gases (e.g.,
water,
~5 hydrogen), boron-containing gases (e.g., boron), sulfur-containing gases
(e.g., sulfur),
carbon-containing gases (e.g., carbon monoxide, carbon dioxide), phosphorus-
containing
gases, silicon-containing gases, and halogen-containing gases (e.g., fluorine,
chlorine,
bromine, iodine). The additional gases) can be co-deposited or reactively
deposited with
material 132. This can result in the deposition/formation of an oxide,
hydroxide, nitride,
2o carbide, phosphide, silicate, boride, sulfide, hydride, nitrate, carbonate,
alkali
thiosulphate (e.g., sodium thiosulphate, potassium thiosulphate), myristate,
sorbate,
stearate, oleate, glutonate, adipate, silicate, phosphide, sulfadiazine,
acetate, lactate,
citrate and/or halide material (e.g., an oxide of a metal-containing material,
a hydroxide
of a metal-containing material, a nitride of a metal-containing material, a
carbide of a
25 metal-containing material, a phosphide of a metal-containing material, a
silicate of a
metal-containing material, a boride of a metal-containing material, a sulfide
of a metal-
containing material, a hydride of a metal-containing material, a halide of a
metal-
containing material, a nitrate of a metal-containing material, a carbonate of
a metal-
containing material, a myristate of a metal-containing material, a sorbate of
a metal-
3o containing material, a stearate of a metal-containing material, an oleate
of a metal-
containing material, a glutonate of a metal-containing material, an adipate of
a metal-



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
containing material, a silicate of a metal-containing material, a phosphide of
a metal-
containing material, a sulfide of a metal-containing material, a sulfadiazine
of a metal-
containing material, a sulfadiazine of a metal-containing material, an acetate
of a metal-
containing material, a lactate of a metal-containing material, a citrate of a
metal-
s containing material, an allcali metal thiosulphate (e.g., sodium metal
thiosulphate,
potassium metal thiosulphate) of a metal-containing material). Without wishing
to be
bound by theory, it is believed that atoms and/or molecules of the additional
gases) may
become absorbed or trapped in the material, resulting in enhanced atomic
disorder. The
additional gases) may be continuously supplied during deposition, or may be
pulsed to
(e.g., for sequential deposition). In embodiments, the material formed can be
constituted
of a material with a ratio of material 132 to additional gases) of about 0.2
or greater.
The presence of dissimilar atoms or molecules in the coating can enhance the
degree of
atomic disorder of the coating due to the difference in atomic radii of the
dissimilar
constituents in the coating.
~ 5 The presence of dissimilar atoms or molecules in the coating may also be
achieved by co-depositing or sequentially depositing one or more additional
metal
elements (e.g., one or more additional antimicrobial metal elements). Such
additional
metal elements include, for example, Au, Pt, Ta, Ti, Nb, Zn, V, Hf, Mo, Si,
Al, and other
transition metal elements. It is believed that the presence of dissimilar
metal elements
20 (one or more primary metal elements and one or more additional metal
elements) in the
coating can reduce atomic diffusion and stabilize the atomically disordered
structure of
the coating. A coating containing dissimilar metal elements can be formed, for
example,
using thin film deposition equipment with multiple targets. In some
embodiments,
sequentially deposited layers of the metal elements are discontinuous (e.g.,
islands within
25 a the primary metal). In certain embodiments, the weight ratio of the
additional metals)
to the primary metals) is greater than about 0.2.
While Fig. 1 shows one embodiment of a deposition system, other embodiments
are possible. For example, the deposition system can be designed such that
during
operation the substrate moves along rollers. Additionally or alternatively,
the deposition
3o system may contain multiple energy sources, multiple targets, andlor
multiple substrates.
36



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
The multiple energy sources, targets and/or substrates can be, for example,
positioned in
a line, can be staggered, or can be in an array.
In certain embodiments, two layers of the material are deposited on the
substrate
to achieve an optical interference effect. Alternatively, the two layers can
be formed of
different materials, with the outer (top) of the two layers being formed of an
antimicrobial, atomically disordered, nanocrystalline silver-containing
material, and the
inner of the two layers having appropriate reflective properties so that the
two layers can
provide an interference effect (e.g., to monitor the thickness of the outer
(top) of the two
layers).
The substrate can be selected as desired. The substrate may be formed of one
layer or multiple layers, which may be formed of the same or different
materials.
In certain embodiments, the substrate can include one or more layers
containing a
bioabsorbable material. Bioabsorbable materials are disclosed, for example, in
U.S.
Patent No. 5,423,859. In general, bioabsorbable materials can include natural
~5 bioabsorbable polymers, biosynethetic bioabsorbable polymers and synthetic
bioabsorbable polymers. Examples of synthetic bioabsorbable polymers include
polyesters and polylactones (e.g., polymers of polyglycolic acid, polymers of
glycolide,
polymers of lactic acid, polymers of lactide, polymers of dioxanone, polymers
of
trimethylene carbonate, polyanhydrides, polyesteramides, polyortheoesters,
2o polyphosphazenes, and copolymers of the foregoing). Examples of natural
bioabsorbable
polymers include proteins (e.g., albumin, fibrin, collagen, elastin),
polysaccharides (e.g.,
chitosan, alginates, hyaluronic acid). Examples of biosynthetic polymers
include
polyesters (e.g., 3-hydroxybutyrate polymers).
In some embodiments, the substrate includes multiple layers (e.g., two layers,
25 three layers, four layers, five layers, six layers, seven layers, eight
layers, nine layers, 10
layers). The layers can be laminated together (e.g., by thermal fusing,
stitching and/or
ultrasonic welding).
One or more layers (e.g., an outer layer) of a mufti-layer substrate can be
formed
of a perforated (and optionally non-adherent) material (e.g., a woven material
or a non-
3o woven material) that can allow fluid to penetrate or diffuse therethrough.
Such materials
include, for example, cotton, gauze, polymeric nets (e.g., polyethylene nets,
nylon nets,
37



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
polypropylene nets, polyester nets, polyurethane nets, polybutadiene nets),
polymeric
meshes (e.g., polyethylene meshes, nylon meshes, polypropylene meshes,
polyester
meshes, polyurethane meshes, polybutadiene meshes) and foams (e.g., an open
cell
polyurethane foam). Examples of commercially available materials include
DELNETTM
P530 non-woven polyethylene veil (Applied Extrusion Technologies, Inc.,
Middletown,
DE), Exu-Dry CONFORMANT2TM non-woven polyethylene veil (Frass Survival
Systems, Inc., NY, NY), CARELLETM material (Carolina Formed Fabrics Corp.),
NYLON90TM material (Carolina Formed Fabrics Corp.), N-TERFACETM material
(Winfield Laboratories, Inc., Richardson, TX), HYPOLTM hydrophilic
polyurethane foam
(W.R. Grace & Co., NY, NY).
One or more layers (e.g., an inner layer) of a multi-layer substrate can be
formed
of an absorbent material (e.g., a woven material or a non-woven material)
formed of, for
example, rayon, polyester, a rayon/polyester blend, polyester/cotton, cotton
and/or
cellulosic fibers. Examples include creped cellulose wadding, air felt, air
laid pulp fibers
~ 5 and gauze. An example of a commercially available material is SONATRATM
8411
70/30 rayoupolyester blend (Dupont Canada, Mississauga, Ontario).
One or more layers (e.g., an outer layer) of a mufti-layer substrate can be
formed
of an occlusive or semi-occlusive material, such as an adhesive tape or
polyurethane film
(e.g., to secure the device to the skin and/or to retain moisture).
2o In some embodiments, the layers in a mufti-layer substrate are laminated
together
(e.g., at intermittent spaced locations) by ultrasonic welds. Typically, heat
(e.g.,
generated ultrasonically) and pressure are applied to either side of the
substrate at
localized spots through an ultrasonic horn so as to cause flowing of at least
one of the
plastic materials in the first and second layers and the subsequent bonding
together of the
25 layers on cooling. The welds can be formed as localized spots (e.g.,
circular spots). The
spots can have a diameter of about 0.5 centimeter or less.
The shape of the substrate can generally be varied as desired. For example,
the
substrate can be in the shape of a film, a fiber or a powder.
The substrate/coating article can be used in a variety of articles. For
example, the
3o article can be in the shape of a medical device. Exemplary medical devices
include
wound closure devices (e.g., sutures, staples, adhesives), tissue repair
devices (e.g.,
38



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
meshes, such as meshes for hernia repair), prosthetic devices (e.g., internal
bone fixation
devices, physical barriers for guided bone regeneration, stems, valves,
electrodes), tissue
engineering devices (e.g., for use with a blood vessel, skin, a bone,
cartilage, a liver),
controlled drug delivery systems (e.g., microcapsules, ion-exchange resins)
and wound
coverings and/or fillers (e.g., alginate dressings, chitosan powders). In some
embodiments, the article is a transcutaneous medical device (e.g., a catheter,
a pin, an
implant), which can include the substrate/coating supported on, for example, a
solid
material (e.g., a metal, an alloy, latex, nylon, silicone, polyester and/or
polyurethane). In
some embodiments, the article is in the form of a patch (e.g., a patch having
an adhesive
layer for adhering to the skin, such as a transdermal patch).
Subsequent to deposition, the material can optionally be annealed. In general,
the
amleal is conducted under conditions to increase the stability (e.g., shelf
life) of the
material while maintaining the desired therapeutic activity of the material.
In certain
embodiments, the material can be annealed at a temperature of about
200°C or less (e.g.,
~5 about room temperature).
The substrate/coating is typically sterilized prior to use (e.g., without
applying
sufficient thermal energy to anneal out the atomic disorder). The energy used
for
sterilization can be, for example, gamma radiation or electron beam radiation.
In some
embodiments, ethylene oxide sterilization techniques are used to sterilize the
2o substrate/coating.
Free Standing Powders
A free standing powder can be prepared by, for example, cold working or
compressing to impart atomic disorder to the powder. In certain embodiments, a
free
25 standing powder is prepared by forming a coating of the material as
described above, and
then removing the material from the surface of the substrate. For example, the
material
can be scraped from the surface of the substrate by one or more scrapers. In
embodiments in which the substrate moves during deposition of the material,
the scrapers
can remove the material as the substrate moves. The scrapers can be, for
example,
3o suspended above the substrate. Such scrapers can be, for example, weighted
and/or
spring loaded to apply pressure sufficient to remove the material as the
substrate moves.
39



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
In some embodiments (e.g., when a continuous belt is used), the scrapers can
be located
above the end rollers to remove the material with a reverse dragging action as
the
substrate rounds the end roller.
A free standing powder can be used to treat a condition in various ways. As an
example, the powder can sprinkled onto the subject's skin. As another example,
the
powder can be inhaled using an inhaler, such as a dry powder inhaler. In some
embodiments, a dry powder can be in the form of an aerosol, which contains,
for
example, at least about 10 (e.g., at least about 20, at least about 30) weight
percent and/or
at most about 99 (e.g., at most about 90, at most about 80, at most about 70,
at most about
60, at most about 50) weight percent of the dry powder.
In certain embodiments (e.g., when the free standing powder is inhaled), the
average particle size of the free standing powder is selected to reduce the
likelihood of
adverse reactions) of the particles in the tissue and/or to deposit the powder
onto specific
anatomical locations (e.g., tissue contacted by the free standing powder
during
~5 inhalation). In some embodiments, the average particle size is selected
(e.g., less than
about 10 microns) so that a relatively small amount of the particles get into
the lower
respiratory tract. In embodiments, a free standing powder can have an average
particle
size of less than about 10 microns (e.g., less than about eight microns, less
than about five
microns, less than about two microns, less than about one micron, less than
about 0.5
2o micron) and/or at least about 0.01 micron (e.g., at least about 0.1 micron,
at least about
0.5 micron).
Powder Impregnated materials
The metal-containing material can be in the form of a powder impregnated
25 material. Such powder impregnated materials can, for example, be in the
form of a
hydrocolloid having the free standing powder blended therein. A powder
impregnated
material can be, for example, in the form of a dressing, such as a
hydrocolloid dressing.
Solutions
3o The material can be in the form of a solution (e.g., a solvent-based
solution). The
solution can be formed, for example, by dissolving a free standing powder of
the material



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
in a solvent for the powder. As an example, a container (e.g., a tea bag-type
container)
with the free standing powder within it can be immersed in the water or
solvent. As
another example, a substrate (e.g., in the form of a strip or a bandage)
carrying the
material can be immersed in the solvent. In certain embodiments, it can be
preferable to
form a solution by dissolving a free standing powder of the material in a
solvent because
this can be a relatively convenient approach to forming a solution. A solution
also refers
to a suspension that contains one or more metal-containing materials. As an
example, a
suspension can be formed by dissolving a metal-containing material (e.g., a
nanocrystalline silver-containing material) in a liquid (e.g., water) for a
period of time
(e.g., several days) so that particles of the metal-containing material are
suspended (e.g.,
by Brownian motion) in the liquid. In some embodiments, a suspended particle
of metal-
containing material can have, for example, a diameter of the order of from
about 10
nanometers to about 20 nanometers. A solution also refers to a dispersion that
contains
one or more metal-containing materials.
~5 In certain embodiments, the solution containing the material is contacted
with the
subject relatively soon after formation of the solution. For example, the
solution
containing the material can be contacted with the subject within about one
minute or less
(e.g., within about 30 seconds or less, within about 10 seconds or less) of
forming the
solution containing the material. In some embodiments, a longer period of time
lapses
2o before the solution containing the material is contacted with the subject.
For example a
period of time of at least about 1.5 minutes (e.g., at least about five
minutes, at least about
minutes, at least about 30 minutes, at least about one hour, at least about 10
hours, at
least about a day, at least about a week) lapses between the time the solution
containing
the material is formed and the solution containing the material is contacted
with the
2s subject.
In some embodiments, lowering the pH of the solution (e.g., to less than about
6.5, such as from about 3.5 to about 6.5) can allow for a higher concentration
of the
dissolved material and/or a faster rate of dissolution. The pH of the solution
can be
lowered, for example, by adding acid to the solution (e.g., by adding COZ to
the solution
3o to form carbonic acid).
41



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
A solution containing the material can be contacted with the subject with or
without the use of a device. As an example, a solution containing the material
can be
contacted with the skin, mouth, ears or eyes as a rinse, a bath, a wash, a
gargle, a spray,
and/or drops. As another example, the solution can be injected using a small
needle
injector andlor a needleless injector. As an additional example, a solution
containing the
material can be formed into an aerosol (e.g., an aerosol prepared by a
mechanical mister,
such as a spray bottle or a nebulizer), and the aerosol can be contacted with
the subject
using an appropriate device (e.g., a hand held inhaler, a mechanical mister, a
spray bottle,
a nebulizer, an oxygen tent). As a further example, a solution containing the
material can
be contacted with the second location via a catheter.
In embodiments in which onychomycosis is being treated, the method can include
first hydrating the nail with urea (1-40%) or lactic acid (10-15%), followed
by treatment
with the metal-containing material, which may contain an appropriate solvent
(e.g.,
DMSO) for penetration through the nail. Alternatively or additionally,
onychomycosis
can be treated by injecting (e.g., via a needleless injector and/or a needle)
the metal-
containing material to the affected area.
Typically, the solvent is a relatively hydrophilic solvent. Examples of
solvents
include water, DMSO and alcohols. In certain embodiments, a water-based
solution is a
buffered solution. In some embodiments, a water-based solution contains
carbonated
2o water. In embodiments, more than one solvent can be used.
In some embodiments, the solution can contain about 0.001 weight percent or
more (e.g., about 0.01 weight percent or more, about 0.02 weight percent or
more, about
0.05 weight percent or more, about 0.1 weight percent or more, about 0.2
weight percent
or more, about 0.5 weight percent or more, about one weight percent or more)
of the
material and/or about 10 weight percent or less (e.g., about five weight
percent or less,
about four weight percent or less, about three weight percent or less, about
two weight
percent or less, about one weight percent or less) of the material.
pharmaceutical Carner Compositions
3o The metal-containing material can disposed (e.g., suspended) within a
pharmaceutically acceptable carrier. The formulation can be, for example, a
semi-solid, a
42



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
water-based hydrocolloid, an oil-in-water emulsion, a water-in-oil emulsion, a
non-dried
gel, and/or a dried gel. Typically, when disposed in a pharmaceutically
acceptable carrier,
the metal-containing material is applied to the skin.
Examples of pharmaceutically acceptable carriers include creams, ointments,
gels,
sprays, solutions, drops, powders, lotions, pastes, foams and liposomes.
The formulation can contain about 0.01 weight percent or more (e.g., about 0.1
weight percent or more, about 0.5 weight percent or more, about 0.75 weight
percent or
more, about one weight percent or more, about two weight percent or more,
about five
weight percent or more, about 10 weight percent or more) of the metal-
containing
1o material and/or about 50 weight percent or less (e.g., about 40 weight
percent or less,
about 30 weight percent or less, about 20 weight percent or less, about 20
weight percent
or less, about 15 weight percent or less, about 10 weight percent or less,
about five
weight percent or less) of the metal-containing material.
In certain embodiments, the metal-containing material can be effectively used
in
~ 5 the oral cavity when in the form of an article (e.g., a tape, a pill, a
capsule, a tablet or
lozenge) that is placed within the oral cavity (e.g., so that the subject can
suck on the
tape, pill, capsule, tablet or lozenge). In some embodiments, the article can
be a
sustained release article (e.g., a sustained release capsule) which can allow
the metal-
containing material to be released at a predetermined rate (e.g., a relatively
constant rate).
2o In some embodiments, an article can include a material (e.g., in the form
of a coating
and/or in the form of a matrix material) that allows the article to pass
through certain
portions of the gastrointestinal system with relatively little (e.g., no)
release of the metal-
containing material, but that allows a relatively large amount of the metal-
containing
material to be released in a desired portion of the gastrointestinal system.
As an
25 example" the article can be an enteric article (e.g., an enteric coated
tablet) so that the
article to passes through the stomach with little (e.g., no) metal-containing
material being
released, and so that the metal-containing material is relatively easily
released by the
article in the intestines.
Formulations can optionally include one or more components which can be
3o biologically active or biologically inactive. Examples of such optional
components
include base components (e.g., water and/or an oil, such as liquid paraffin,
vegetable oil,
43



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
peanut oil, castor oil, cocoa butter), thickening agents (aluminum stearate,
hydrogen
lanolin), gelling agents, stabilizing agents, emulsifying agents, dispersing
agents,
suspending agents, thiclcening agents, coloring agents, perfumes, excipients
(starch,
tragacanth, cellulose derivatives, polyethylene glycols, silicones,
bentonites, silicic acid,
talc), foaming agents (e.g., surfactants), surface active agents,
preservatives (e.g., methyl
paraben, propyl paraben) and cytoconductive agents (e.g., betaglucan). In some
embodiments, a formulation includes petrolatum. In certain embodiments, a
pharmaceutical carrier composition can include a constituent (e.g., DMSO) to
assist in
the penetration of skin.
While the foregoing has described embodiments in which a single condition is
treated, in some embodiments multiple conditions can be treated. The multiple
conditions can be the same type of condition (e.g., multiple skin or
integument
conditions) or different types of conditions. For example, a dressing formed
of one or
more substrates coated with an appropriate metal-containing material (e.g.,
antimicrobial,
~ 5 atomically disordered, silver-containing material) can be applied to an
area of the skin
having multiple skin or integument conditions (e.g., a burn and psoriasis) so
that the
metal-containing material treats the multiple skin or integument conditions.
Moreover, while the foregoing has described embodiments that involve one
method of contacting a subj ect with the metal-containing material, in other
embodiments,
2o more than one method of contacting a subject with the metal-containing
material can be
used. For example, the methods can include one or more of ingestion (e.g.,
oral
ingestion), injection (e.g., using a needle, using a needleless injector),
topical
administration, inhalation (e.g., inhalation of a dry powder, inhalation of an
aerosol)
and/or application of a dressing.
25 Furthermore, while the foregoing has described embodiments in which one
form
of the metal-containing material is used, in other embodiments, more than one
form of
the metal-containing material can be used. For example, the methods can
include using
the metal-containing material in the form of a coating (e.g., a dressing), a
free standing
powder, a solution and/or a pharmaceutical carrier composition.
30 ~ Moreover, the metal-containing material can be used in various industrial
applications. For example, the metal-containing material can be used to reduce
and/or
44



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
prevent microbial growth on industrial surfaces (e.g., industrial surfaces
where microbial
growth may occur, such as warm and/or moist surfaces). Examples of industrial
surfaces
include heating pipes and furnace filters. W certain embodiments, the metal-
containing
material can be disposed (e.g., coated or sprayed) on the surface of interest
to reduce
and/or prevent microbial growth. This can be advantageous in preventing the
spread of
microbes via, for example, heating and/or air circulation systems within
buildings.
The following examples are illustrative and not intended as limiting.
Examples
Treatment of Hyperproliferative Skin Conditions
Example 1 - Preparation of Nanocrystalline Silver Coatings on Dressings
This example shows the preparation of a bilayer nanocrystalline silver coating
on
a dressing material. A high density polyethylene dressing, DELNETTM or
CONFORMANT 2TM was coated with a silver base layer and a silver/oxide top
layer to
generate a coloured anti-microbial coating having indicator value. The coating
layers
~ 5 were formed by magnetron sputtering under the conditions set out in the
following table.
Sputterin~LConditions: Base Layer Top La ~~er
Target 99.99% Ag 99.99% Ag
Target Size 20.3 cm diameter 20.3 cm 'diameter
Working Gas 96/4 wt% Ar/OZ 9614 wt% Ar/O2
2o Working Gas Pressure 5.33 Pa (40 mT) 5.33 Pa (40 mT)
Power 0.3 kW 0.15 kW
Substrate Temperature 20 ° C 20 ° C
Base Pressure 3.0 X 10-6 Torr 3.0 X 10-6 Torr
Anode/Cathode Distance 100 mm 100 mm
25 Sputtering Time 7.5 - 9 min 1.5 min
Voltage 369 - 373 V 346 V
The resulting coating was blue in appearance. A fingertip touch was sufficient
to
cause a colour change to yellow. The base layer was about 900 nm thick, while
the top
so layer was 100 nm thick.



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
To establish that silver species were released from the coated dressings, a
zone of
inhibition test was conducted. Mueller Hinton agar was dispensed into Petri
dishes. The
agar plates were allowed to surface dry prior to being inoculated with a lawn
of
Staphylococcus aureus ATCC#25923. The inoculant was prepared from Bactrol
Discs
(Difco, M.), which were reconstituted as per the manufacturer's directions.
Immediately
after inoculation, the coated materials to be tested were placed on the
surface of the agar.
The dishes were incubated for 24 hr. at 37°C. After this incubation
period, the zone of
inhibition was calculated (corrected zone of inhibition = zone of inhibition -
diameter of
the test material in contact with the agar). The results showed a corrected
ZOI of about
10 mm, demonstrating good release of silver species.
The coating was analyzed by nitric acid digestion and atomic absorption
analysis
to contain 0.24 +/- 0.04 mg silver per mg high density polyethylene. The
coating was a
binary alloy of silver (>97%) and oxygen with negligible contaminants, based
on
secondary ion mass spectroscopy. The coating, as viewed by SEM, was highly
porous
~ 5 and consisted of equiaxed nanocrystals organized into coarse columnar
structures with an
average grain size of 10 nm. Silver release studies in water demonstrated that
silver was
released continuously from the coating until an equilibrium concentration of
about 66
mg/L was reached (determined by atomic absorption), a level that is 50 to 100
times
V
higher than is expected from bulk silver metal (solubility <_ lmg/L).
2o By varying the coating conditions for the top layer to lengthen the
sputtering time
to 2 min, 15 sec., a yellow coating was produced. The top layer had a
thickness of about
140 nm and went through a colour change to purple with a fingertip touch.
Similarly, a
purple coating was produced by shortening the sputtering time to 1 min, to
achieve a top
layer thickness of about 65 nm. A fingertip touch caused a colour change to
yellow.
25 To form a three layer dressing, two layers of this coated dressing material
were
placed above and below an absorbent core material formed from needle punched
rayon/polyester (SONTARATM 8411). With the silver coating on both the first
and third
layers, the dressing may be used with either the blue coating side or the
silver side in the
skin facing position. For indicator value, it might be preferable to have the
blue coating
3o visible. The three layers were laminated together by ultasonic welding to
produce welds
between all three layers spaced at about 2.5 cm intervals across the dressing.
This
46



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
allowed the dressing to be cut down to about 2.5 cm size portions for smaller
dressing
needs while still providing at least one weld in the dressing portion.
The coated dressings were sterilized using gamma radiation and a sterilization
dose of 25 kGy. The finished dressing was packaged individually in sealed
polyester
peelable pouches, and has shown a shelf life greater than 1 year in this form.
The coated
dressings can be cut in ready to use sizes, such as 5.1 x 10.2 cm strips, and
slits formed
therein before packaging. Alternatively, the dressings may be packaged with
instructions
for the clinician to cut the dressing to size and form the desired length of
the slit for the
medical device.
Additional silver coated dressings were prepared in a full scale roll coater
under
conditions to provide coatings having the same properties set out above, as
follows:
~ the dressing material included a first layer of silver coated DELNET, as set
out
above, laminated to STRATEX, AET, g.ONP2-A/QW, which is a layer of 100%
rayon on a polyurethane film.
~ Silver Foam Dressing - three layers of silver coated high density
polyethylene
prepared as above, alternating with two layers of polyurethane foam, L-00562-6
Medical Foam, available from Rynel Ltd., Bootbay, Maine, USA.
Exam~,le 2 - Preparation of Nanocrystalline Silver Powders
~'~ ~~ Nanocrystalline silver powde~:wvas prepared by preparing silver
coatings orb;
2o silicon wafers, under the conditions set forth in the table above, and then
scraping the
coating off using a glass blade.
Nanocrystalline silver powder was also prepared by sputtering silver coatings
on
silicon wafers using Westaim Biomedical NGRC unit, and then scraping the
coating off.
The sputtering conditions were as follows:
Target: ~ 99.99% Ag
Target Size: 15.24 cm X 1216.125 cm
Working Gas: 75:25 wt% Ar/Oz
Working Gas Pressure: 40 mTorr
Total Current: 40 A
3o Base Pressure: 5.0 X10-5 Torr
Sandvik Belt Speed: 340 mm/min
47



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Voltage: 370 V
The powder has a particle size ranging from 2 ~,m to 100 ~Cm, with crystallite
size
of 8 to 10 nm, and demonstrated a positive rest potential.
Example 3 - Treatment of Psoriasis
This patient was a 58 year old female with psoriatic plaques covering up to
sixty
percent of her body. For this patient, psoriatic plaques first occurred ten
years ago and
have been treated with the following:
1. Adrenal corticosteroids. Injections gave relief from pruritus and general
discomfort. Treatments led to a rebound effect; i.e. psoriasis would flare up
after
treatments wore off. Corticosteroids were discontinued.
2. UV Light and Methotrexate treatments. UV light treatments were given in
conjunction with methotrexate. The UV light treatments caused burns and new
lesions.
The methotrexate caused severe nausea. Both treatments were discontinued.
3. Ice Cap Spray. This treatment contained a potent corticosteroid, and gave
~ 5 some relief but it was taken off the market and is no longer available.
4. Soriatone (acetretin 10 mg). This systemic retinoid treatment was
associated
with joint aches and was discontinued.
S. Diet. The patient was attempting to control the disease through diet.
Nanocrystalline silver was tested as follows. Nanocrystalline silver was
deposited
20 on sheets of high-density polyethylene (HDPE) using a vapour deposition
process as set
forth in
Example 1. Two sheets of this coated HDPE were laminated together around a
core of non-woven rayon polyester, as set forth in Example 1. A 50 rnm X 50 mm
(2" X
2") piece of this composite material was saturated with water and placed
centrally on a
25 one and a half year old 150 mm X 100 inm (6" X 4") psoriatic plaque on the
patient's
flank. The nanocrystalline silver coated material was covered with a piece of
low
moisture vapour transmission thin polymer film. The polymer sheet extended 50
mm
(2") beyond the nanocrystalline silver coated HDPE to provide control data
regarding
occlusion of the psoriatic plaque.
3o The dressing was removed after three days. There was no discernible change
in
the plaque at this time. However two days later the area that was covered with
the
48



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
nanocrystalline silver had the appearance of normal skin while the rest of the
plaque was
still rough and unchanged, including the untreated but occluded area.
The nanocrystalline silver therapy caused the treated psoriatic plaque to
resolve.
Example 4 - Treatment of Psoriasis
The subject was a 5~ year old female with psoriatic plaques over up to sixty
percent of her body. Psoriatic plaques had first occurred 10 years ago and had
been
treated with the following:
1. Adrenal corticosteroids. Injections gave relief from pruritus and general
discomfort. Treatments led to a rebound effect i.e. psoriasis would flare up
after
treatments wore off. Corticosteroids were discontinued.
2. TJV Light and Methotrexate treatments. UV light treatments were given in
conjunction with methotrexate. The W light treatments caused burns and new
lesions.
The methotrexate caused severe nausea. Both treatments were discontinued.
3. Ice Cap Spray. This treatment contained a potent corticosteroid, and gave
~ 5 some relief but it was taken off the market and is no longer available.
4. Soriatone (acetretin 10 mg). This systemic retinoid treatment was
associated with joint aches and was discontinued.
5. Diet. The patient was attempting to control the disease through diet.
Nanocrystalline silver was tested as follows. Nanocrystalline silver was
deposited
20 on sheets of high-density polyethylene (HDPE) using a vapour deposition
process as set
forth in Example 1 (top layer). Two sheets of this coated HDPE were laminated
together
around a core of non-woven rayon polyester, as set forth in Example 1. A 50 mm
X 50
mm (2" X 2") piece of this composite material was saturated with water and
placed
centrally on a 125 mm X 100 mm (5" X 4") psoriatic plaque on the patient's
upper left
25 thigh. The nanocrystalline silver coated material was covered with a piece
of low
moisture vapour transmission thin polymer film. The polymer sheet extended 50
mm
(2") beyond the nanocrystalline silver coated HDPE to provide control data
regarding
occlusion of the psoriatic plaque.
The dressing was removed and the plaque examined after two days. The area that
3o was covered with the nanocrystalline silver was free of scaling and only
slightly
erythematous while the rest of the plaque was still erythenatous and scaly,
including the
49



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
untreated but occluded area. The plaque was redressed with a similar 50 mm X
50 mm
(2" X 2") piece of nanocrystalline silver coated dressing, which was left in
place for a
further period of 2 days. The area that was covered with the nanocrystalline
silver
remained free of scale and only slightly erythenatous, while the rest of the
plaque was
still erythenatous and scaly, including the area under the occlusive film.
The nanocrystalline silver therapy caused the treated psoriatic plaque to
resolve.
Example 5 - Preparation of Nanocrystalline Gels
A commercial carboxymethyl cellulose/pectin (Duoderm ConvatecTM) was
combined with nanocrystalline silver powder prepared as in Example 2 to
produce a gel
1 o with 0.1 % w/v. silver. Carboxymethyl cellulose (CMC) fibers were coated
by magnetron
sputtering, under conditions similar to those set out in Example 1 for the top
layer to
produce a defective nanocrystalline silver coating. The CMC was then gelled in
water by
adding 2.9 g to 100 mL volume. A~1 alginate fibrous substrate was directly
coated with a
defective nanocrystalline silver coating by magnetron sputtering under coating
conditions
~5 similar to those set forth in Example 1 for the top layer. The alginate
(5.7 g) was added
to 100 mL volume of water to create a gel. A commercial gel containing CMC and
alginate (Purilon gel ColoplastTM) was mixed with an atomic disordered
nanocrystalline
silver powder prepared as in Example 2 to give a gel product with 0.1 % w/v
silver. A
commercially available gel (LubridermTM - glyceryl polyrnethacrylate) was
blended with
2o atomic disordered nanocrystalline silver powder prepared as in Example 2,
to prepare a
gel with a silver content of 0.1 % w/v. A further gel was formulated with, on
w/v basis,
0.1 % methyl paraben, 0.02 % propyl paraben, 0.5% polyvinyl alcohol (AirvolTM
PVA
540), 2% CMC, 0.1 % nanocrystalline silver powder prepared as in Example 2,
and was
brought up to 1000 g with water.
Treatment of Inflammatory Skin Conditions
Example 1 - Preparation of Nanocrystalline Silver Coatings on Dressings
This example shows the preparation of a bilayer nanocrystalline silver coating
on
a dressing material. A high density polyethylene dressing, DELNETTM or
3o CONFORMANT 2TM was coated with a silver base layer and a silver/oxide top
layer to
generate a coloured antimicrobial coating having indicator value as described
in Example



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
1 of the Treatment of Hyperproliferative Skin conditions examples. The coating
layers
were formed by magnetron sputtering under the conditions set out in the
following table.
Example 2 - Preparation of Nanocrystalline Silver Coating on HDPE Mesh
The silver coated mesh was produced, as set forth in Example 1, by sputtering
silver onto Delnet, a HDPE mesh (Applied Extrusion Technologies, Inc.,
Middletown,
DE, USA) using Westaim Biomedical TMRC unit under the following conditions:
Target: 99.99% Ag
Target Size: 15.24 cm X 152.4 cm
Working Gas: 99.375:0.625 wt% Ar/02
Working Gas Pressure: 5.33 Pascals (40 mTorr)
Total Current: 22 A
Base Pressure: 5.0 X10-5 Torr
Sandvik Belt Speed: 577 mm/min
Voltage: 367 V
The coating was tested and found to have a weight ratio of reaction product to
silver of between 0.05 and 0.1. The dressing was non-staining to human skin.
Example 3 - Preparation of Atomic Disordered Nanocrystalline Silver Powders
Nanocrystalline silver coatings were prepared by sputtering silver in an
oxygen
2o containing atmosphere directly onto an endless stainless steel belt of a
magnetron
sputtering roll coater, or onto silicon wafers on the belt. The belt did not
need to be
cooled. The coatings were scraped off with the belt with suspended metal
scrapers as the
belt rounded the end rollers. For the coated silicon wafers, the coatings were
scraped off
with a l~nife edge. The sputtering conditions were as follows:
Target: 99.99% Ag
Target Size: 15.24 cm X 1216.125 cm
Working Gas: 75:25 wt% Ar/02
Working Gas Pressure: 5.33 Pascals (40 milliTorr)
3o Total Current: 40 A
51



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Base Pressure: 5.0 X10-5 Torr (range: 1 X 10-4- 9 X 10-~
Torr or 1 X 10-2 - 1.2 X 10-4 Pa)
Sandvik Belt Speed: 340 rmn/min
Voltage: 370 V
Note - pressure conversions to Pa herein may not. be accurate, most accurate
numbers are in torn mTorr units.
The powder had a particle size ranging from 2 ,um to 100 ,um, with grain or
crystallite size of 8 to 10 nm (i.e., nanocrystalline), and demonstrated a
positive rest
potential.
Similar atomic disordered nanocrystalline silver powders were formed as set
forth
hereinabove by magnetron sputtering onto cooled steel collectors, under
conditions
taught in the prior Burrell et al. patents to produce atomic disorder.
Example 4 - In vitro Activity of Silver' Solution against Propionibacterium
acne
An in vitf°o test was conducted to determine if silver solutions
according to the
~ 5 present invention effectively control Pf~opioiaibaete~ium acne. The silver
solution was
obtained by static elution of ActicoatTM Burn Wound Dressing (lot #: 00403A-
O5,
Westaim Biomedical Corp., Fort Saskatchewan, Canada) with nanopure water in a
ratio
of one square inch of dressing in five milliliters of water for 24 hours at
room
temperature. The silver concentration of the silver solution was determined by
an atomic
2o absorption method. The silver elute was diluted with nanopure water to 20
~,g/ml. The
P~opionibactef°ium ache (ATCC No. 0919) was provided by Biofilm
Research Group,
University of Calgary.
The inoculum was prepared by inoculating freshly autoclaved and cooled tubes
of
Tryptic soy broth (TSB) with P. acne and incubating them for 2 days at 37
° C in an
25 anaerobic jar. At this time, the optical density of the suspensions was ~
0.3 at a
wavelength of 625 nm.
The bacterial suspension (100 ,uL) was mixed with 100 ,uL of the silver
solution
being tested. The final concentration of silver in these mixtures was 10
ug/ml. The
mixtures were incubated in an anaerobic jar at 37°C for two hours. The
silver was
so neutralized by addition of 0.4% STS (0.85% NaCI, 0.4% Sodium thioglycolate,
1%
TweenTM 20) and the solution was serially 10-fold diluted with phosphate-
buffered
52



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
saline. 20 ,uL aliquots of the original solution and subsequent dilutions were
plated onto
TSA drop plates. The drops were allowed to dry and the plates were incubated
in an
anaerobic jar at 37°C for 72 hours at which time the colonies were
counted. The control
consisted of 100 ,uL of bacterial suspension mixed with 100 ,uL of nanopure
water and
treated as above.
The results showed that the silver solution according to the present
invention, at a
final concentration of 10 ,ug/ml, gave 4.3 logaritlnn reduction in viable P.
acne counts in
two hours.
Example 5 - Treatment of Acne
A sixteen year old female was diagnosed with acne vulgaris. She had numerous
red papules and pustules on her forehead. Various skin cleansing regimes and
antibiotic
(erythromycin and clindomycin) treatments had been tried and had failed to
control the
acne. Prior to bedtime, the papules and pustules on one side of her forehead
were
moistened and covered with a nanocrystalline silver coated high density
polyethylene
~ 5 mesh, prepared as in Example 1 (single layer, blue coating). The mesh was
then
occluded with a thin film dressing which remained in place for 10 hours. Upon
removal,
the papules and pustules were no longer red and were only slightly raised.
Some brown
staining of the skin was observed.
Example 6 - Treatment of Acne
2o A sixteen year old male was diagnosed with acne vulgaris. He had numerous
raised, red papules and pustules on his forehead. Various skin cleansing
regimes and
antibiotic treatments had been tried and had failed to control the acne. The
patient was
placed on isotretinoin treatment which controlled his acne well. He did
develop a single
large pustule on his forehead which was embarrassing for him. Prior to
bedtime, the
25 pustule was moistened and covered with a nanocrystalline silver coated high
density
polyethylene mesh prepared as in Example 2. The mesh was then occluded with a
thin
film dressing which remained in place for 10 hours. Upon removal the pustule
was no
longer red and was only slightly raised. A second treatment resulted in the
disappearance
of the pustule.
3o Example 7 - Treatment of Acne
53



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
A sixteen year old female was diagnosed with acne vulgaris. She had numerous
red papules and pustules on her forehead. Various skin cleansing regimes and
antibiotic
(erythromycin and clindomycin) treatments had been tried and had failed to
control the
acne. Prior to bedtime, the papules and pustules on one side of her forehead
were
moistened and covered with a nanocrystalline silver coated high density
polyethylene
mesh, prepared as in Example 2. The mesh was then occluded with a thin film
dressing
which remained in place for 10 hours. Upon removal the papules and pustules
were no
longer red and were only slightly raised. A second treatment resulted in the
disappearance of the papules and virtual elimination of the pustules. The
silver coated
mesh, when prepared as set forth in Example 2, did not result in any staining
of the skin.
Example 8 - Treatment of Adult Acne with Silver-Impregnated Hydrocolloid
Dressing
A 49 year old white male experienced occasional acne vulgaris. He had painful,
raised, red papules and pustules on his shoulders. The patient was treated
with a thin
~5 hydrocolloid dressing (Craig Medical Products Ltd., Clay Gate House 46
Albert Rd.
North Reigate, Surrey, United Kingdom) which was impregnated with 1
nanocrystalline silver powder formed with atomic disorder as in Example 3.
Following
cleansing, the pustule was covered with a small disc of the dressing, which
remained in
place for 24 hours. Upon removal, the pustule was no longer painful, red, or
raised.
2o Example 9 - Treatment of Eczema
A twenty-nine year old white female presented with acrodermatitis. The
erythematous area was located on the dorsal surface of the first web space of
the left
hand. It was bounded by the metacarpal bones of the thumb and index forger.
The
patient also complained of pruritus associated with the dermatitis. A gel
consisting of
25 0.1% nanocrystalline silver powder (formed with atomic disorder as in
Example 3) and
2% carboxymethylcellulose was applied to the inflamed area before bedtime.
There was
an immediate antipruritic effect that provided the patient with relief in the
short term.
The next morning all evidence of acrodermatitis (i.e. redness disappeared) was
gone. The
condition had not returned after two weeks.
3o Example 10 - Allergic Contact Dermatitis
54



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Slcin allergic contact hypersensitivity is caused by excessive infiltration of
eosinophils. An animal model may be used for ifz vivo evaluation of eosinophil
infiltration in the contact sensitivity reaction and to determine whether it
is associated
with allergic slcin conditions such as contact dermatitis. On a gross
histology level, this
can be measured by the degree of erythema and edema at the dermatitis site.
Current
drugs used for treatment of this and other related eczema conditions include
high potency
steroids (UltravateTM), medium potency steroids (EloconTM) and non steroidal
anti-
inflammatory compounds (ProtopicTM or tacrolimus). These compounds do not
always
work and may have undesirable side effects. Several commercially available
anti-
inflarmnatory products were compared to a nanocrystalline silver powder for
the
treatment of allergic contact dermatitis as follows.
Four healthy domestic pigs (approximate weight 20 kg) were used in the study.
All pigs had normal skin prior to induction of eczema with 10% 2,4-
dinitrochlorobenzene
(DNCB) in acetone. The animals were housed in appropriate animal facilities
with 12
hour light-dark cycles. The pigs were fed antibiotic-free feed and water ad
libitum. The
pigs were housed and cared for in accordance with Canadian Council of Animal
Care
guidelines. On day 0, the hair on both left and right back and side were
clipped. The
DNCB solution was painted over this area. This was repeated on day 7 and 11.
On day
11, the solution was painted approximately 4 hours before treatment was
initiated.
2o Treatment groups are shown in the following table. ProtopicTM (tacrolimus),
EloconTM and UltravateTM were purchased as creams from the local pharmacy. The
nanocrystalline silver powder (1 g/L) was mixed into a 2% sodium carboxymethyl
cellulose (CMC) and water solution at 30°C using a magnetic stirrer at
a high speed
(Vista Scientific). Petrolatum, commercially known as VaselineTM, was used as
a control
for EloconTM and UltravateTM.
p-i~ # Treatment (Left Side) Control (Ryht Side) Day of
Treatment
1 ProtopicTM (tacrolimus) , ProtopicTM Control Day 0
so 2 Medium Potency Steroid Petrolatum Day 0
(EloconTM)



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
3 2% CMC + 1% 2% CMC Day 0
nanocrystalline silver
(Vista Scientific)
4 High Potency Steroid Petrolatum Day 0
(UltravateTM)
Pigs were placed under general anesthetic with ketamine (KetaleanTM, MTC
Pharmaceuticals, Cambridge, ON; 4-500 mg) and halothane (MTC Pharmaceuticals).
The skin was wiped with a moist gauze and allowed to dry. Bandages (n = 8)
containing
each treatment were applied to the left side of the thoracolumbar area of the
pig, while
control bandages (n = 8) were applied to the right side of the thoracolumbar
area of the
pig. Following placement of bandages, they were covered with TegadennTM (3M
Corp.,
Minneapolis, MN) which was secured with an ElastoplastTM (Smith and Nephew,
Lachine, QC) wrap. Bandages with active agents were changed daily. The skin
~ 5 associated with each bandage site was scored for severity of erythema (0=
normal, 1=
slight, 2= moderate, 3= severe, 4= very severe) and swelling (0= normal, 1=
slight, 2=
moderate, 3= severe, 4= very severe). This was performed on days 0, 1, 2 and
3.
All pigs remained healthy during the study. Results are shown in the following
tables, and indicated in Figs. 3 and 4. Figs. 3 and 4 show the efficacy of the
2o nanocrystalline silver powder compared to ProtopicTM, EloconTM and
UltravateTM in the
treatment of contact dermatitis in the pig model.
Treatment (Erythema D_a~0 Day_1 Day 2 Dad
l


Nanocrystalline silver3 2 1 0


25 ProtopicTM 3 3 1.9 0.4


EloconTM 3 2.4 2.6 2.6


UltravateTM 3 3 3 3


Treatment (Edema) Day 0 Day 1 Day 2 Day
3


so Nanocrystalline 2 1 0 0
silver


ProtopicTM 2 2 2 0


56



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
EloconTM 2 2 2 0
UltravateTM 2 2 1 0
The pigs treated with the high (UltravateTM) and medium (EloconTM) strength
steroids showed little to no improvement in the degree of erythema associated
with
contact dermatitis. They did, however, improve in terms of edema in that at
Day 3, no
swelling was apparent. ProtopicTM showed a marked improvement when compared to
the
steroids in both the degree of erythema and edema. The largest improvement
occurred
with the nanocrystalline silver powder suspended in a 2% carboxymethyl
cellulose gel.
Both erythema and edema scores were lower after a single treatment and were
normal
after Day 2 (edema) and Day 3 (erythema) of treatment. Clearly the
nanocrystalline
silver product was more efficacious in treating contact dermatitis than the
commercially
available products.
Example 11 - Preparation of Gels
No. 1
~5 A commercial carboxymethyl cellulose/pectin gel (DuoDERMTM, ConvaTec
Canada, 555, Dr. Frederik Philips, Suite 110, St-Laurent, Quebec, H4M 2X4) was
combined with nanocrystalline silver powder prepared as set forth in Example 3
to
produce a gel with 0.1% silver. A logarithmic reduction test was performed as
follows in
the gel using Pseudomonas aeYUginosa. The inoculum was prepared by placing 1
2o bacteriologic loopful of the organism in 5 mL of trypticase soy broth and
incubating it for
3-4 h. The inoculum (0.1 mL) was then added to 0.1 mL of gel and vortexed
(triplicate
samples). The mixture was incubated for one-half hour. Then 1.8 mL of sodium
thioglycollate-saline (STS) solution was added to the test tube and vortexed.
Serial
dilutions were prepared on 10-1 to 10-x. A 0.1 mL aliquot of each dilution was
plated in
25 duplicate into Petri plates containing Mueller-Hinton agar. The plates were
incubated for
48 h and then colonies were counted. Surviving members of organisms were
determined
and the logarithmic reduction compared to the initial inoculum was calculated.
The
logarithmic reduction for this mixture was 6.2, indicating a significant
bactericidal effect.
No. 2
3o Carboxymethyl cellulose (CMC) fibers were coated directly to produce an
atomic
disordered nanocrystalline silver coating, using magnetron sputtering
conditions similar
57



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
to those set forth in Example 1. The CMC was then gelled in water by adding
2.9 g to
100 mL volume. Tlus material was tested using the method of No. 1. The
material
generated a 5.2 logarithmic reduction of Pseudomoraas ae~ugifaosa ,
demonstrating that
the gel had a significant bactericidal effect.
s No.3
An alginate fibrous substrate was directly coated with an atomic disordered
nanocrystalline silver coating using magnetron sputtering conditions similar
to those set
forth in Example 1. The alginate (5.7 g) was added to 100 mL volume of water
to create
a gel. This material was tested using the method of No. 1. The material
generated a 5.2
logarithmic reduction of Pseudomon.as aer~ugi~2osa, demonstrating that the gel
had a
significant bactericidal effect.
No. 4
A commercial gel containing CMC and alginate (Purilin gel, Coloplast) was
mixed with a atomic disordered nanocrystalline silver powder to give a product
with
~ 5 0.1 % silver. This was tested as above with both Pseudomonas aeruginosa
and
Staphylococcus auf°eus. Zone of inhibition data was also generated for
this gel as
follows. An inoculum (Pseudomonas aerugiyaosa and Staphylococcus au~eus) was
prepared as in No. 1 and 0.1 mL of this was spread onto the surface of Mueller-
Hinton
agar in a Petri dish. A six mm hole was then cut into the agar at the center
of the Petri
2o dish and removed. The well was filled with either 0.1 mL of the silver
containing gel, a
mupirocin containing cream or a mupirocin containing ointment. The Petri
plates were
then incubated for 24 h and the diameter of the zone of inhibition was
measured and
recorded.
The silver containing gel produced 9 mm zone of inhibition against both
2s Pseudomo~ras ae~ugifaosa and Staphylococcus auf°eus, while the
mupirocin cream and
ointment produced 42 and 48 mm zones against Staphylococcus aureus and 0 mm
zones
against Pseudomonas aeYUgifaosa.
The silver containing gel reduced the Pseudoy~aofzas aer~uginosa and
Staphylococcus aureus properties by 4.4 and 0.6 log reductions, respectively,
showing
3o good bactericidal activity. The mupirocin cream and ointment generated 0.4
and 0.8, and
0.8 and 1.6, log reductions against Staphylococcus aur~eus afad Pseudomoraas
ae~uginosa,
58



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
respectively. The silver gel had both a greater bactericidal effect and
spectrum of activity
than the mupirocin containing products.
Nos. 5-10
The formula for Nos. 5-10 are summarized in the following table. Zones of
inhibitions were determined as in No. 4 and log reductions were determined as
in No. 1.
All formulae provided a broader spectrum of activity and a greater
bactericidal
effect than did mupirocin in a cream or ointment form. The mupirocin cream
produced
zones of inhibition of 42 and 0, and log reduction of 0.4 and 0.8, against
Staphylococcus
auf°eus and Pseudomonas aeruginosa, respectively.
# CMC PVA Ag Beta- Methyl Propyl CZOI CZOI Log Log
(%) (%) Powder glucanparabenparaben red'nred'n
(%) S. P. S. P.
aureusaeruginosaaureusaeruginosa


5 2 0.1 11 13 1.4 >6


6 2 0.5 0.1 0.1 0.02 14 15 3.3 >6


7 2 0.5 0.1 13 14 2 N/A


8 2 0.5 0.1 0.1 14 14 2 N/A


g 2 0.5 0.1 0.20 14 14 2 N/A


10 2 0.5 0.1 0.5 0.1 0.20 14 14 2 >6


No. 11
A commercially available gel (glyceryl polymethacrylate) was blended with
nanocrystalline silver powder to produce a gel with a silver content of 0.1%.
This gel
was tested as in Nos. 5-10 and was found to produce zones of 15 mm against
both
Staphylococcus aureus and Pseudorraonas aeruginosa. Log reductions of 1.7 and
>5
were produced against Staphylococcus aureus and Pseudomoyt.as aeruginosa. This
gel
product had a greater spectrum of activity than did mupirocin cream or
ointment.
Examt~le 12 - Treatment of Adult Acne with Nanocrystalline Silver Gel Occluded
2o by a Hydrocolloid Dressing
59



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
A 49 year old white male experienced occasional acne vulgaris. He had painful,
raised, red papules and pustules on his shoulders. The patient was treated
with gel
formulation No. S as set forth in Example 11. Gel formulation No. 5 was
applied to the
problem area of the patient's shoulders and then occluded by a thin
hydrocolloid dressing
(Craig Medical Products Ltd., Clay Gate House 46 Albert Rd. North Reigate,
Surrey,
United Kingdom). The dressing remained in place for 24 hours. Upon removal the
pustule was no longer painful, red, or raised.
Treatment of Mucosal or Serosal Conditions
Example 1- Preparation of Nanocrystalline Silver Coatings on Dressings
This example shows the preparation of a bilayer nanocrystalline silver coating
on
a dressing material. A high density polyethylene dressing, DELNETTM. or
CONFORMANT 2TM was coated with a silver base layer and a silver/oxide top
layer to
generate a coloured antimicrobial coating having indicator value as described
in Example
~ 5 1 of the Treatment of Hyperproliferative Skin conditions examples. The
coating layers
were formed by magnetron sputtering under the conditions set out in the
following table.
Example 2 - Preparation of Atomic Disordered Nanocrystalline Silver Powders
Atomically disordered, nanocrystalline silver powder was prepared as described
in Example 3 in the Treatment of W flammatory Skin conditions examples above.
2o Example 3
Silver solutions were prepared by immersing AgHDPE mesh from dressings
prepared as in Example 1 in reverse osmosis water that had been pretreated
with C02 in
order to reduce the pH. Two different concentrations of silver solutions were
prepared by
this method, the concentrations being 85 ~,g/ml, and 318 ~,g/ml. Solutions of
silver
25 nitrate were also prepared to use as comparisons in the experiments. The
concentrations
of the silver nitrate were 103 ppm of silver and 295 ppm of silver as
determined by
Atomic Absorption Spectroscopy.
The solutions were in turn placed in an ultrasonic nebulizer that created
small
droplets containing dissolved and suspended parts of the silver solution. The
output from
3o the nebulizer was directed into a chamber made from a stainless steel frame
and base.
Petri dishes containing Mueller Hinton agar streaked with 4 h old cultures of



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Pseudomonas aef°ugiraosa and Staphylococcus aureus, were exposed to the
silver solution
aerosols and the silver nitrate aerosols.
The results of the tests show that silver aerosols of this invention transmit
the
antimicrobial activity of the dressings to remote sites, and such aerosols are
more
effective than comparable concentrations of silver nitrate.
In many instances the delivery of antimicrobial materials may most
expeditiously
be accomplished by using aerosols (e.g. treatment of pneumonia). The drawback
of
aerosols is the requirement for a high concentration of the active ingredient
to ensure that
a sufficient amount is delivered to achieve the biological effect desired
without causing
problems with the carrier solvent (e.g. water). It is preferably that the
equipment for
producing an aerosol that contains the dissolved and suspended components of
nanocrystalline silver form droplets of aerosol direct from the liquid form,
and the aerosol
droplets must be small enough to reach the lungs. This means the droplets
sh~uld be less
than approximately 10 ~.m. To meet these requirements the aerosol is not
created by first
~ 5 evaporating the liquid then condensing it to form droplets. Rather,
aerosols are generated
by 1) mechanical disruption of the liquid, or 2) air under pressure passing
through some
form of orifice that combines the air and the liquid in a way that creates
droplets instead
of evaporating the liquid.
Several experiments were carried out with silver solutions of this invention
and
2o silver nitrate solutions to determine if the antimicrobial activity of the
dressing could be
transferred through a direct droplet aerosol to a Petri dish.
a) Methods
i) Equipment
The method used for the current tests was the mechanical method in the form of
25 an ultrasonic nebulizer. For convenience an ultrasonic humidifier was used.
The liquid
containing the dissolved and suspended silver from the dressing of Example 1
was placed
in the water reservoir of the humidifier. When power was applied to the
humidifier
aerosol droplets of dissolved and suspended silver were generated and flowed
from the
output nozzle.
3o A test chamber was c~nstructed using a stainless steel frame with a
transparent
plastic covering. The frame was placed on a stainless steel base plate. The
output nozzle
61



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
from the humidifier was modified so that the aerosol could be directed into
the chamber
at a height of approximately 30 cm from the base. The plates and other test
samples were
placed on the stainless steel plate and exposed to the aerosol for a
prescribed length of
time.
s ii) Solutions
Solution 1 - A silver containing solution was prepared by immersing 518 sq.
inches of the dressing from Example 1 in 1 L of reverse osmosis water, which
was treated
with COZ to maintain a pH of 6.5. After 20 minutes the concentration of silver
in the
water was 85 ,ug/ml.
Solution 2 - A solution containing 370 ~,g/ml of silver from a dressing from
Example 1 was prepared as follows: 1 L of reverse osmosis water was purged
with
corninercial grade carbon dioxide until the pH was 4.3.
Sufficient dressing was added to bring the pH up to 6.5. At that time, the
silver
concentration was 370 ~.g/ml.
15 Solution 3 - Ag as AgN03 was prepared by dissolving 0.157 g of AgNO3 into 1
L
of reverse osmosis water and mixed until dissolved. The solution was analyzed
by
Atomic Absorption Spectroscopy and found to be 102.9 ppm of silver.
Solution 4 - Ag as AgNO3 was prepared by dissolving 0.427 g of AgN03 into 1 L
of reverse osmosis water and mixed until dissolved. The solution was analyzed
by
2o Atomic Absorption Spectroscopy and found to be 295 ppm of silver.
iii) Aerosolization
Petri dishes, containing Mueller Hinton agar, were streaked with 4 h old
cultures
of Pseudo~zon.as aerugiraosa or Staphylococcus aur~eus. The plates were then
weighed
and their exposed outer surfaces were coated with Parafilm to prevent
condensation from
2s occurring on these surfaces. These plates were placed in the aerosol
chamber uncovered.
The ultrasonic nebulizer was then started and run for 53 minutes. The plates
were then
removed from the chamber, the plastic was removed and the dishes re-weighed so
that
the amount of moisture loss/gain could be determined.
The plates were then placed in a 35°C incubator for 16 h. After
incubation the
so pattern and amount of growth was assessed on the plates for both organisms.
iv) Viability Assessment
62



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Three of the six plates made for each organism were tested to determine if the
antimicrobial effect was cidal or static in nature. This was accomplished by
rinsing or
placing a piece of the clear section of agar in the Petri dish plates into
Tryptic soy broth
in a test tube and incubating for 4 h or 16 h. If the medium turned turbid in
4 h it would
indicate that the antimicrobial affect was bacteriostatic in nature. If the
organisms took
more than 16 h to grow, as indicated by turbidity, it was considered an
indication that
both static and cidal effects occurred. If no growth occurred the effect was
bactericidal.
v) Results - The results are summarized in the following table.
Solutions 1 and 3
Silver from AgN03
Dressing


Organism P. aeYUginosaS. aureus P. aerugiyaosaS. au~eus


Ag concentration 85 85 99 99


(,ug/ml)


About 6.5 about 6.5


pH of test solution6.5 6.5


53 53 53


Exposure Time 53


(minutes)


Exposed are (sq. 9.8 9.8 9.8 9.8
in.)


Exp 0.8 0.8 1:05 1.05


Weight Gain (g)


0 0 0 0


Growth at 16 h 0 ++ 0 ++++


(0-++++) at 48
h


No No No


Viable 4 h No No No N/A


16h No


63



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Solutions 2 and 4
Silver from AgN03
Dressing


Organism P. aeruginosaS. aur~eusP. aeruginosaS. aureus


370 370 300 300


Ag concentration


(~,g/ml)


About 6.3 about 6.3


pH of test solution6.5 6.5


53 53 53


Exposure Time 53


(minutes)


Exposed are (sq. 9.8 9.8 9.8 9.8
in.)


Exp 1.14 1.14 1.12 1.12


Weight Gain (g)


0 0 0 0


Growth at 16 h 0 0 0 +++


(p-++++) at 48
h


No No No


Viable 4 h No No No N/A


16h No


vi) Discussion
At the low concentration of silver in solution, the dressing generated silver
was
effective in controlling the growth of both organisms while the silver nitrate
only
prevented the growth of P. aerugirzosa. Viability tests showed that at the low
concentration, neither form of silver was completely bacteriocidal although
the poor
growth on the dressing aerosol treated plates compared to the silver nitrate
treated plates
suggests that a significant log reduction occurred in the dressing aerosol
treated plates.
64



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
At a higher concentration of silver, both dressing generated silver (370
~.g/ml) and
AgN03 (300 ~,g/ml) were effective at controlling P. aeruginosa. Since no re-
growth
occurred, it is assumed that the agent at this concentration were
bactericidal. Silver
generated from the dressing was more effective than AgN03 at controlling S.
aureus. No
re-growth occurred on any plates or in the broth indicating a total kill of
the organism
while in the AgN03 treatment, a large number of organisms grew at 16 h.
Based on weight gain during aerosol treatments a dose per unit area can be
calculated. In each case for solution 1 the dose was 8.5 ~.g/sq. inch while
for solution 2
the dose was 38 ,ug/sq. inch. These doses, on a per lung basis, would be less
than 10 mg
of silver per hour of treatment. Each hour of treatment with dressing
generated silver
aerosols appears to provide at least 48 h of protection. Therefore the dose
per day, from
the high concentration treatment, would be about 5 mg or a little less than
the silver
released by 2 sq. inches of SSD per day.
A most significant advantage of using dressing generated silver may be the
lack
~ 5 of a toxic cation such as N03 or sulfadiazine.
Overall, the example demonstrated that the dressing generated aerosols axe
operative to transmit the antimicrobial activity of the dressings to remote
sites.
Furthermore, the dressing generated aerosols were more effective than
comparable
concentrations of silver nitrate.
2o Example 4 - Aerosolized Silver Solutions in Rats
a) Materials And Methods
i) Solutions From Atomically Disordered Silver Dressings
A solution was prepared by sparging C02 through 400 ml of reverse osmosis
water for 30 minutes at a flow rate of 1 L/min. The beaker of water was
covered with a
25 piece of parafilm to assist in maintaining a saturated C02 environment.
This process
resulted in the pH of the water dropping to about 4. At this point,
approximately 600
square inches of silver-coated net (AgHDPE) prepared as in Example 1 was added
to the
water and stirred for approximately 40 minutes resulting in an elevation of
the pH to
approximately 6.5. The solution was then transferred to a medical nebulizer
and
3o connected to an oxygen cylinder with a flow rate of 10 L/min. The outflow
of the
nebulizer was connected to a sealed animal chamber housing the rats to be
dosed. Only



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
the "test" rats (15 randomly assigned animals) received the dosing. The rats
received two,
one-hour aerosol administrations of the solution on the day of infection.
Thereafter, the
test rats were dosed three times per day for an additional three days.
ii) Animals
Thirty male Sprague-Dawley rats were obtained from the University of Calgary,
Alberta, Canada breeding colony. These animals were specific-pathogen free and
weighed approximately 300 g. The animals were housed in groups of 5 in plastic
cages
with wire mesh tops. The rats had access to fresh water and rat chow ad
libitum. All
animals were maintained in an appropriate facility with 12-hour light/dark
cycles and
constant temperature and humidity, according to facility standard operating
procedures.
The protocol was approved by the University of Calgary Animal Care Committee
and
was conducted in accordance with guidelines established by the Canadian
Council on
Animal Care.
iii) Bacteria
~ 5 The bacteria used for infection of these animals were Pseudomonas
aef~uginosa
strain 579. The dose was previously titrated to ascertain that a dose of up to
101° CFU
was not lethal for the animals. The bacteria were grown overnight in Tryptic
soy broth,
washed once in sterile PBS, and re-suspended in a 1/10 volume of sterile PBS.
iv) W fection
2o The rats were anesthetized by inhalation of 2% halothane. A 50 microliter
volume of bacterial suspension was intratracheally administered into the
bronchi of each
rat. This was performed non-surgically on intubated animals. The infection
process
resulted in the instillation of approximately 2x109 CFU into the lungs of each
animal.
v) Sampling
25 On each day, a number of animals were sacrificed. The lungs of the animals
were
aseptically removed, homogenized, and plated to determine bacterial levels. A
few
animals were also subjected to bronchoalveolar lavage prior to removal of the
lungs. In
several cases, lung homogenates and/or lavage fluids were reserved for silver
analysis.
After the first batch of the silver solution was prepared, total silver
analysis
3o indicated that there was about 225 ppm of total silver in the solution. The
solution was
reserved for several hours until after next dosing of the animals. A second
silver analysis
66



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
indicated that the total silver in solution had dropped to about 166 ppm. The
reason for
the drop was immediately apparent as the silver had visibly precipitated out
of solution
and had deposited on the surface of the nebulizer. One other batch of freshly
prepared
solution had a total silver concentration of 337 ppm. Regardless of the actual
numbers,
the process of generating the silver solution results in a significant
quantity of silver in
the solution that is aerosolized into the dosing chamber.
The dosing chamber is not perfect. Although significant amounts of mist are
generated into the chamber, the rates tend to lie on top, of one another and
are probably
exposed to vastly different levels of the silver mist.
1 o vi) Results
i) Pulmonary Bacterial Levels
Day Log CFU/Test Lung Log CFU/Control Lung
1 6.2 7.3
4.1 7.8
3 p 6.2
3.5 4.8
The bacteriological results gathered from the lungs of the treated and control
animals demonstrated a sharper decline in the numbers of bacteria present in
the lungs
~ 5 following treatment with silver mist as compared to controls. The results
indicated that,
in spite of the small sample sizes and inconsistent exposures, a difference
could still be
noted. There was considerable variation in the numbers of bacteria recovered
from
individual animals within each treatment group and, therefore, there was no
significant
difference in the results. Gross examination of excised lungs suggested that
there may
2o have been less apparent damage to the lungs in the animals treated with the
silver mist as
compared to the untreated, infected animal. This was very encouraging given
the
potential anti-inflammatory effects of the nanocrystalline silver technology.
ii) Pulmonary Silver Levels
Rat Description Total Silver Level Average
Sacrifice Date
67



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
36999 Silver mist 1 O.SOppm


36999 Silver mist 2 1.13 ppm 0.74ppm


36999 Silver mist 3 0.58 ppm


37000 Silver mist 4 0.73 ppm


37000 Silver mist 5 0.70 ppm 0.72 ppm


37000 Control 1 0.08 ppm


37000 Control 2 0.10 ppm 0.09 ppm


The results of the silver analysis appear to indicate that the amount of
silver in the
lung either plateaus or each dose of silver mist deposits a certain amount of
silver within
the lung and this level is significantly diminished prior to the next dosing
of the animals.
The results of this experiment indicated that the method employed to prepare
the
silver mist solution was reasonably reproducible and yielded relatively high
concentrations of silver in solution. However, the silver was prone to
precipitation and
should be fleshly prepared prior to each dosing period. A lengthy period
between
preparation and dosing, although resulting in a decrease in the amount of
silver in
solution, did not result in a complete elimination of the silver from the
solution or even
result in the silver concentration dropping to very low levels.
The method employed for exposing the rats to the mist is also prone to
significant
variation due to the piling up of the rats and the resultant inconsistent
exposure to the
silver-containing mist. However, the silver analyses suggested that a
reasonably uniform
~ 5 dose of silver was achieved when only a few animals were present within
the dosing
chamber.
Regardless of the difficulties associated with the experiment, the results
were
indicative of a therapeutic modality for pulmonary infections. The results
showed that
the presence of silver mist was effective in more rapidly clearing the
bacterial load of the
2o infected lungs than is the host immune system alone. The apparently less
severe
pathology associated with the rat lungs treated with the silver mist showed
that the
treatment was effective for more than simply assisting in the killing of
invading
organisms.
68



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Example 5- Pulmonary Anti-inflammatory Activity
A solution form nanocrystalline silver coated dressings (AgHDPE) from Example
1 was prepared by sparging C02 through 1000 ml of reverse osmosis water using
commercial COa Soda Syphon Charger. This process resulted in the pH of the
water
dropping to about 4. At this point, approximately 333 m1 of the carbonated
water was
decanted into a plastic bottle and 333 square inches of nanocrystalline silver-
coated net
was added to the water. The nanocrystalline silver-coated net and water were
placed in
37°C shaker incubator and shaken at 180 RPM for 30 minutes to elevate
the pH to
approximately 5.8. The solution was then transferred to a beaker and stirred
vigorously
for 2 minutes to raise the pH to approximately at 7.3. The dissolution
solution was
transferred to a commercial nebulizer which was connected to a medical air
cylinder with
a flow rate of approx. 20 L/min. The outflow of the nebulizer was connected to
a animal
chamber housing the rats to be dosed. Only the "test" rats (12 randomly
assigned
animals) received the dosing. The rats received two -1/2. hour aerosol
administrations of
the test solution on the day of infection. Thereafter, the test rats were
dosed 3 times per
day for an additional one and a half days.
Thirty male Spragu-Dawley rats were obtained. These animals are specific-
pathogen free and weighed approximately 400 g. The animals are housed in
groups of
four in plastic cages with wire mesh tops. The rats had access to fresh water
and rat
2o chow ad libitufn. All animals were maintained in an appropriate facility
standard
operating procedures.
The bacteria used for infection of these animals were Pseudo~aonas aerugiraosa
strain 5588. The dose was previously titrated to ascertain that a dose of up
to 109 CFU
was not lethal for the animals. The bacteria were gromn overnight in Tryptic
soy broth,
washed once in sterile PBS and resuspend in sterile PBS. The final
concentration of the
inoculum was 4X 109 CFU/ml.
The rats were anesthetized by inhalation of 2% halothane. A 400 microliter
volume of bacterial suspension was intratracheally administered in the bronchi
of each
rat. This was performed non-surgically on intubated animals. The infection
process
3o resulted in the instillation of approximately 109 CFU into the lungs of
each animal.
The three treatment groups of rats and treatment schedules were as follows:
69



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Group 1 W fected, not treated (12 Rats)
Group 2 Infected, animal will be treated by intramuscularly injection of
Tobramycin at 30mg/kg (l2mg/rat) once daily (12 Rats)
Group 3 Infected and treated, using nanocrystalline silver solution and
nebulizer
(Nebulized Ag), three times a day (12 Rats)
Day One
10:00 AM Infection
4:00 PM First treatment (For Group 2, Nebulized Ag for Group 3)
8:00 PM Nebulized Ag treatment for Group 3
Day Two
9:00 AM Inj ection treatment for Group 2, Nebulized Ag for Group 3
1:00 PM Sacrifice and sample six Rats in each group
3:00 PM Nebulized Ag treatment for Group 3
8:00 PM Nebulized Ag treatment for Group 3
~ 5 Day Three
9:00 AM Inj ection treatment for Group 2, Nebulized Ag for Group 3
1:00 PM Sacrifice and sample six Rats in each group
On each day, six rats of each group of animals were sacrificed. The lungs of
the
animals were aseptically removed, homogenized and plated to determine
bacterial levels.
2o Lung samples were collected for histological examination. Three lung
homogenates were
reserved for silver analysis. Lungs were grossly scored (absent = 0, mild = l,
moderate =
2, and severe = 3) based on the degree and involvement of consolidation,
hemorrhage, -
edema and necrosis based upon gross observation.
Histopathology was scored (0-4) based upon the degree of consolidation and
25 inflammation (neutrophil infiltration). The entire right middle lobes of
all rats were
collected for histopathology. As whole lobes were selected there was no bias
toward any
sample. All samples were fixed in neutral buffered formalin at the time the
lung was
removed from the thorax. It was fixed overnight, dehydrated and embedded in
was.
Sections were obtained which were hydrated and stained with haematoxylin and
eosin.
so All sections were examined by a veterinary pathologist who was blinded to
the
treatment groups, until after the samples were scored and comments were
provided. The
7o



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Scores and comments are provided in Table 5. (0= normal, 1= slight, 2
moderate, 3
severe, 4 very severe).
Tissue Colony Counts:
At 24 hours, there was not a significant reduction in the number of colony
forming
units (cfu) in the nebulized Ag group compared to the control but at 48 hours
there was a
significant reduction in the bacterial numbers in the nebulized Ag animals.
The
Tobramycin treated animals had a similar cfu counts to the controls at time 24
hours and
48 hours.
24 h animal Control Tobramycin Nebulization


1 0 2 1


2 0 3 1


3 3 1 0


4 3 3 0


5 2 2 3


6 3 2 1


48 h animal Control Tobramycin Nebulization


7 2 1 1


8 1 2 1


9 1 1 0


1 1 0


11 3 1 1


12 Dead Dead Dead


Histopathology of Lung Samples:
Both the control and the Tobramycin treated rats had similar pathology. These
are outlined in Table 6. At 24 and 48 hours severe infiltration of
polymorphonuclear
leukocytes (PMN's) into the interstitial spaces of the lung was observed.
These cellular
elements could also be identified in alveolar and bronchiolar spaces but to a
lesser extent.
The pulmonary vessels were dilated and the alveolar spaces were filled with
71



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
proteinaceous material. The silver-nebulized rats had occasional infiltration
of PMN's
and no evidence of accumulation of fluids in alveolar or bronchiolar spaces.
Histopathology of Lung Samples Removed from Rats
Treatment TimeInflamConsolComments


Score Score


Conhol (1) 24 3 3 Severe infiltration of PMN into
interstitial spaces.


Proteinaceous secretion in alveolar
spaces. Occasional


PMN in alveolar and bronchiolar
space, Consolidation


in affected areas. Involvement
of 70I of sample.


Interstitial Pneumonia.


Control (2) 24 3 3 Severe infiltration of PMNs into
interstitial spaces.


Proteinaceous secretion in alveolar
spaces. Occasional


PMN in alveolar and bronchiolar
space, Consolidation


in affected areas. Involvement
of 80% of sample.


Interstitial Pneumonia


Tobramycin 24 3 3 Severe infiltration of PMNs into
(1) interstitial spaces.


Proteinaceous secretion in alveolar
spaces. Occasional


PMN in alveolar and bronchiolar
space. Consolidation


in affected areas. Involvement
of 90% of sample.


Interstitial Pneumoiua.


Tobramycin 24 3 3 Severe infiltration of PMNs into
(2) interstitial spaces.


Proteinaceous secretion in alveolar
spaces. Occasional


PMN in alveolar and bronchiolar
space. Consolidation


in affected areas. Involvement
of 80% of sample.


Interstitial Pneumonia.


Nebulized 24 0 I No PMNs in area. Slight consolidation.
Ag (I) Normal Lung


Nebulized 24 I 1 Slight infiltration of PMNs around
Ag (2) vessels and


brocheoli.


Control (1) 48 3 3 Severe infiltration of PMNs into
interstitial spaces.


Proteinaceous secretion in alveolar
spaces. Occasional


PMN in alveolar and bronchiolar
space. Consolidation


in affected areas. Involvement
of 80% of sample.


Interstitial Pneumonia.


Control (2) 48 2 2 Severe infiltration of PMNs info
interstitial spaces.


Proteinaceous secretion in alveolar
spaces. Occasional


72



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
PMN in alveolar and bronchiolar
space. Consolidation


in affected areas. Involvement
of 60% of sample.


Interstitial Pneumonia.


Tobramycin 48 3 3 Severe infiltration of PMNs into
(1) interstitial spaces.


Proteinaceous secretion in alveolar
spaces. Occasional


PMN iii alveolar and bronchiolar
space. Consolidation


in affected areas. Involvement
of 70% of sample.


Interstitial Pneumonia.


Tobramycin 48 3 3 Severe infiltration of PMNs into
(2) interstitial spaces.


Proteinaceous secretion in alveolar
spaces. Occasional


PMN in alveolar and bronchiolar
space. Consolidation


in affected areas. Involvement
of 70% of sample.


Interstitial Pneumonia. Slight
infiltration of PMNs


around vessels and brocheoli.


Nebulized 48 1 0 Slight infiltration of PMNs around
Ag (1) vessels and


brocheoli.


Nebulized Normal lung.
Ag (2)


The nebulized nanocrystalline silver reduced bacterial colonization in
Pseudomonas infected lungs reduced injury as determined by gross pathology
(consolidation, hemorrhage, edema) in Pseudomonas infected lungs. Further, the
nanocrystalline silver delivered by aerosol reduced pulmonary inflammation
(primarily
PMN infiltration) in Pseudomonas infected lungs compared to Tobramycin (IM).
Example 6 - Pulmonary Anti-inflammatory Activity
A solution was prepared by sparging C02 through 1000 ml of reverse osmosis
water using commercial CO2 Soda Syphon Charger. This process results in the pH
of the
water dropping to about 4. At this point, approximately 333 ml of the
carbonated water
was decanted into a plastic bottle and 333 square inches of nanocrystalline
silver-coated
net was added to the water. The nanocrystalline silver-coated net and water
were placed
in 37 ° C shaker incubator and shaken at 180 RPM for 30 minutes to
elevate the pH to
approximately 5.8. The solution was then transferred to a beaker and stirred
vigorously
for 2 minutes to raise the pH to approximately at 7.3. The solution had a
final silver
concentration of approximately 400 ppm.
73



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Test solutions of silver nitrate (400 ppm) and silver acetate (400 ppm) were
prepared by dissolving the silver salts in deionized water. A colloidal silver
solution (20
ppm) in was obtained from a cormnercial source.
The dissolution solutions were transferred to commercial nebulizers which were
connected to a Medical air cylinder with a flow rate of approx. 20 L/min. The
outflows
of the nebulizers were connected to an animal chamber housing the rats to be
dosed. All
rats (40 randomly assigned animals) received the dosing. The rats received two
- ~/~ hour
aerosol administrations of the test solutions on the day of infection.
Thereafter, the test
rats were dosed 3 times per day for an additional one and a half days.
Forty male Sprague-Dawley rats were obtained. These animals are specific-
pathogen free and weighed approximately 400 g. The animals were housed in
groups of
four in plastic cages with wire mesh tops. The rats had access to fresh water
and rat
chow ad libitum. All animals were maintained in an appropriate facility with
12-hour
light/dark cycles and constant temperature and humidity, according to facility
standard
~ 5 operating procedures.
The bacteria used for infection of 20 these animals were Pseudofnoraas
aeruginosa strain 5588. The dose was previously titrated to ascertain that a
dose of up to
109 CFU was not lethal for the animals. The bacteria were grown overnight in
Tryptic
soy broth, washed once in sterile PBS and resuspend in sterile PBS. The final
2o concentration of the inoculmn was 4 X 109 CFU/ml.
The rats were anesthetized by inhalation of 2% halothane. A 400 microliter
volume of bacterial suspension was intratracheally administered into the
bronchi of each
rat. This was performed non-surgically on intubated animals. The infection
process
resulted in the instillation of approximately 109 CFU into the lungs of each
animal.
2s Group 1 & 2: Not infected and infected, treated with nebulized silver
nitrate. (10
Rats)
Group 3 & 4: Not infected and infected, treated with nebulized colloidal
silver.
(10 Rats)
Group 5 & 6: Not infected and infected, treated with nebulized nanocrystalline
3o silver. (10 Rats)
74



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Group 7 & 8: Not infected and infected, treated with nebulized silver acetate.
( 10 Rats)
The treatment schedule was as follows:
Day One Day Two
10:00 AM Infection 9:00 AM Third Treatment
4:00 PM First Treatment 1:00 PM Sacrifice, sample 5 rats/Gp
8:00 PM Second Treatment
All rats of each group of animals were sacrificed after 24 h. The lungs of the
animals were aseptically removed, homogenized and plated to determine
bacterial levels.
Lung samples were collected for histological examination. Three lung
homogenates were
reserved for silver analysis. Lungs were grossly scored (absent = 0, mild = 1,
moderate =
2, and severe = 3) based on the degree of involvement of consolidation,
hemorrhage,
edema and necrosis based upon gross observation.
Histopathology was scored (0-4) based upon the degree of consolidation and
inflammation (neutrophil infiltration). The entire right middle lobes of all
rats were
collected for histopahtology. As whole lobes were selected there was no bias
toward any
sample. All samples were fixed in neutral buffered formalin at the time the
lung was
removed from the thorax. It was fixed overnight, dehydrated and embedded in
wax.
Sections were obtained which were hydrated and stained with haematoxylin and
eosin.
2o All sections were examined by a veterinary pathologist who was blinded to
the
treatment groups, until after the samples were scored and comments were
provided, with
scoring being (0 = normal, 1 = slight, 2 = moderate, 3= severe, 4 = very
severe).
All rats in the silver nitrate, silver acetate and colloidal silver groups had
lung that
were grossly scored as moderately to severely inflamed while the lungs of the
~25 nanocrystalline group were grossly scored as normal to slightly inflamed.
This was
confirmed by the histopathological analyses.
The nanocrystalline derived silver solution had pulmonary anti-inflammatory
properties while the other forms of silver did not.
Example 7 - Treatment of an Infected Throat with a Nanocrystalline Silver
3o Derived Solution



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
A forty-nine year old male was suffering from an infected throat. The
condition
was accompanied by fever and a severe pain that made swallowing,very difficult
and
limited the patients ability to sleep. A solution of nanocrystalline derived
silver was
prepared using a method similar to Example 1. This solution was gargled for
one minute
and repeated 3 times over the next ten minutes. Within an hour the pain was
reduced to
the point where the patient could sleep. The treatment was repeated every four
hours for
16h and then once 8h later. The throat infection was cleared as determined by
the
elimination of fever and pain. No further symptoms occurred.
Example 8 - Preparation of Gels
Gels were prepared as described above in Example 11 in the Treatment of
Inflammatory Skin Conditions examples above.
Apoutosis Induction/MMP Modulation
Example 1- Preparation of Nanocrystalline Silver Coatings on Dressings
~ 5 This example shows the preparation of a bilayer nanocrystalline silver
coating on
a dressing material. A high density polyethylene dressing, DELNETTM or
CONFORMANT 2TM was coated with a silver base layer and a silver/oxide top
layer to
generate a coloured antimicrobial coating having indicator value as described
in Example
1 of the Treatment of Hyperproliferative Skin conditions examples. The coating
layers
2o were formed by magnetron sputtering under the conditions set out in the
following table.
Example 2 - Preparation of Atomic Disordered Nanocrystalline Silver Powders
Atomically disordered, nanocrystalline silver powders were prepared as
described
in Example 3 in the Treatment of Inflammatory Skin Conditions examples above.
Example 3 - Preparation of Gels
25 Gels were prepared as described above in Example 11 in the Treatment of
Inflammatory Skin Conditions examples above.
Example 4 - Effects of Antimicrobial Silver on Apoptosis and Matrix
Metalloproteinases in a Porcine Model
A porcine model was used to examine the effects of an antimicrobial metal
3o formed with atomic disorder, preferably silver, on apoptosis and matrix
metalloproteinases. Young, commercially produced, specific pathogen free
domestic
76



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
swine (20 - 25 kg) were used in these studies. The animals were conditioned in
an
animal facility for one week prior to any experimental manipulation.
Typically, three
animals were used in each experiment. The animals received water and hog
ration
(UnifeedTM, Calgary, Alberta) without antibiotics ad libitum, were housed
individually in
suspended stainless steel cages (5' x 6'), and maintained in a controlled
environment with
12 hours of light per day. The study was approved by the University of Calgary
Animal
Care Committee and was conducted in accordance with guidelines established by
the
Canadian Council on Animal Care.
Antimicrobial silver metal was administered in the form of a dressing. The
dressings included:
i) AB - nanocrystalline silver-coated dressing (the non-foam, three-layer
dressing as set out in Example 1), comprising two layers of silver-coated high
density
polyethylene (HDPE) bonded on either side of an absorbent rayoupolyester core;
ii) AgHDPE - nanocrystalline silver coated HDPE layers aseptically separated
~5 from the absorbent core of the AB dressings;
iii) Control - identical in construction to the AB dressing except that the
HDPE
was not coated with nanocrystalline silver;
iv) Gauze - the absorbent rayon/polyester core of the AB dressings;
v) cHDPE - the uncoated HDPE aseptically removed from the absorbent core of
2o the control dressings; and
vi) SN - sterile piece of the gauze dressing to which 24 ,ug silver /square
inch
(from silver nitrate) was added. This amount of silver is equivalent to the
amount of
silver released from a square inch of the AB dressing immersed in serum over a
24 hour
period, as determined by atomic absorption analysis.
25 Dressings (i) - (iii) were gamma sterilized (25 kGy) prior to use. All
dressings
were moistened with sterile water prior to application to the incision. In
some cases, the
incisions were covered with a layer of occlusive polyurethane (TegadermTM, 3M
Corp.,
Minneapolis, MN).
Three isolates of bacteria were used in the inoculum, including Pseudomonas
3o aeYUgiraosa, Fusobacterium sp., and coagulase-negative staphylococci (CNS)
(Culture
Collection, University of Calgary, Calgary, Alberta). The bacterial strains
were grown
77.



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
under appropriate conditions overnight prior to the day of surgery. On the
morning of
surgery, the organisms were washed with sterile water and resuspended to a
final density
of approximately 10' CFU/mL. The bacteria were mixed together in a ratio of
1:0.5:1
(Pseudomoiaas:CNS:Fusobacterium) in water. Sufficient inoculum was prepared to
wet
the incision created in each experiment. This procedure resulted in the
incisions initially
being evenly contaminated with approximately S X 104 CFU/cm2.
Prior to treatment, animals were sedated by an intramuscular injection of a
mixture of 10 mg/kg ketamine (KetaleanTM, MTC Pharmaceuticals, Cambridge, ON )
and
0.2 mg/kg acepromazine (AtravetTM, Ayerst Laboratories), followed by complete
anesthesia induced by mask inhalation of 1- 2 % halothane (MTC
Pharmaceuticals).
Following induction of anesthesia, the dorsal and lateral thorax and abdomen
of each
animal was clipped using a #40 Osler blade and the skin subsequently scrubbed
with a
non-antibiotic soap, and allowed to dry prior to incision.
Animals typically received 20 full-thickness incisions, 10 on each side of the
dorsal thorax. The incisions were created using a 2 cm diameter trephine. An
epinephrine solution was then applied to the incisions to provide for complete
hemostasis
prior to inoculation. The incisions were contaminated by covering them with
gauze
sponges soaked with the bacterial inoculum. The wet sponges were covered with
an
occlusive barrier and allowed to stand for 15 minutes. In some instances, an
incision was
2o then sampled to determine the initial inoculum. Following any requisite
sampling, the
incisions were dressed with the appropriate dressings and covered with an
occlusive layer
that was secured with ElastoplastTM tape (Smith & Nephew, Lachine, QC). All
animals
received narcotic pain medication (TorbugesicTM, Ayerst Laboratories,
Montreal, QC, 0.2
mg/kg), as required.
The experimental and control groups are summarized in the following table:
Animal # Left Side Silver Treatment) Right Side (Controls)
Pig 1 silver nitrate (SN) on gauze gauze moistened with water
Pig 2 AgHDPE cHDPE
3o Pig 3 AB control
78



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
A 2 cm diameter circle of the appropriate dressing was applied to each
incision.
For Pig 1, incisions on the left side were dressed with silver nitrate-
moistened (SN)
gauze, while control incisions on the right side received water-moistened
gauze dressing.
For Pig 2, the incisions on the left side were dressed with silver-coated HDPE
(AgHDPE), while the control incisions on the right side received non-coated
HDPE
(cHDPE). For Pig 3, the incisions on the left side were dressed with AB
dressing, while
incisions on the right side received the vehicle control. For these
experiments, each
incision was individually covered with an occlusive film dressing (TegadermTM,
3M
Corp., Mimleapolis, MN).
Each day following incision (up to 5 days), the dressing materials from each
of
the experimental and control groups were collected and pooled within each
group. These
dressing materials were then placed in conical centrifuge tube containing
glass wool. The
tubes and contents were centrifuged to remove all liquid from the dressings.
The glass
wool was then placed into a 5-mL syringe and pressed to recover the incision
fluid from
~ 5 each of the six sample sets. The incisions were rebandaged in an identical
manner to the
original dressing format each time. Incision fluid which collected under the
occlusive
dressing was also aspirated and reserved for analysis. Due to the small
volumes collected
from each incision, it was necessary to pool the collected fluid from each of
10 incisions
dressed with the same type of dressing. All incision fluids were stored at -
80°C until
2o analysis.
Prior to enzyme zymography or activity assays, the protein concentrations of
the
incision fluid samples were compared to ensure that the protein levels in each
sample
were similar. The samples were diluted 1:100 in water and assayed using the
BCA
Protein Assay SystemTM (Pierce Chemical, Rockford, IL). A standard curve was
25 concurrently constructed using dilutions of bovine serum albumin. Incision
fluid was
diluted in water and then mixed with an equal volume of sample buffer (0.06 M
Tris-
HCI, pH 6.8; 12% SDS; 10% glycerol; 0.005% bromophenol blue). The samples were
then electrophoresed on 10% polyacrylamide (BioRad, Mississauga, ON) gels
containing
0.1% gelatin. The proteins were then incubated in renaturing buffer (2.5%
TritonTM X-
30 100) for 90 minutes at 37°C. Following enzyme renaturation, the gels
were incubated
overnight in substrate buffer (50 mM Tri-HCl, pH 7.8; 5 mM CaCl2; 200 mM NaCI;
79



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
0.02% Brij-35) with or without 10 mM 1,10 phenanthroline. The gels were
subsequently
stained with a standard Coomassie Blue stain and destained in methanol/acetic
acid.
Unless otherwise indicated, all chemicals were obtained from Sigma-Aldrich
(Oakville,
ON).
The incision fluid samples were assayed for the total amount of protein
present.
These values were between 30 - 80 mg/mL. The samples from individual animals
were
even more similar, varying by only 10 - 20 mg/mL in the pooled incision fluid.
i) Assay for Activity of Total MMPs
The total MMP activity of the incision fluid samples was determined by
1o incubating diluted incision fluid with a quenched fluorescein-conjugated
substrate
(EnzChek DQ gelatinTM, Molecular Probes, Eugene, OR) for approximately 20
hours.
Following incubation, the samples were read in a fluorometer (excitation 1=480
nm;
emission 1=520 nm). Activity was compared to a collagenase standard as well as
experimental versus control incision fluids.
~ 5 Fig. 4 shows the change in total MMP activity from differently treated
incision
sites over a five-day period. The silver-coated HDPE (AgHDPE) results were
essentially
identical to those obtained using the silver-coated dressing (AB). Similarly,
the gauze,
non-coated HDPE (cHDPE), and control dressings yielded results essentially
identical to
each other and to untreated incisions under occlusion from which incision
fluid was
2o collected. Only the results from the control, silver-coated dressing (AB),
silver-coated
HDPE (AgHDPE), and silver nitrate moistened gauze (SN) are thus shown. The
total
MMP activity of the incision fluid sample from the control dressing was low
for the first
few days, then rose dramatically and remained high for the duration of the
experiment.
Similarly, the silver-nitrate moistened gauze (SN) demonstrated an almost
identical
25 pattern of total MMP activity. Results from the silver-coated dressing (AB)
yielded
dramatically different results. The level of MMP activity remained steady for
the
duration of the experiment and did not spike to high levels. Instead, it
remained at a level
roughly 60% lower than the highest level of activity reached in control or
silver-nitrate
moistened gauze (SIB.
3o ii) Assay for Activity of Gelatinases



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Gelatinases include MMP-2 (secreted by fibroblasts and a wide variety of other
cell types) and MMP-9 (released by mononuclear phagocytes, neutrophils,
corneal
epithelial cells, tumor cells, cytotrophoblasts and keratinocytes). The
gelatinases degrade
gelatins (denatured collagens) and collagen type IV (basement membrane).
Zymograms
were run to examine changes in the levels and activity of MMP-9 and MMP-2 over
the
duration of the experiment.
Results of the zymograms for the control and silver nitrate moistened gauze
(SN)
appeared to be identical. The levels of MMP-9 declined over the period
examined,
particularly levels of the active form of MMP-9. The silver-coated dressing
(AB)
demonstrated higher levels of active MMP-9 than for the control. On Day 2, the
silver-
coated dressing (AB) showed lower levels of active MMP-9 than in the control.,
On Day
4, the silver-coated dressing (AB) showed little active MMP-9. In the control,
the
amount of the latent enzyme appeared to decrease while the active form of MMP-
9
increased, particularly up to Day 4.
~ 5 There was not much difference in the levels of MMP-2 activity for the
silver-
coated dressing (AB) over the duration of the experiment. However, there was
an
increase in the level of active MMP-2 in the control dressing by Day 5. It was
also
observed that the levels of some other, unidentified, gelatinolytic enzymes
also decreased
in the silver-coated dressing (AB) compared to the control.
2o iii) Assay of Total Protease Activity
Since MMPs have proteolytic activity, the total protease activity in incision
fluid
samples was assessed by incubating the samples with 3 mg/mL azocasein in 0.05
M Tris-
HCI, pH 7.5 for 24 hours at 37°C. The undigested substrate was then
precipitated by the
addition of 20% trichloroacetic acid. The absorbance of the supernatant was
then
25 assessed using a spectrophotometer, 1=400 nm. The absorbance was compared
to a
standard curve prepared with bovine pancreatic trypsin.
Results paralleled those obtained in the total MMP assay above. The incision
fluid samples for the control and silver nitrate moistened gauze (SN)
demonstrated a
pronounced increase in activity after Day 2 (Fig. 5). Incision fluid from the
silver nitrate
3o moistened gauze (SN) also demonstrated a marked increase in the total
protease activity
compared to control dressing incision fluid (Fig. 4). However, the total
protease activity
81



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
in the incision fluids of the silver coated dressings (AB) remained constant
over the
duration of the experiment.
Antimicrobial silver was thus demonstrated to be effective in modulating
overall
MMP activity. However, silver nitrate was not effective in modulating MMP
activity in
spite of the Ag+ concentration being approximately the same levels as were
expected to
be released from the silver-coated dressing (AB) over the same period of time
(24 h)
between applications. The reason for the difference in effects may be related
to the
inherent nature of the two silver formulations. In the case of silver nitrate,
although the
silver was added to provide a similar final concentration of Ag+ as was
anticipated to be
released from the silver-coated dressing (AB), the Ag+ ions were added at
once. It would
thus be expected that the serum proteins and chlorides within the incision
fluid would
quickly inactivate a large portion of the Ag+. In the case of the silver-
coated dressing
(AB), the silver is continuously released to maintain a steady-state
equilibrium,
maintaining an effective level of silver in the incision for a prolonged
period.
~ 5 iv) Apoptosis
Histological assessment of cell apoptosis was carried out in order to
determine
whether the silver-coated dressing (AB) affected apoptosis within the
incision.
Histological Observations of Porcine Tissue
Samples of tissue from the incisions were collected daily for the duration of
the
2o experiment, except for Day l, and examined for evidence of apoptosis. The
samples
were fixed in 3.7% formaldehyde in PBS for 24 hours, then embedded in
paraffin, and
cut into 5 mm thick sections. The samples were then de-waxed with Clearing
SolventTM
(Stephan's Scientific, Riverdale, NJ) and rehydrated through an ethanol:water
dilution
series. The sections were treated with 20 mg/mL proteinase K (Qiagen,
Germantown,
25 MD) in lOmM Tris-HCl (pH 7.4) for 30 minutes at room temperature.
Terminal deoxynucleotidyl transferase nick end labeling (TUNEL staining) was
performed using an In Situ Cell Death Detection POD KitTM (Boehringer
Mannheim,
W dianapolis, III. Using this technique, cells which stain brown are those
being
eliminated by apoptosis. Endogenous peroxidase was blocked with 3% hydrogen
3o peroxide in methanol for 10 minutes at room temperature then cells were
permeabilized
with 0.1% TritonTM X-100 (in 0.1% sodium citrate) for 2 minutes on ice. After
82



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
permeabilization, the samples were treated with the terminal transferase
enzyme solution
incubated in a humidified chamber at 37°C for 60 minutes. Following
labelling, the
samples were washed once with 1.0% TritonTM X-100 and twice with PBS. The
sections
were incubated with Converter-PODTM (Boehringer Mannheim, Indianapolis, IN) in
a
humidified chamber at 37°C for 30 minutes, and repeated washing with
1.0% TritonTM
X-100 and PBS. Subsequently, the samples were incubated with DAB substrate
(Vector
Laboratory Inc., Burlingame, CA) for 10 minutes at room temperature and washed
with
1.0% TritonTM X-100 and PBS. It was also necessary to counterstain the
sections with
hematoxylin nuclear counterstain (Vector Laboratory Inc., Burlingame, CA) for
10
seconds.
The prepared samples were then ready to be observed by light microscopy for
evidence of apoptosis. For a positive control, the permeabilized sections were
treated
with 100 mg /mL DNase I in PBS for 10 minutes at room temperature to induce
DNA
strand breaks. For negative controls, the terminal transferase enzyme, POD or
DAB were
~5 omitted between each labelling step.
In all samples examined, there was little difference between the control and
silver
nitrate moistened gauze (SN). However, significant apoptosis of the cell
population was
observed in incisions of the silver-coated dressing (AB). In the control
incision, there
were significant numbers of polymorphonuclear leukocytes (PMNs) and few
fibroblasts,
2o while in incisions of the silver-coated dressing (AB), there were
significantly more
fibroblasts and few PMNs.
Histopatlaological Scoring of Pof~cine Tissue
Animals were anesthetized as described above of Days 1, 4, and 7. A mid-
incision biopsy was collected with a sterile 4 mm biopsy punch. The tissue was
fixed in
25 10% neutral buffered formalin, embedded in methacrylate and sectioned (2 -
5 mm
thick). The sections were stained with Lee's methylene blue and basic fuschin
to
demonstrate the cellular organization and bacteria. A pathologist blinded to
the
treatments scored the sections based on the presence of fibroblasts, PMNs and
bacteria as
follows: 0 = absent; + = occasional with 1-5 per high power field of view; ++
= moderate
3o with 6-20 per high power field of view; +++ _- abundant with 21-SOper high
power field
83



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
of view; ++++ = very abundant with more than 50 per high power field of view
(see the
following table).
Day Post- Dressing Fibroblasts PMNs Bacteria
Incision


1 Silver coated++ ++ +


1 Control 0 +++ ++++


4 Silver coated++++ ++ 0


4 Control + ++++ ++++


7 Silver Coated++++ + 0


7 Control +++ +++ +++


The microscopic observation of the biopsy samples revealed that the
infiltrating
cell types were significantly different between the control and silver-coated
dressings
(AB). The control incisions were characterized by a large numbers of PMNs,
while the
silver-coated dressings (AB) demonstrated a larger proportion of fibroblasts
and
monocytes. The relative abundance of the fibroblasts in incisions of the
silver-coated
dressings (AB) became increasingly pronounced through to Day 7, as compared to
the
control incisions that remained populated largely by PMNs and monocytes. The
staining
method enabled staining also of bacteria, which was abundant in the control
incision but
generally absent in the incisions of the silver-coated dressings (AB).
15 Incisions treated with the nanocrystalline antimicrobial silver thus
demonstrated
more extensive apoptosis than did cells from incisions treated with either
control or silver
nitrate dressings. During the first two days following incision, the cell type
which
demonstrated the most pronounced increase in apoptosis were neutrophils. This
suggests
that part of the reason for the moderated neutrophil presence and the
resultant modulation
20 of MMP levels was due to neutrophil apoptosis. It has been shown that the
number of
84



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
apoptotic cells increases as the incision closes and that this is part of the
mechanism
involved in the decrease in cellularity of the maturing scar tissue
(Desmouliere, A.,
Badid, C., Bochaton-Piallat, M. and Gabbiani, G. (1997) Apoptosis during wound
healing, fibrocontractive diseases and vascular wall injury. Int. J.
BiocTZern. Cell Biol. 29:
19-30.). The results suggest that the maturing of the nascent dermal and
epidermal
tissues may also be accelerated in the presence of the nanocrystalline
antimicrobial metal-
containing materials. The findings indicated that acceleration in healing
induced by the
nanocrystalline antimicrobial metal-containing materials is associated with a
reduction of
local MMP activity, as well as with an increased incidence of cell apoptosis
within the
incision.
Example 5 - Clinical Study on the Effect of Silver-Coated Dressings on MMPs
and Cytokines
This study was conducted to assess the effect of the silver-coated dressing on
the
concentrations of MMPs and cytokines in non-healing wounds over time during
~ 5 treatment. The modulation of the levels of active MMPs and cytokines may
alleviate the
inflarmnatory response in a wound, allowing the wound to advance through the
subsequent stages of wound healing culminating in a healed wound.
A total of 10 patients with non-healing venous stasis ulcers were randomly
. assigned to treatment with a silver-coated dressing (5 patients) or a
control dressing (5
2o patients). The silver-coated dressing was prepared as in Example 1. The
control dressing
was identical in construction to the silver-coated dressing of Example 1,
except that the
HDPE was not coated with silver. The ulcers were dressed in appropriate
pressure
dressings to correct the underlying medical problem. Samples of the ulcer
fluid were
collected before treatment (day 0) and at weekly intervals (days 1, 7, 14 and
21) by
25 removing the silver-coated dressing or control dressing, and replacing the
dressing with
TegadermTM occlusive dressing (3M Corp., Minneapolis, MN) for one hour to
allow
wound fluids to collect. The fluid samples were aspirated from below the
dressing in a
syringe, and were frozen at -~0°C until assayed.
Assays were conducted for active MMP-9, active MMP-2, Tumor necrosis factor-
a (TNF-oc) and Interleukin-1 ~3 (IL-1 [3). High levels of MMP-9 and MMP-2 are
predominant in non-healing wounds, with levels decreasing over time in normal
healing



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
wounds. Released by activated macrophages, TNF-a and IL-1 (3 are indicators of
wound
inflammation. Levels of TNF-a and IL-1 (3 are elevated in non-healing wounds
and
increase release of pro-MMPs, for example, MMP-9 and MMP-2.
To measure the levels of active MMP-9 and MMP-2, enzyme capture assays
(BioTrak, NJ) were conducted. In this method, active enzyme is detected
through
activation of a modified pro-detection enzyme and the cleavage of its
chromogenic
peptide substrate. The resultant color is read by spectrophotometer, and the
concentration
of MMP is determined by interpolation of a standard curve, expressed in ng/ml
(see
results in Figs. 6 and 7).
To assay the levels of cytokines, IL-1 (3 levels were measured using a
sandwich
immunoassay (BioTrak, NJ), while TNF- oc levels were measured by a high
sensitivity
sandwich antibody assay (BioTrak, NJ). In both methods, endogenous cytokine is
bound
to an immobilized antibody and then detected by an addition of a
biotinylated,antibody,
followed by a colorimetric substrate. The color is measured by a
spectrophotometer, and
~ 5 the concentrations of TNF-a, and IL-1 (3 are determined by interpolation
of a standard
curve and expressed as pg/ml (see results in Figs. 8 and 9).
Total protein levels were measured for each sample to standardize the measures
of
the MMPs and cytokines. Total protein levels were measured using BCA Protein
Assay
SystemTM (Pierce Chemical, Rockford, IL). No protein level of any sample was
2o significantly different from the total mean.
Fig. 6 is a graph showing the concentrations (ng/ml) of active MMP-9 in fluid
samples recovered from ulcers dressed with silver-coated dressing (Silver) and
control
dressing (Control) at days 0, 1, 7, 14 and 21. The levels of active MMP-9
decreased to a
normal level, and were suppressed over time with the silver-coated dressing
compared to
25 the control dressing, demonstrating a modulating effect of the silver-
coated dressing.
Fig. 7 is a graph showing the concentrations (ng/ml) of active MMP-2 in fluid
samples recovered from ulcers dressed with silver-coated dressing (Silver) and
control
dressing (Control) at days 0, 1, 7, 14 and 21. The levels of active MMP-2 were
not
significantly different with the silver-coated dressing and the control
dressing.
so Fig. 8 is a graph showing the concentrations (pg/ml) of TNF-a, in fluid
samples
recovered from ulcers dressed with silver-coated dressing (Silver) and control
dressing
86



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
(Control) at days 0, 1, 7, 14 and 21. The levels of TNF-a were suppressed over
the
treatment period, and did not increase significantly over the treatment period
with the
silver-coated dressing, while the levels in the control dressing increased,
demonstrating a
modulating effect of the silver-coated dressing.
s Fig. 9 is a graph showing the concentrations (pg/ml) of IL-1 (3 in fluid
samples
recovered from ulcers dressed with silver-coated dressing (Silver) and control
dressing
(Control) at days 0, 1, 7, 14 and 21. The levels of IL-1 [3 were not
significantly different
with the silver-coated dressing and the control dressing.
The study suggests that the modulation of the MMP-9 and TNF-oc levels is
responsible for improved wound healing and reduced inflammation with silver-
coated
dressings. In comparison, the levels of MMPs and cytokines did not decrease
over time
with the control dressings.
This example and Example 4 above, taken together with the evidence that the
silver materials herein disclosed are capable of reducing inflammation (see co-
pending
1s United States Patent Application Nos. 10/131,568; 10/131,511; 10/131,509;
10/131,513;
and 10/128,208 filed April 23, 2002; and co-pending United States Patent
Application
No. 09/840,637 filed April 23, 2001, and United States Provisional Patent
Application
No. 60/285,884 filed April 23, 2001) demonstrates a method of reducing
inflammation in
a patient in need thereof, by contacting an area of inflammation or an
inflammatory cell
2o with a therapeutically effective amount of the antimicrobial metal-
containing materials in
a crystalline form. The antimicrobial metal-containing materials are
characterized by
sufficient atomic disorder, such that the metal, in contact with an alcohol or
water-based
electrolyte, releases atoms, ions, molecules, or clusters of at least one
antimicrobial metal
at a concentration sufficient to modulate the release of one or both of MMP-9
and TNF-
25 oc. Excessive TNF production has been reported in diseases, such as cancer
and
autoimmune diseases, which are characterized by elevated MMP activity. In this
regard,
use of the nanocrystalline silver of the present invention, when in
therapeutically
effective amounts, provides the dual modulation of MMP-9 and TNF-oc to
alleviate the
particular condition.
87



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Additional Examples
Examt~le 1
6 milligrams of antimicrobial metal-containing material with atomic disorder,
in
free-standing powder form, are sprinkled lightly onto 6.5 cm2 of burned
tissue, and
thereafter wet with a light spray of water or wound exudate or TDWL (Trans
Dermal
Water Loss) or other bodily fluids, so as to provide an antimicrobial
treatment to the
burned tissue. The treatment is repeated every 24 hours until the therapeutic
effects are
no longer needed.
Example 2
0.5 milligrams of antimicrobial metal-containing material with atomic
disorder, in
free-standing powder form, are inj ected, using a small-needle drug delivery
system or a
needle-less drug delivery system, into gum tissue so as to treat gingivitis.
The treatment
is repeated every 3 days until the therapeutic effects are no longer needed.
Example 3
~5 A solution of antimicrobial metal-containing material with atomic disorder
is
prepared by dissolving 6 milligrams of antimicrobial metal-containing material
with
atomic disorder in 1 gram of water. The solution of antimicrobial metal-
containing
material with atomic disorder is applied as a rinse or bath or wash to a wound
site so as to
provide an antimicrobial treatment to the wound site. The treatment is
repeated every 24
2o hours until the therapeutic effects are no longer needed.
Example 4
A solution of antimicrobial metal-containing material with atomic disorder is
prepared by dissolving 6 milligrams of antimicrobial metal-containing material
with
atomic disorder in 1 gram of water. The solution of antimicrobial metal-
containing
25 material with atomic disorder is applied to the interior of the bladder via
a catheter so as
to provide antimicrobial treatment to the bladder. The treatment is repeated
every 8 hours
until the therapeutic effects are no longer needed.
Example 5
A solution of antimicrobial metal-containing material with atomic disorder is
3o prepared by dissolving 6 milligrams of antimicrobial metal-containing
material with
atomic disorder in 1 gram of water. The solution of antimicrobial metal-
containing
88



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
material with atomic disorder is injected (using a small-needle or needle-less
injection
system) under the toenails or into the nail bed and/or the surrounding tissue
of a person
suffering from onychomycosis so as to provide an antimicrobial treatment to
the tissue.
The treatment is repeated 2 times a day until the therapeutic effects are no
longer needed.
Example 6
Summary
Solutions of nanocrystalline noble metal-containing materials were prepared by
immersing Acticoat~ burn dressings (distributed by Smith & Nephew) in reverse
osmosis water that had been pretreated with C02 in order to reduce the pH. Two
different concentrations of antimicrobial metal-containing material with
atomic disorder
solutions were prepared by this method, the concentrations being 85 mg/mL and
318
mg/mL. Solutions of silver nitrate were also prepared to use as comparisons in
the
experiments. The concentrations of the silver nitrate were 103 ppm of silver
and 295
ppm of silver as determined by Atomic Absorption Spectroscopy.
~ 5 The solutions were in turn placed in an ultrasonic nebulizer that created
small
droplets containing dissolved and suspended parts of the solution of
nanocrystalline noble
metal-containing material. The output from the nebulizer was directed into a
chamber
made from a stainless steel frame and base. Petri dishes containing Mueller
Hinton agar
streaked with 4 h old cultures of Pseudomonas aerugiona and Staphylococcus
aureus
2o were exposed to nanocrystalline noble metal aerosols and the silver nitrate
aerosols.
The results of the tests show that nanocrystalline noble metal aerosols
transmit the
antimicrobial activity of the dressings to remote sites, and nanocrystalline
noble metal
aerosols are more effective than comparable concentrations of silver nitrate.
Introduction
25 In many instances the delivery of antimicrobial materials may most
expeditiously
be accomplished by using aerosols (e.g., in the treatment of pneumonia). The
drawback
of aerosols is the requirement for a high concentration of the active
ingredient to ensure
that a sufficient amount is delivered to achieve the biological effect desired
without
causing problems with the carrier solvent (e.g., water). The essential
requirement of the
so equipment for producing an aerosol that contains dissolved and suspended
components of
antimicrobial metal-containing material with atomic disorder is that it must
form droplets
89



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
of aerosol directly from the liquid form, and the aerosol droplets must be
small enough to
reach the lungs. This means that the droplets should be preferably less than
approximately 10 mm. To meet these requirements, the aerosol cannot be created
by first
evaporating the liquid and then condensing it to form droplets, since this
would remove
the desired antimicrobial metal-containing material with atomic disorder from
the
aerosol. There are two methods that can be used to relatively easily form the
droplets
directly: (1) mechanical disruption of the liquid; and (2) air, under
pressure, passing
through some form of orifice that combines the air and the liquid in a way
that creates
droplets instead of evaporating the liquid.
Several experiments were carned out with antimicrobial metal-containing
material with atomic disorder and silver nitrate solutions to determine if the
antimicrobial
activity of the dressing could be transferred through a direct droplet aerosol
to a Petri
dish.
Equipment
~ 5 The method used to create an aerosol for these tests was the mechanical
method in
the form of an ultrasonic nebulizer. For convenience, an ultrasonic
humidifier.was used.
The liquid containing the dissolved and suspended antimicrobial metal-
containing
material with atomic disorder was placed in the water reservoir of the
humidifier. When
power was applied to the humidifier, aerosol droplets of dissolved and
suspended
2o antimicrobial metal-containing material with atomic disorder were generated
and flowed
from the output nozzle.
A test chamber was constructed using a stainless steel frame with a
transparent
plastic covering. The frame was placed on a stainless steel plate. The output
nozzle from
the humidifier was modified so that the aerosol could be directed into the
chamber at a
25 height of approximately 30 cm from the base. The plates and other test'
samples were
placed on the stainless steel plate and exposed to the aerosol for a
prescribed length of
time.
Solution 1
A solution of antimicrobial metal-containing material with atomic disorder was
so prepared by immersing 518 sq. inches ofActicoat~ burn dressing in 1L of
reverse



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
osmosis water, which was treated with COZ to maintain a pH of 6.5. After 20
minutes the
concentration of silver in the water was 85 mg/mL.
Solution 2
A solution containing 370 mg/mL of silver from a Acticoat~ dressing was
prepared as follows: 1 L of reverse osmosis water was purged with cormnercial
grade
carbon dioxide until the pH was 4.3. Sufficient Acticoat~ dressing was added
to bring
the pH up to 6.5. At that time, the silver concentration was 370 mg/mL.
Solution 3
Ag as AgN03 was prepared by dissolving 0.157 g of AgN03 into 1 L of reverse
osmosis water and mixed until dissolved. The solution was analyzed by Atomic
Absorption Spectroscopy and found to be 102.9 ppm of silver.
Solution 4
Ag as AgN03 was prepared by dissolving 0.427 of AgN03 into 1 L of reverse
osmosis water and mixed until dissolved. The solution was analyzed by Atomic
~5 Absorption Spectroscopy and found to be 295 ppm of silver.
Aerosolization
Petri dishes, containing Mueller Hinton agar, were streaked with 4 h old
cultures
of Pseudomonas aeruginosa or Staphylococcus aureus. The plates were then
weighed and
their exposed outer surfaces were coated with Parafilm to prevent condensation
from
20 occurring on these surfaces. These plates were placed in the aerosol
chamber uncovered.
The ultrasonic nebulizer was then started and run for 53 minutes. The plates
were then
removed from the chamber, the plastic was removed and the dishes re-weighed so
that the
amount of moisture loss/gain could be determined.
The plates were then placed in a 35°C incubator for 16 h. After
incubation the
25 pattern and amount of growth was assessed on the plates for both organisms.
Viability Assessment
Three of the six plates made for each organism were tested to determine if the
antimicrobial effect was cidal or static in nature. This was accomplished by
rinsing or
placing a piece of the clear section of agar in the Petri dish plates into
Tryptic soy broth in
3o a test tube and incubating for 4 h or 16 h. If the medium turned turbid in
4 h it would
indicate that the antimicrobial affect was bacteriostatic in nature. If the
organism took
91



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
more than 16 h to grow, as indicated by turbidity, it was considered an
indication that
both static and cidal effects occurred. If no growth occurred, the effect was
bactericidal.
Results
The results for Solutions 1 and 3 are summarized in the following two table.
Antimicrobial AgNo3
Metal-Containing
Material
With


Atomic Disorder


Organism


Ps. AeruginosaS. aureus Ps. AeruginosaS, aureus


Ag concentration85 85 99 99


(~g/mL)


pH of test 6.5 6.5 Approx. Approx. 6.5
solution 6.5


Exposure 53 53 53 53
time


(minutes)


Exposed area9.8 9.8 9.8 9.8
(sq.in)


Weight gain 0.8 0.8 1.05 1.05
(g)


Growth at 0 0 0 +-H-+
16h


(0-++++) 0 ++ 0 ++++
at 48h


Viable 4h No Yes No Yes


16h Yes Yes Yes Yes


The results for Solutions 2 and 4 are summarized in the following two table.
Antimicrobial AgNo3
Metal-Containing
Material
With


Atomic Disorder


Organism


Ps. aeruginosaS, aureus Ps. aeruginosaS, aureus


Ag concentration370 370 300 300


(ltglmL)


pH of test 6.5 6.5 Approx. 6.3 Approx. 6.3
solution


Exposure 53 53 53 53
time


(minutes)


Exposed area9.8 9.8 9.8 9.8
(sq.in)


Weight gain 1.14 1.14 1.12 1.12
(g)


Growth at 0 0 0 0
16h


(0-++++) 0 0 0 +++
at 48h


Viable 4h No No No No


16h No No No N/A


92



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Discussion
At the low concentration of silver in solution, the Acticoat~ dressing
generated
silver was effective at controlling the growth of both organisms while the
silver nitrate
only prevented the growth of Ps. aeruginosa. Viability tests showed that at
the low
concentration, neither form of silver was completely bactericidal although the
poor
growth on the plates treated with antimicrobial metal-containing material with
atomic
disorder compared to the silver nitrate treated plates suggests that a
significant log
reduction occurred in the plates treated with the aerosol of antimicrobial
metal-containing
material with atomic disorder.
At a higher concentration of silver, both antimicrobial metal-containing
material
with atomic disorder (370 mghnL) and AgN03 (300 mg/mL) were effective at
controlling
P. aeruginosa. Since no retgrowth occurred, it is assumed that the agent at
this
concentration was bactericidal. Antimicrobial silver with atomic disorder was
more
effective than Ag NO3 at controlling S. aureus. No re-growth occurred on any
plates or in
~ 5 the broth indicating a total kill of the organism while, in the Ag N03
treatment, a large
number of organisms grew at 16h.
Based on weight gain during aerosol treatments, a dose per unit area can be
calculated. In each case for Solution 1, the dose was 8.5 mg/sq. inch, while
for Solution
2, the dose was 38 mg/sq. inch. These doses, on a per lung basis, would be
less than 10
2o mg of silver per hour of treatment. Each hour of treatment with
antimicrobial silver with
atomic disorder aerosols appears to provide at least 48 h of protection.
Therefore, the
dose per day, from the high concentration treatment, would be about 5 mg or a
little less
than the silver released by 2 sq. inches of SSD per day.
The most significant advantage of using antimicrobial silver with atomic
disorder
25 may be the lack of a toxic action such as N03 or sulfadiazine.
Conclusions
(1) Aerosols of antimicrobial metal-containing material with atomic disorder
transmit the antimicrobial activity of the dressings to remote sites.
(2) Aerosols of antimicrobial metal-containing material with atomic disorder
3o are more effective than comparable concentrations of silver nitrate.
(3) The dose delivered is acceptable and would
93



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
not appear to be excessive.
(4) No toxic cations (N03 or sulfadiazine) are
introduced to the patient.
Example 7 (Gels of Antimicrobial Metal-Containing Material With Atomic
Disorder
Gel products of antimicrobial metal-containing material with atomic disorder
encompass both wet and dry materials.
A wet gel product of antimicrobial metal-containing material with atomic
disorder
is a product that provides moisture to a dry skin condition (psoriasis,
eczema, acne,
wound, etc.) and facilitates autolytic debridement of necrotic tissue. It also
delivers the
antimicrobial and anti-inflammatory properties of the suspended antimicrobial
metal-
containing material with atomic disorder powders.
In many instances it is also beneficial to supply biologically active
molecules to
elicit a specific response such as cell migration, etc. Since these
biologically active
molecules are susceptible to microbial degradation if not protected, it is
beneficial to
include them in gels of antimicrobial metal-containing material with atomic
disorder that
will provide the necessary protection.
Dry gel products of antimicrobial metal-containing material with atomic
disorder
are physically stabilized (dry or cross-linked) materials that provide
lubricious,
2o antimicrobial, antithrombogenic, and anti-inflammatory properties to a
variety of
implantable, trancutaneous or topically applied devices. The coatings may also
provide
other benefits such as accelerating or otherwise facilitating tissue
integration by creating
a favorable environment for cell proliferation. This favorable environment may
be
created by including cyto-conductive agents or anti-adhesion agents such as
bone
25 morphogenetic proteins, B-glucan hyaluronic acids in the gel. The gel may
be stabilized
by cross-linking the gel components (collagen, gelatin, etc.) or by drying the
coated
materials.
Examples of the primary gelling agents are listed in the following table.
Biologically active ingredients that may be used, in any combination with the
primary
3o gelling agents, are given in the subsequent table. Materials that should
not be used with
gels of antimicrobial silver with atomic disorder are given in the final
table.
94



CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Material Percentage Composition


Carboxymethyl cellulose (CMC)0.1-10


Polyvinyl alcohol (PVA) 0.1-10


Collagen ~ 0.1-10


Pectin 0.1-10


Gelatin 0.1-10


Chitin 0.1-10


Chitosan 0.1-10


Alginate 0.1-10


Poly (a,-amino acids)


Polyester


Poly-1-caprolactone


PEG


Cocoa butter


Sepigel


Biologically Active IngredientsPercentage Composition


Methyl paraben <3


Propylparaben <3


B-glucan <5


Hyaluronic acid <5


Epidermal growth factor <1


Platelet derived growth factor<1


Transforming growth factor <1


Vascular endothelial growth <I
factor


Interleukins <1


Heparin <5


Bone morphogenetic proteins <1


Non-Compatible Materials Percentage Composition


Chloride salts >0.01


Aldehydes >0.01


Ketones >0.01


Long chain alcohols >0.01


Glycerol >0.01


Triethanolamine >0.01





CA 02500836 2005-03-31
WO 2004/037187 PCT/US2003/033446
Example 8 (Examples of Gels with Antimicrobial Metal-Containing Material
With Atomic Disorderl
Gels were prepared as described above in Example 11 in the Treatment of
Inflammatory Skin Conditions examples above.
Other embodiments are in the claims.
96

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2003-10-22
(87) PCT Publication Date 2004-05-06
(85) National Entry 2005-03-31
Examination Requested 2008-10-15
Dead Application 2015-07-07

Abandonment History

Abandonment Date Reason Reinstatement Date
2010-10-22 FAILURE TO PAY APPLICATION MAINTENANCE FEE 2011-03-25
2011-12-21 R30(2) - Failure to Respond 2012-10-18
2014-07-07 R30(2) - Failure to Respond
2014-10-22 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2005-03-21
Maintenance Fee - Application - New Act 2 2005-10-24 $100.00 2005-10-06
Registration of a document - section 124 $100.00 2005-12-20
Registration of a document - section 124 $100.00 2005-12-20
Registration of a document - section 124 $100.00 2005-12-20
Maintenance Fee - Application - New Act 3 2006-10-23 $100.00 2006-10-05
Maintenance Fee - Application - New Act 4 2007-10-22 $100.00 2007-10-04
Request for Examination $800.00 2008-10-15
Maintenance Fee - Application - New Act 5 2008-10-22 $200.00 2008-10-20
Maintenance Fee - Application - New Act 6 2009-10-22 $200.00 2009-10-22
Registration of a document - section 124 $100.00 2010-04-23
Reinstatement: Failure to Pay Application Maintenance Fees $200.00 2011-03-25
Maintenance Fee - Application - New Act 7 2010-10-22 $200.00 2011-03-25
Maintenance Fee - Application - New Act 8 2011-10-24 $200.00 2011-10-21
Maintenance Fee - Application - New Act 9 2012-10-22 $200.00 2012-10-15
Reinstatement - failure to respond to examiners report $200.00 2012-10-18
Maintenance Fee - Application - New Act 10 2013-10-22 $250.00 2013-10-16
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
SMITH & NEPHEW (OVERSEAS)LIMITED
Past Owners on Record
BURRELL, ROBERT E.
GILLIS, SCOTT H.
LAM, KAN
MOXHAM, PETER H.
NAYLOR, ANTONY G.
NUCRYST PHARMACEUTICALS CORP.
SCHECHTER, PAUL
STILES, JAMES ALEXANDER ROBERT
WRIGHT, JOHN B.
YIN, HUA QING
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2005-03-31 2 75
Claims 2005-03-31 15 611
Drawings 2005-03-31 5 68
Description 2005-03-31 96 5,418
Representative Drawing 2005-03-31 1 2
Cover Page 2005-06-21 2 44
Claims 2012-10-18 5 184
Description 2013-10-07 96 5,491
Claims 2013-10-07 2 70
Fees 2005-10-06 1 28
PCT 2005-03-31 7 377
Assignment 2005-03-31 5 151
Prosecution-Amendment 2005-11-25 1 43
Assignment 2005-12-20 20 515
Prosecution-Amendment 2006-01-18 3 115
Prosecution-Amendment 2006-02-14 1 34
Fees 2006-10-05 1 31
Prosecution-Amendment 2007-06-06 2 51
Prosecution-Amendment 2007-08-28 1 37
Fees 2007-10-04 1 31
Prosecution-Amendment 2008-10-15 1 38
Fees 2008-10-20 1 35
Fees 2009-10-22 1 39
Assignment 2010-04-23 9 313
Fees 2011-03-25 1 44
Prosecution-Amendment 2011-06-21 4 156
Fees 2011-10-21 1 43
Fees 2012-10-15 1 44
Prosecution-Amendment 2012-10-18 9 352
Prosecution-Amendment 2013-04-05 2 98
Prosecution-Amendment 2013-10-07 9 378
Fees 2013-10-16 1 33
Prosecution-Amendment 2014-01-07 3 145