AU2002362836A1 - Compositions and methods for delivery of poorly water soluble drugs and methods of treatment - Google Patents

Compositions and methods for delivery of poorly water soluble drugs and methods of treatment

Info

Publication number
AU2002362836A1
AU2002362836A1 AU2002362836A AU2002362836A AU2002362836A1 AU 2002362836 A1 AU2002362836 A1 AU 2002362836A1 AU 2002362836 A AU2002362836 A AU 2002362836A AU 2002362836 A AU2002362836 A AU 2002362836A AU 2002362836 A1 AU2002362836 A1 AU 2002362836A1
Authority
AU
Australia
Prior art keywords
composition
nanoparticulates
paclitaxel
particle size
micrometers
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2002362836A
Other versions
AU2002362836B2 (en
Inventor
Jeffrey M Jonas
Roger A Rajewski
Bala Subramaniam
Katherine Fern Terranova
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
CritiTech Inc
Original Assignee
CritiTech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by CritiTech Inc filed Critical CritiTech Inc
Priority claimed from PCT/US2002/032820 external-priority patent/WO2003032906A2/en
Publication of AU2002362836A1 publication Critical patent/AU2002362836A1/en
Application granted granted Critical
Publication of AU2002362836B2 publication Critical patent/AU2002362836B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Description

COMPOSITIONS AND METHODS FOR DELIVERY OF POORLY WATER SOLUBLE DRUGS AND METHODS OF TREATMENT
BACKGROUND OF THE INVENTION
The present embodiment of the invention is directed generally to the prevention and treatment of disease such as neoplastic cell growth and proliferation and, more specifically, to compositions of poorly water soluble compounds, such as, antimitotics and antibiotics, and methods of delivering such compositions for the prevention and treatment of cancers and tumors.
Cancer is one of the leading causes of death in the United States and is characterized by uncontrolled increases in abnormal or neoplastic cells that form a tumor mass and invade adjacent tissues. Malignant cells spread by way of the blood system, the lymphatic system to lymph nodes, by migration of cancer cells within the fluids of the peritoneal cavity, and to distant sites in a process known as metastasis.
Numerous compounds are known which are useful in the prevention and treatment of various types of cancer. In order to effectively deliver these compounds by intravenous administration, it is generally preferred that the compounds be in solution to avoid or reduce the risk of blood clotting or other adverse effects that could result if the compounds were delivered in particulate form. Unfortunately, many of these compounds have poor solubility in water, the preferred solvent, and must be delivered using solvents which can cause adverse patient reactions that must in turn be prevented or controlled through the administration of other compounds. For example, paclitaxel is a known inhibitor of cell division or mitosis and is widely used in the treatment of ovarian, breast, lung, esophageal, bladder, head and neck cancers. Paclitaxel is a natural product originally purified from the bark of yew trees, but now obtained by semisynthesis from 10-desacetylbaccatin, a precursor purified from yew leaves. Paclitaxel, however, is poorly water soluble and is conventionally solubilized in Cremophor EL, a formulation comprising 50% ethyl alcohol and 50% polyethoxylated castor oil. Cremophor EL is believed to result in histamine release in certain individuals and patients receiving paclitaxel in that delivery method must normally be protected with a histamine Hi-receptor antagonist, an H2-receptor antagonist and a corticosteroid to prevent severe hypersensitivity reactions. Other compounds cannot be effectively administered because they are not soluble in any known solvent that can be tolerated by patients in need of cancer prevention or treatment. As a result, these anti-cancer agents are unavailable for use in cancer prevention or treatment using conventional methods of administration.
While anti-cancer compounds are commonly administered by intravenous injection to patients in need of treatment, it is also known to inject cisplatin and carboplatin into the peritoneal cavity. A comparative study of intravenous versus intraperitoneal administration of cisplatin has been published by Alberts, et al. in the New England Journal of Medicine, 335, 1950-1955 (1996). Dedrick, et al., have published a pharmacokinetic rationale for the advantage of intraperitoneal versus intravenous administration of cisplatin in Cancer Treatment Reports, 62, 1-11 (1978).
Similarly, intraperitoneal delivery of cisplatin as an infusion is discussed in Principles of Clinical Pharmacology (Atkinson, et al., Academic Press 2001). To date, however, there do not appear to be any published reports of intraperitoneal delivery of suspensions of poorly water-soluble anti-cancer compounds.
SUMMARY OF THE INVENTION A composition comprising nanop articulates of at least one antimitotic drag, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
A composition comprising nanoparticulates of at least one antimitotic drug, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers in a suspension medium.
A method of administering intraperitoneally a composition comprising nanoparticulates of at least one antimitotic drug in a suspension medium, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
A method of administering intravenously a composition comprising nanoparticulates of at least one antimitotic drug in a suspension medium, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers. A composition comprising nanoparticulates of paclitaxel, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
A composition comprising nanoparticulates of paclitaxel, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers in a suspension medium.
A method of administering intraperitoneally a composition comprising nanoparticulates of paclitaxel in a suspension medium, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
A method of administering intravenously a composition comprising nanoparticulates of paclitaxel in a suspension medium, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
A composition comprising nanoparticulates of at least one antibiotic drug, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers. A composition comprising nanoparticulates of at least one antibiotic drug, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers in a suspension medium.
A method of administering intraperitoneally a composition comprising nanoparticulates of at least one antibiotic drug in a suspension medium, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
A method of administering intravenously a composition comprising nanoparticulates of at least one antibiotic drug in a suspension medium, where the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
BRIEF DESCRIPTION OF THE DRAWINGS The present embodiment of the invention is described in detail below with reference to the attached drawing figures, wherein:
FIG. 1 is a graph illustrating the results from a study of cancer bearing mice treated with nanoparticulate paclitaxel administered intravenously compared with controls and paclitaxel in Cremophor solution. FIG. 2 is a graph illustrating the results from a study of cancer bearing mice treated with nanoparticulates of paclitaxel administered intraperitoneally compared with controls and paclitaxel in Cremophor solution.
FIG. 3 is a graph illustrating the results from a study of cancer bearing mice treated with macroparticulates of paclitaxel, 20 to 60 microns in size, administered intraperitoneally compared with controls, paclitaxel in Cremophor solution and nanoparticulate paclitaxel administered intraperitoneally.
FIG. 4 is a photograph of the body wall of a cancer bearing mouse treated with the saline control. FIG. 5 is a photograph of the body wall of a cancer bearing mouse treated with 48 mg/kg of nanoparticulates of paclitaxel in suspension administered intraperitoneally.
FIG. 6 is a photograph of the diaphragm of a cancer bearing mouse treated with the saline control. FIG. 7 is a photograph of the diaphragm of a cancer bearing mouse treated with 48 mg/kg of nanoparticulates of paclitaxel in suspension administered intraperitoneally.
FIG. 8 is a photograph of an external view of a cancer bearing mouse treated with the saline control. FIG. 9 is a photograph of an external view of a cancer bearing mouse treated with 48 mg/kg of nanoparticulates of paclitaxel in suspension administered intraperitoneally.
FIG. 10 is a photograph of the kidneys of a cancer bearing mouse treated with the saline control. FIG. 11 is a photograph of the kidneys of a cancer bearing mouse treated with 48 mg/kg of nanoparticulates of paclitaxel in suspension administered intraperitoneally.
FIG. 12 is a photograph of the peritoneal organs of a cancer bearing mouse treated with the saline control. FIG. 13 is a photograph of the peritoneal organs of a cancer bearing mouse treated with 48 mg/kg of nanoparticulates of paclitaxel in suspension administered intraperitoneally.
DESCRIPTION OF THE PREFERRED EMBODIMENT OF THE INVENTION The present embodiment of the invention is generally directed to compositions comprising suspensions of poorly water soluble compounds recrystallized in nanoparticulate sizes ranging from 0.1 to 5 μ.m, and more preferably from 0.4 to 2 μm. In addition, the embodiment of the invention is directed to methods for preparation and administration of these compositions to a patient for prevention and treatment of disease states. In particular, the embodiment of the invention is directed to compositions comprising suspensions of poorly water-soluble compounds, such as antimitotics and antibiotics, in nanoparticulates and methods of prevention and treatment of chronic disease states, such as cancer, by intraperitoneal and intravenous administration of such compositions. Various processes are disclosed in U.S. Patent Nos. 5,833,891 and
6,113,795, which are incorporated by reference herein in their entireties, for producing particle sizes as small as 0.1 to 10 βm for compounds. Because particles that are smaller than one to two microns can pass through the smallest capillaries in the human body, it is desirable to determine whether suspensions of small size particles of anti-cancer compounds could be injected into the blood stream and produce a therapeutic effect without causing blood clotting or other undesirable side effects as a result of aggregation of the small particles into larger particles or aggregation of platelets on the surface of the particles.
Antimitotics as used herein, include, but are not limited to: paclitaxel; derivatives of paclitaxel; taxanes; epithilones, Vinca alkaloids, such as vinblastine, vincristine, vindesine, vinorelbine; camptothecin analogs; and epipodophyllotoxins, such as etoposide and teniposide. Poorly water-soluble antibiotics, include, but are not limited to, actinomycin D, mitomycin, daunorubicin, doxorubicin and idarubicin.
Poorly water soluble compounds, as used herein, are compounds that include: insoluble compounds that have <0.01 mg/ml solubility, very slightly soluble compounds that have 0.1-1 mg/ml solubility, slightly soluble compounds that have 1-10 mg/ml solubility and sparingly soluble compounds that have 10-33 mg/ml solubility. The compositions of the present embodiment of the invention may include other pharmaceutically acceptable ingredients, excipients and adjuvants. The nanoparticulate intraperitoneal delivery described in this application may ameliorate some of the side effects of administering a poorly water-soluble drug by allowing a lower dose to be delivered over a long period of time.
Example 1 In this example, using the process described in U.S. Patent Nos. 5,833,891 and 6,113,795, paclitaxel was recrystallized to an average particle size of about 700 nanometers and a particle size distribution such that greater than 95% of the particles were below one micron in size as determined by aerodynamic Time-of-Flight particle sizing. Four groups of mice that had previously been injected with cancerous cells and had developed ovarian cancer were treated with one of the following: 1) a phosphate buffered saline alone, used as a control 2) a Cremophor EL solution alone, used as a control 3) paclitaxel in Cremophor EL solution injected by intravenous (IV) administration, or 4) nanoparticulate paclitaxel suspended in phosphate buffered saline and injected by IV administration. The mice were injected with the treatment, comparative and control compositions on the fiftieth day after inoculation with cancer cells. Four doses of the compositions were injected every other day.
The saline control group survived for a maximum of 110 days post cancer cell injection and the Cremophor control group survived for a maximum of 113 days post cancer cell injection. By day 125, the last of the nanoparticulate paclitaxel group expired and the group injected with paclitaxel in Cremophor was 80% expired. There appears to be no statistical difference in overall survival of mice treated by IV with nanoparticulate paclitaxel in suspension and paclitaxel in Cremophor solution. The results of the IN injection study are shown in FIG. 1.
Notably, the mice survived the direct TV injection of the suspension of nanoparticulate paclitaxel in phosphate buffered saline without the need to add anticlotting agents such as heparin or agents such as surfactants or emulsifiers to prevent aggregation of the particles. While the preferred formulations would include these additional ingredients to further reduce the opportunity for clotting, the nanoparticulate paclitaxel did not appear to cause blockage or infarct of fine capillaries. Surprisingly, it was determined that IV injection of the nanoparticulate suspension of paclitaxel was as effective as the solution of paclitaxel in Cremophor EL in lengthening the survival time for mice inoculated with cancer cells. As a result, it may be possible to deliver a suspension of nanoparticulate paclitaxel intravenously with the same therapeutic effect as a solution of paclitaxel in Cremophor, but without the adverse effects of Cremophor.
Example 2
In this example, using the process described in U.S. Patent Nos.
5,833,891 and 6,113,795, paclitaxel was recrystallized to an average particle size of about 700 nanometers and a particle size distribution such that greater than 95% of the particles were below one micron in size as determined by aerodynamic Time-Of-Flight particle sizing. Eight groups of mice that had previously been injected with cancerous cells and had developed ovarian cancer were treated with one of the following: 1) a phosphate buffered saline alone, used as a control; 2) a Cremophor EL solution alone, used as a control; 3) paclitaxel in Cremophor EL solution, 12 mg/kg administered intraperotenel administration; 4) paclitaxel in Cremophor EL solution, 18 mg/kg injected by IP administration; 5) paclitaxel in Cremophor solution, 36 mg/kg injected by IP administration; 6) nanoparticulate paclitaxel suspended in phosphate buffered saline, 18 mg/kg administered intraperitoneally; 7) nanoparticulate paclitaxel suspended in phosphate buffered saline, 36 mg/kg administered intraperitoneally; or 8) nanoparticulate paclitaxel suspended in phosphate buffered saline, 48 mg/kg administered intraperitoneally. The mice were injected with the treatment, comparative, and control compositions on the fiftieth day after inoculation with cancer cells. Four doses of the compositions were injected every other day.
The longest surviving Cremophor control mouse lasted until 79 days post cancer cell injection. For the phosphate buffered saline control group, the last member of the control group expired on day 87. For the paclitaxel in Cremophor group, the last mouse survived up to day 99 for the 18 mg/kg dose and day 105 for the 12 mg/kg dose. The 36 mg/kg dosage group did not survive treatment. For the nanoparticulate paclitaxel in phosphate buffer saline group, administered intraperitoneally, the last mouse survived up to day 162 for the 18 mg/kg dose, day 181 for the 36 mg/kg does, and day 220 for the 48 mg/kg does. This represents a significant increase in survival in comparison to IV administration. The results of the intraperitoneal injection study are shown in FIG. 2.
It was determined that intraperitoneal injection of the suspension of paclitaxel nanoparticulates significantly lengthened the survival time of the mice in comparison to the intraperitoneal injection of solubilized paclitaxel in Cremophor. Further, as can be seen from FIG. 4-FIG. 13, the mouse treated with nanoparticulate paclitaxel, administered intraperitoneally, developed fewer cancerous tumors and spreading of the cancer was less aggressive than the cancer in the saline control mouse.
FIG. 4 is a photograph of the body wall of a mouse treated with the saline control showing numerous cancerous tumors. FIG. 5 shows the body wall of a mouse treated with 48 mg/kg of nanoparticulates of paclitaxel in suspension administered peritoneally and shows few, if any, cancerous tumors.
The diaphragm of a mouse treated with the saline control is depicted in Fig 6, where numerous cancerous tumors can be seen. The diaphragm of a mouse treated with 48 mg/kg of nanoparticulates of paclitaxel in suspension administered peritoneally, as shown in FIG. 7, does not show the same proliferation of cancerous tumors. FIG. 8 is a photograph of an external view of a mouse treated with the saline control where abdominal cavity of the mouse is distended from the spreading of the cancer and the accumulation of ascetic fluids. On the other hand, the external view of the mouse treated with nanoparticulates of paclitaxel in FIG. 9 is normal. FIG. 10 is a photograph of the kidneys with numerous cancerous tumors of a mouse treated with the saline control. FIG. 11 is a photograph of the healthy kidneys of a mouse treated with nanoparticulates of paclitaxel in suspension administered peritoneally. FIG. 12 is a photograph of the cancerous growths on the peritoneal organs of a mouse treated with the saline control, while FIG. 13 does not show any cancerous growths on the peritoneal organs of a mouse treated with nanoparticulates of paclitaxel. Example 3 In this example, nine groups of mice that had previously been injected with cancerous cells and had developed ovarian cancer were treated with one of the following: 1) a phosphate buffered saline alone, used as a control; 2) a Cremophor EL solution alone, used as a control; 3) paclitaxel in Cremophor EL solution, 18 mg/kg injected by IP administration; 4) macroparticulate paclitaxel, 18 mg/kg administered intraperitoneally; 5) macroparticulate paclitaxel , suspended in phosphate buffered saline, 36 mg/kg administered peritoneally; 6) macroparticulate paclitaxel suspended in phosphate buffered saline, 48 mg/kg administered intraperitoneally; 7) nanoparticulate paclitaxel suspended in phosphate buffered saline, 18 mg/kg administered intraperitoneally; 8) nanoparticulate paclitaxel suspended in phosphate buffered saline, 36 mg/kg administered intraperitoneally; or 9) nanoparticulate paclitaxel suspended in phosphate buffered saline, 48 mg/kg administered intraperitoneally. The mice were injected with the treatment, comparative, and control compositions on the fiftieth day after inoculation with cancer cells. Four doses of the compositions were injected every other day.
The longest surviving Cremophor control mouse lasted until 92 days post cancer cell injection. For the phosphate buffered saline control group, the last member of the control group survived until day 93. For the paclitaxel in Cremophor group, the maximal survival time was to day 115. For the macroparticulate (20-60 μm particle size) paclitaxel in phosphate buffer saline group, the maximal survival time was 137 days for the 18 mg/kg dose, 150 days for the 36 mg/kg dose and 151 days for the 48 mg/kg dose. For the nanoparticulate paclitaxel in phosphate buffer saline, the last member of the group expired on day 162 for the 48 mg/kg dosage group, day 179 for the 36 mg/lcg dosage group and day 205 for the 18 mg/kg dosage group. The survival time for mice treated with macroparticulate paclitaxel was greater than the survival time of mice treated with paclitaxel in Cremophor. However, the mice treated with nanoparticulate paclitaxel had the longest survival time. The results of this intraperitoneal injection study are shown in FIG. 3. The present embodiment of the invention has been described in relation to particular embodiments which are intended in all respects to be illustrative rather than restrictive. Alternative embodiments will become apparent to those skilled in the art to which the present invention pertains without departing from its scope. From the foregoing, it will be seen that this invention is one well adapted to attain all the ends and objects hereinabove set forth together with other advantages which are obvious and which are inherent to the compositions and methods of making and using such compositions herein disclosed. Since many possible embodiments may be made of the invention without departing from the scope thereof, it is to be understood that all matter herein set forth or shown in the accompanying drawing is to be interpreted as illustrative and not in a limiting sense.

Claims (30)

CLA S What is claimed is:
1. A composition comprising: nanoparticulates of at least one antimitotic drug, wherein the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
2. The composition of claim 1, wherein the antimitotic drug is selected from the group consisting of: paclitaxel, paclitaxel derivatives, taxanes, epithilones, Vinca alkaloids, camptothecin analogs, epipodophyllotoxins and combinations thereof.
3. The composition of claim 1, wherein the nanoparticulates have a particle size from 0.4 to 2 micrometers.
4. The composition of claim 2, wherein the antimitotic drug is a Vinca alkaloid.
5. The composition of claim 4 wherein the antimitotic drug is selected from the group consisting of: vinblastine, vincristine, vindesine and vinorelbine.
6. The composition of claim 2, wherein the amitotic drug is a camptothecin analog.
7. The composition of claim 2, wherein the antimitotic drug is an epipodophyllotoxin.
8. The composition of claim 2, wherein the antimitotic drug is a paclitaxel derivative.
9. The composition of claim 2, wherein the antimitotic drug is a taxane.
10. The composition of claim 7, wherein the antimitotic drug is selected from the group consisting of: etoposide and teniposide.
11. The composition of claim 1, further comprising: a suspension medium.
12. A method of administering the composition of claim 11.
13. The method of claim 12, wherein the composition is administered intravenously.
14. The method of claim 12, wherein the composition is administered intraperitoneally.
15. The composition of claim 11, wherein the suspension is phosphate buffered saline.
16. The composition of claim 11, further comprising anticlotting agents.
17. The composition of claim 11, further comprising surfactants.
18. A composition comprising: nanoparticulates of paclitaxel, wherein the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
19. The composition of claim 18, wherein the nanoparticulates have a particle size from 0.4 to 2 micrometers.
20. The composition of claim 18, further comprising: a suspension medium.
21. A method of administering the composition of claim 20.
22. The method of claim 21, wherein the composition is administered intravenously.
23. The method of claim 21, wherein the composition is administered intraperitoneally.
24. A composition comprising: nanoparticulates of an antibiotic drag, wherein the nanoparticulates have a particle size from 0.1 micrometer to 5 micrometers.
25. The composition of claim 24, wherein the nanoparticulates have a particle size from 0.4 to 2 micrometers
26. The composition of claim 24, wherein the antibiotic is selected from the group consisting of: actinomycin D, mitomycin, daunorubicin, doxorubicin and idarubicin.
27. The composition of claim 24, further comprising: a suspension medium.
28. A method of administering the composition of claim 27.
29. The method of claim 28, wherein the composition is administered intravenously.
30. The method of claim 28, wherein the composition is administered intraperitoneally.
AU2002362836A 2001-10-15 2002-10-15 Compositions and methods for delivery of poorly water soluble drugs and methods of treatment Expired AU2002362836B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US32929101P 2001-10-15 2001-10-15
US60/329,291 2001-10-15
PCT/US2002/032820 WO2003032906A2 (en) 2001-10-15 2002-10-15 Delivery of poorly soluble drugs

Publications (2)

Publication Number Publication Date
AU2002362836A1 true AU2002362836A1 (en) 2003-07-03
AU2002362836B2 AU2002362836B2 (en) 2008-05-22

Family

ID=23284727

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002362836A Expired AU2002362836B2 (en) 2001-10-15 2002-10-15 Compositions and methods for delivery of poorly water soluble drugs and methods of treatment

Country Status (9)

Country Link
US (2) US20030087837A1 (en)
EP (1) EP1478343B1 (en)
JP (1) JP2005508354A (en)
CN (1) CN1592607A (en)
AU (1) AU2002362836B2 (en)
CA (1) CA2461556A1 (en)
ES (1) ES2625340T3 (en)
IL (2) IL160927A0 (en)
WO (1) WO2003032906A2 (en)

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090004277A1 (en) * 2004-05-18 2009-01-01 Franchini Miriam K Nanoparticle dispersion containing lactam compound
GT200500310A (en) 2004-11-19 2006-06-19 ORGANIC COMPOUNDS
SA06270147B1 (en) 2005-06-09 2009-12-22 نوفارتيس ايه جي Process for the Synthesis Of 5-(methyl-1h-imidazol-1-yl)-3-(trifluoromethyl)-benzenamine
MX2008014953A (en) * 2006-05-26 2009-03-05 Bayer Healthcare Llc Drug combinations with substituted diaryl ureas for the treatment of cancer.
WO2010005726A2 (en) 2008-06-16 2010-01-14 Bind Biosciences Inc. Therapeutic polymeric nanoparticles with mtor inhibitors and methods of making and using same
WO2010005723A2 (en) 2008-06-16 2010-01-14 Bind Biosciences, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8318211B2 (en) * 2008-06-16 2012-11-27 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising vinca alkaloids and methods of making and using same
WO2010068866A2 (en) 2008-12-12 2010-06-17 Bind Biosciences Therapeutic particles suitable for parenteral administration and methods of making and using same
EA201100765A1 (en) 2008-12-15 2012-04-30 Бинд Биосаиэнсис Long-term circulation nanoparticles
CN102811743B (en) 2009-12-11 2015-11-25 佰恩德治疗股份有限公司 The stabilization formulations of lyophilizing treatment granule
ES2780156T3 (en) 2009-12-15 2020-08-24 Pfizer Therapeutic compositions of polymeric nanoparticles with high glass transition temperature or high molecular weight copolymers
KR20150056618A (en) 2012-09-17 2015-05-26 바인드 쎄라퓨틱스, 인크. Process for preparing therapeutic nanoparticles
EP3116547B1 (en) 2014-03-14 2019-06-05 Pfizer Inc Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using same
JP6921759B2 (en) * 2015-06-04 2021-08-18 クリティテック・インコーポレイテッド Collection device and usage
WO2017049083A2 (en) 2015-09-16 2017-03-23 Dfb Soria, Llc Delivery of drug nanoparticles and methods of use thereof
EP3854381A1 (en) 2016-04-04 2021-07-28 Crititech, Inc. Methods for solid tumor treatment
EP3512560A4 (en) * 2016-09-13 2020-05-27 Rasna Research Inc. Dactinomycin compositions and methods for the treatment of myelodysplastic syndrome and acute myeloid leukemia
KR102658256B1 (en) 2017-03-15 2024-04-16 디에프비 소리아, 엘엘씨 Topical therapy for the treatment of skin malignancies using taxane nanoparticles.
WO2018170210A1 (en) 2017-03-15 2018-09-20 Dfb Soria, Llc Topical therapy for the treatment of vulvar intraepithelial neoplasia (vin) and genital warts using nanoparticles of taxanes
WO2018170207A1 (en) 2017-03-15 2018-09-20 Dfb Soria, Llc Topical therapy for the treatment of skin keratoses using nanoparticles of taxanes
WO2018227037A1 (en) 2017-06-09 2018-12-13 Crititech, Inc. Treatment of epithelial cysts by intracystic injection of antineoplastic particles
CN115919815A (en) 2017-06-14 2023-04-07 克里蒂泰克公司 Methods of treating pulmonary diseases
EP3691631A1 (en) 2017-10-03 2020-08-12 Crititech, Inc. Local delivery of antineoplastic particles in combination with systemic delivery of immunotherapeutic agents for the treatment of cancer
KR20200132963A (en) 2018-03-16 2020-11-25 디에프비 소리아, 엘엘씨 Topical therapy for the treatment of cervical intraepithelial neoplasia (CIN) and cervical cancer using taxane nanoparticles
US20190365698A1 (en) 2018-05-31 2019-12-05 Crititech, Inc. Use of Antineoplastic Agents to Stimulate the Immune System for Treatment of Cancer
KR20210016511A (en) 2018-05-31 2021-02-16 크리티테크, 인크. Methods of isolating tumor specific immune cells from a subject for adoptive cell therapy and cancer vaccines
WO2020072090A1 (en) 2018-10-03 2020-04-09 Crititech, Inc. Use of antineoplastic agents to stimulate the immune system for production of tertiary lymphoid structures (tls)

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4489055A (en) * 1978-07-19 1984-12-18 N.V. Sopar S.A. Process for preparing biodegradable submicroscopic particles containing a biologically active substance and their use
US5707634A (en) * 1988-10-05 1998-01-13 Pharmacia & Upjohn Company Finely divided solid crystalline powders via precipitation into an anti-solvent
US5399363A (en) * 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
US5145684A (en) * 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
CA2086874E (en) * 1992-08-03 2000-01-04 Renzo Mauro Canetta Methods for administration of taxol
US5439686A (en) * 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
US5916596A (en) 1993-02-22 1999-06-29 Vivorx Pharmaceuticals, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US6447796B1 (en) * 1994-05-16 2002-09-10 The United States Of America As Represented By The Secretary Of The Army Sustained release hydrophobic bioactive PLGA microspheres
DE4440337A1 (en) 1994-11-11 1996-05-15 Dds Drug Delivery Services Ges Pharmaceutical nanosuspensions for drug application as systems with increased saturation solubility and dissolution rate
US6106866A (en) * 1995-07-31 2000-08-22 Access Pharmaceuticals, Inc. In vivo agents comprising cationic drugs, peptides and metal chelators with acidic saccharides and glycosaminoglycans, giving improved site-selective localization, uptake mechanism, sensitivity and kinetic-spatial profiles, including tumor sites
US5834025A (en) * 1995-09-29 1998-11-10 Nanosystems L.L.C. Reduction of intravenously administered nanoparticulate-formulation-induced adverse physiological reactions
JP3986086B2 (en) 1996-03-01 2007-10-03 ザ ユニバーシティ オブ カンザス Particle precipitation method and coating method using near-critical and supercritical anti-solvents
US5833891A (en) * 1996-10-09 1998-11-10 The University Of Kansas Methods for a particle precipitation and coating using near-critical and supercritical antisolvents
US5874029A (en) * 1996-10-09 1999-02-23 The University Of Kansas Methods for particle micronization and nanonization by recrystallization from organic solutions sprayed into a compressed antisolvent
US6441025B2 (en) * 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US6458939B1 (en) * 1996-03-15 2002-10-01 Millennium Pharmaceuticals, Inc. Compositions and methods for the diagnosis, prevention, and treatment of neoplastic cell growth and proliferation
WO1998017656A1 (en) * 1996-10-24 1998-04-30 Institute Armand-Frappier A family of canadensol taxanes, the semi-synthetic preparation and therapeutic use thereof
KR100530601B1 (en) * 1996-12-25 2005-11-23 니폰 가야꾸 가부시끼가이샤 Fine cisplatin powder and process for the production thereof
CA2628857C (en) * 1996-12-30 2011-09-13 Batelle Memorial Institute Formulation and method for treating neoplasms by inhalation
KR100923172B1 (en) 1997-06-27 2009-10-22 아브락시스 바이오사이언스, 엘엘씨 Novel Formulations of Pharmacological Agents
US6106566A (en) * 1998-01-29 2000-08-22 Micron Electronics, Inc. Upgradable electronic module and system using same
US6113795A (en) * 1998-11-17 2000-09-05 The University Of Kansas Process and apparatus for size selective separation of micro- and nano-particles
US6270806B1 (en) * 1999-03-03 2001-08-07 Elan Pharma International Limited Use of peg-derivatized lipids as surface stabilizers for nanoparticulate compositions
US6267989B1 (en) * 1999-03-08 2001-07-31 Klan Pharma International Ltd. Methods for preventing crystal growth and particle aggregation in nanoparticulate compositions
EP1178786A4 (en) * 1999-05-21 2006-03-01 American Bioscience Inc Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US6620351B2 (en) 2000-05-24 2003-09-16 Auburn University Method of forming nanoparticles and microparticles of controllable size using supercritical fluids with enhanced mass transfer
US7425668B2 (en) 2006-08-30 2008-09-16 Alternative Seed Strategies Llc Field pea cultivar CDC 0007

Similar Documents

Publication Publication Date Title
US8221779B2 (en) Compositions and methods for the delivery of poorly water soluble drugs and methods of treatment
AU2002362836A1 (en) Compositions and methods for delivery of poorly water soluble drugs and methods of treatment
EP2508207B1 (en) Nanoparticles loaded with chemotherapeutic antitumoral Drug
US20150342872A1 (en) Use of Paclitaxel Particles
JP2002537333A (en) Synergistic antitumor composition
CN109771663B (en) Preparation and application of acid-responsive anticancer nano-drug
KR20180098702A (en) Compositions and methods of delivery of pharmacological agents
CN100594902C (en) Nano micelle preparation of Catharanthus roseus alkaloids antineoplastic drugs with coating of phospholipid derived from polyethylene glycol
CN102292109A (en) Polymer micelle composition for treatment of resistant cancer cells
CN110538328A (en) polypeptide compound, drug-loaded nanoparticle, preparation method of polypeptide compound and drug-loaded nanoparticle, drug composition and application of drug composition
CN102198085B (en) Triblock copolymer loaded taxane medicaments micelle and freeze-drying preparation, and preparation method and application thereof
CN113768878B (en) Elemene cabazitaxel double-targeting bionic liposome and preparation method and application thereof
DE60025494T2 (en) EPOTHILONE COMPOSITIONS
WO2009121997A2 (en) Pegylated nanoparticles containing a biologically active molecule and use thereof
CA2285391A1 (en) Methods and compositions for treatment of ovarian cancer
CN102078301A (en) Taxotere nano preparation carried by albumin and phospholipid and method preparing same
CA3199407A1 (en) Pharmaceutical composition comprising insoluble camptothecin compound-containing nanoparticle for treatment of cancer and combination therapy thereof
CN102600084A (en) Rubescensin-galactosylation chitosan nano particle preparation and preparation method thereof
KR20180001112A (en) Layer by layer assembly of albumin conjugate and pharmaceutical composition using the same
CN111568893A (en) Docetaxel-resveratrol-co-loaded nano long-circulating liposome as well as preparation method and application thereof
US20210220295A1 (en) Treatment of cancer by guanidinium derivatives
KR20180059346A (en) Liposome for treating cancer containing both linalool nanoparticles and anticancer drug