CA2086874E - Methods for administration of taxol - Google Patents

Methods for administration of taxol

Info

Publication number
CA2086874E
CA2086874E CA002086874A CA2086874A CA2086874E CA 2086874 E CA2086874 E CA 2086874E CA 002086874 A CA002086874 A CA 002086874A CA 2086874 A CA2086874 A CA 2086874A CA 2086874 E CA2086874 E CA 2086874E
Authority
CA
Canada
Prior art keywords
paclitaxel
infusion
dosage
cancer
accordance
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.)
Expired - Fee Related
Application number
CA002086874A
Other languages
French (fr)
Other versions
CA2086874C (en
CA2086874A1 (en
Inventor
Renzo Mauro Canetta
Elizabeth Eisenhauer
Marcel Rozencweig
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.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=25448992&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2086874(E) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Publication of CA2086874A1 publication Critical patent/CA2086874A1/en
Publication of CA2086874C publication Critical patent/CA2086874C/en
Application granted granted Critical
Publication of CA2086874E publication Critical patent/CA2086874E/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents

Abstract

Paclitaxel dosages of about 135 mg/m2 or greater are administered via infusions of less than 6 hours duration; the method makes it possible to provide paclitaxel infusions on an out-patient basis to patients who do not otherwise require hospitalization. In a preferred embodiment, about 135 mg/m2 of paclitaxel in a cremaphor emulsion is infused over a 3-hour duration, following patient pretreatment with steroids, antihistamines, and H2-receptor antagonists sufficient to prevent fatal anaphylactic-like reactions, and preferably sufficient to reduce the occurrence of severe anaphylactic-like reactions in greater than 90% of patients treated. In an alternative embodiment, between 135 mg/m2 and about 175 mg/m2 of paclitaxel is provided in a 3-hour infusion, following pretreatment to minimize hypersensitivity responses. A method for rechallenging patients with paclitaxel episodes of acute hypersensitivity reactions is also disclosed, thus enabling patients to continue paclitaxel therapy who would otherwise be deprived of treatment.

Description

2 n ~ ~ %-7 4 _ METHODS FOR ADMINISTRATION OF TAXOL

FIELD OF THE lNV~NllON
The present invention is directed to methods of cancer treatment with paclitaxel, and more particularly is directed to improvements in the administration of paclitaxel in the treatment of cancer.

BACKGROUND OF THE lNV~N-llON
Paclitaxel is a naturally occurring compound which has shown great promise as an anti-cancer drug. For example, paclitaxel has been found to be an active agent against drug-refractory ovarian cancer by McGuire et al. See "Taxol*: A
Unique Anti-Neoplastic Agent With Significant Activity Against Advanced Ovarian Epithelial Neoplasms," Ann. Int. Med., 111, 273-279 (1989).
Unfortunately, paclitaxel has extremely low solubility in water, which makes it difficult to provide a suitable dosage form. In fact, in Phase I clinical trials, severe allergic reactions were caused by the emulsifiers administered in conjunction with paclitaxel to compensate for paclitaxel's low water solubility; at least one patient's death was caused by an allergic reaction induced by the emulsifiers. Dose limiting toxicities include neutropenia, peripheral neuropathy, and hypersensitivity reactions.
Brown et al, in "A Phase I Trial of Taxol Given By A 6-Hour Intravenous Infusion" J of Clin Oncol, Vol. 9, No. 7, pp.

1261-1267 ~(July, 1991) report on a Phase I Trial in which paclitaxel was provided as a 6-hour IV infusion every 21 days without premedication. 31 patients received 64 assessable ~Trademark ~9 ~ O ~ 6 ~-7 4 ...._ courses of paclitaxel. One patient had a severe (or acute) hypersensitivity reaction, which required discontinuation of the infusion and immediate treatment to save the patients life. Another patient experienced a hypersensitivity reaction, but it was not so severe as to require discontinuing the infusion. Myelosuppression was dose-limiting, with 2 fatalities due to sepsis. Non-hematologic toxicity was of Grade 1 and 2, except for one patient with Grade 3 mucositis and 2 patients with Grade 3 neuropathy. The neuropathy consisted of reversible painful parestnesias, requiring discontinuation of paclitaxel in two patients. Four partial responses were seen (3 in patients with non-small-cell lung cancer, and one in a patient with adenocarcinoma of unknown primary). The maximum tolerated dose reported was 275 mg/m2, and the recommended Phase II starting dose was 225 mg/m2. The incidence of hypersensitivity reaction was reported to be schedule-dependent, with 6 to 24-hour infusions of drug having a 0~ to 8~ incidence of hypersensitivity reactions. It was also reported that hypersensitivity reactions persist with or without premedication despite prolongation of infusion times.
Since these Phase I studies were conducted on terminally ill patients suffering from a variety of cancers, the efficacy of the paclitaxel treatments could not be determined.
In a study by Kris et al., paclitaxel formulated with Cremaphor EL in dehydrated alcohol was given as a 3-hour IV
infusion every 21 days, with the administered dosage ranging from 15 to 230 mg/m2 in nine escalation steps. Kris et al.
concluded that l'with the severity and unpredictability of the hypersensitivity reactions, further usage of paclitaxel is not indicated with this drug formulation on this administration schedule. Il See Cancer Treat. Rep., Vol. 70, No. 5, May 1986.
B

~ n 8 ~ ~-7 ~ ~
Since early trials using a bolus injection or short (1-3 hour) infusions induced anaphylactic reactions or other hypersensitivity responses, further studies were carried out in which paclitaxel was administered only after premedication with steroids (such as dexamethasone), antihistamines (such as diphenhydramine), and H2-antagonists (such as cimetidine or ranitidine), and the infusion time was extended to 24 hours in an attempt to eliminate the most serious allergic reactions.
Various Phase I and Phase II study results have been published utilizing 24-hour infusions of paclitaxel with maximum total dosages of 250 mg/m2, generally with the course being repeated every 3 weeks. Patients were pretreated with dexamethasone, diphenhydramine, and cimetidine to offset allergic reactions.
See Einzig, et al., "Phase II Trial of Taxol in Patients with Metastatic Renal Cell Carcinoma," Cancer Investigation, 9(2) 133-136 (1991), and A.B. Miller et al., "Reporting Results of Cancer Treatment," Cancer, Vol 47, 207-214 (1981).
Koeller et al, in "A Phase I Pharmacokinetic Study of Taxol Given By a Prolonged Infusion Without Premedication,"
Proceedings of ASCO, Vol. 8 (March, 1989), recommends routine premedication in order to avoid the significant number of allergic reactions believed to be caused by the cremaphor (polyethoxylated castor oil) vehicle used for paclitaxel infusions. Patients received dosages ranging from 175 mg/m2 to 275 mg/m2.
Wiernik et al, in "Phase I Clinical and Pharmacokinetic Study of Taxol," Cancer Research, 47, 2486-2493 (May 1, 1987), also report the administration of paclitaxel in a cremaphor vehicle by IV infusion over a 6-hour period in a Phase I
study. Grade 3-4 hypersensitivity reactions incurred in 4 of 13 courses. The starting dose for the study was 15 mg/m2 B

~0 ~6874 (one-third of the lowest toxic dose in dogs). Doses were escalated, and a minimum of 3 patients were treated at each dose level until toxicity was identified, and then 4-6 patients were treated at each subsequent level. The study concluded that neurotoxicity and leucopenia were dose-limiting, and the recommended Phase II trial dose was 250 mg/m2 with premedication.
Other exemplary studies on paclitaxel include: Legha et al, "Phase II Trial of Taxol in Metastatic Melanoma," Vol. 65 (June 1990) pp. 2478-2481; Rowinsky et al, "Phase I and Pharmacodynamic Study of Taxol in Refractory Acute Leukemias,"
Cancer Research, 49, 4640-4647 (August 15, 1989); Grem et al, "Phase I Study of Taxol A~m'n' stered as a Short IV Infusion Daily For 5 Days," Cancer Treatment Reports, Vol. 71 No. 12, (December, 1987); Donehower et al., "Phase I Trial of Taxol in Patients With Advanced Cancer," Cancer Treatment Reports, Vol.
71, No. 12, (December, 1987); Holmes et al, "Phase II Study of Taxol in Patients (PT) with Metastatic Breast Cancer (MBC),"
Proceedings of the American Society of Clinical Oncology, Vol.
10, (March, 1991, pp. 60. See also Suffness, "Development of Antitumor Natural Products at the National Cancer Institute,"
Gann Monograph or Cancer Research, 31 (1989) pp. 21-44 (which recommends that paclitaxel only be given as a 24-hour infusion).
Weiss et al., in "Hypersensitivity Reactions from Taxol,"
Journal of Clinical Oncology Vol. 8, No. 7 (July 1990) pp.
1263-1268, reported that it was difficult to determine a reliable overall incidence of hypersensitivity reactions, HSRs, because of the wide variations in paclitaxel doses and schedules used, and the unknown degree of influence that changing the infusion schedule and using premedication has on B

~0 8B8-7~
HSR incidents. For example, of five patients who received paclitaxel in a 3-hour infusion at greater than 190 mg/m2 with no premedication, three had reactions, while only one out of 30 patients administered even higher doses over a 6-hour infusion with no premedication had a reaction. Therefore, this suggests that prolonging the infusion to beyond 6 hours is sufficient to reduce HSR incidents. Nevertheless, Weiss et al. found that patients receiving 250 mg/m2 of paclitaxel administered via a 24-hour infusion still had definite HSRs.
Thus, while prolonging drug infusion to 6 or 24-hours may reduce the risk for an acute reaction, this conclusion cannot be confirmed, since 78~ of the HSR reactions occurred within ten minutes of initiating the paclitaxel infusion, which indicates that the length of time planned for the total infusion would have no bearing. Further, concentration of paclitaxel in the infusion may also not make a difference since substantial numbers of patients had reactions to various small paclitaxel dosages. Finally, not only is the mechanism of paclitaxel HSR unknown, it is also not clear whether paclitaxel itself is inducing HSRs, or if the HSRs are due to the excipient (Cremophor (trade-mark) EL; Badische Anilin und Soda Fabrik AG [BASF], Ludwigshafen, Federal Republic of Germany). Despite the uncertainty as to whether or not premedication had any influence on reducing the severity or number of HSRs, prophylactic therapy was recommended, since there is no known danger from its use.
The conflicting recommendations in the prior art concerning whether premedication should be used to avoid hypersensitivity reactions when using prolonged infusion durations, and the lack of efficacy data for infusions done over a six hour period has led to the use of a 24-hour ~2 ~ a~ 7 ~

infusion of high doses (above 170 mg/m2) of paclitaxel in a Cremaphor EL emulsion as an accepted cancer treatment protocol.
Although it appears possible to minimize the side effects of administering paclitaxel in an emulsion by use of a long infusion duration, the long infusion duration is inconvenient for patients, and is expensive due to the need to monitor the patients for the entire 6 to 24-hour infusion duration.
Further, the long infusion duration requires that patients spend at least one night in a hospital or treatment clinic.
Thus, it is highly desirable to develop a paclitaxel infusion protocol which would allow for recipients to be treated on an out-patient basis. Since paclitaxel infusions are generally preceded by premedication, and require post-infusion monitoring and record keeping, it is highly desirable that the infusion duration not exceed 6 hours, yet the infusion dosage should provide the patient sufficient paclitaxel to have an anti- neoplastic effect, while not exceeding dose-limiting toxicities. It is also desirable to minimize premedication since this increases patient discomfort and increases the expense and duration of treatment.
Even if infusion duration cannot be shortened, it is also desirable to avoid the high dosages of paclitaxel presently believed necessary to have an anti-neoplastic effect, which induce a variety of adverse side-effects, including respiratory distress, cardiovascular irregularities, flu-like symptoms, gastrointestinal distress, hematologic complications, genitourinary effects, neuropathy, alopecia, and skin rashes.

~ . .

~ O ~ 6 ~-7 ~
,, Further, due to the extremely limited supply of paclitaxel, and the high dosage requirement for each patient, the demand for paclitaxel greatly exceeds the supply.
Therefore, it is highly desirable to reduce paclitaxel dosages, if possible, to both extend the supply of paclitaxel and reduce the toxic side effects of paclitaxel. It is also highly desirable to decrease the time required to administer paclitaxel to patients to minimize patient discomfort and expense.
Thus, there is a need for a new method of administration of paclitaxel which utilizes less paclitaxel and/or requires less infusion time.
Therefore, it is a primary object of the present invention to provide a new method for administering paclitaxel over a shorter period of time than the present 6 to 24-hour infusion protocols, while minimizing toxic effects induced by the administration of paclitaxel.
It is another object of the present invention to provide a new method for administration of paclitaxel which reduces the amount of paclitaxel administered to a patient, without sacrificing the anti-neoplastic effects desired by administering paclitaxel.
It is yet a further object of the present invention to provide a new method for administration of paclitaxel which utilizes both lower dosages of paclitaxel and shorter infusion periods, without sacrificing the anti-neoplastic benefits of the administration of paclitaxel.

~ O' ~ 7 ~ ~
SUMMARY OF THE lNV~L. ~lON
These and other objects of the present invention are accomplished by new methods of administration of paclitaxel.
In a preferred embodiment, paclitaxel infusions are provided over a duration of less than six hours, preferably about three hours, utilizing dosages of between about 135 mg/m2 and about 275 mg/m2, preferably between about 135 mg/~ and about 175 mg/m2, after patients are pretreated to alleviate or minimize hypersensitivity responses. In a preferred embodiment, anti-neoplastic effects are achieved in patients suffering fromcancer through administration of about 135 mg/m2 paclitaxel administered via a 3-hour infusion following premedication to reduce or eliminate hypersensitivity responses. These results are surprising in view of the conventional understanding that a bolus injection or short (1-3 hour infusions) will induce anaphylactic reactions or other hypersensitivity responses, and that only premedication coupled with extension of the infusion time to 6-24 hours would reduce or eliminate the most serious allergic reactions.
In an alternative embodiment, 135 mg/m2 paclitaxel is administered via a 24-hour infusion following premedication.
Preferably, patients are premedicated with steroids, antihistamines, and H2-antagonists sufficient to at least prevent an anaphylactoid shock capable of causing patient death in greater than 95% of cancer patients treated, and avoid acute hypersensitivity reactions in greater than 90% of cancer patients treated.
To ameliorate myelosuppression associated with paclitaxel administration, particularly associated with high dosages of paclitaxel, granulocyte-colony stimulating factor (G-CSF) is given as a daily subcutanious injection started 24-hours after B

~0 ~8-74 the completion of a paclitaxel infusion. With the use of G-CSF, paclitaxel dosages of up to 275 mg/m2 can be safely administered using the methods of the present invention.
Other aspects of the present invention include a method of administering paclitaxel to a patient suffering from cancer by monitoring certain clinical parameters, and temporarily haulting paclitaxel administration when infusion related toxicities exceed safe limits. After the parameters return to near baseline, therapy is restarted, preferably with 72 hours.
In a preferred embodiment, premedication is given intravenously prior to restarting the infusion. For patients suffering severe HSRs near the end of, or after, a paclitaxel infusion, the HSR is treated, and the patient is rechallenged during the next course by providing premedication intravenously, followed by using a modified 24-hour infusion, in which the infusion is initiated at about one fourth the planned infusion 24-hour rate; if there are no severe HSR
symptoms after about six hours, the infusion is continued at the 24-hour rate. Additional aspects include the administration of multiple courses of paclitaxel over regular periods of time, such as at approximately 21-day intervals, or when adverse effects of a previous course or infusion substantially subside.

DETAILED DESCRIPTION OF THE lNv~NllON
Despite the conventional understanding that it is necessary to infuse patients over a 24-hour period with high dosages of paclitaxel (greater than 170 mg/m2) following premedication to minimize or eliminate hypersensitivity responses, while obtaining the desired anti-neoplastic effect, it has been surprisingly discovered that paclitaxel can be ~n~874 safely administered to cancer patients via infusions lasting less than 6 hours at dosages of about 135 mg/m2 to about 175 mg/m2. In a preferred embodiment, paclitaxel is administered via an infusion having a duration of about three hours, with a paclitaxel dosage of about 135 mg/m2 or about 175 mg/m2. Of great significance is a surprising discovery that the short term infusion causes less myelosuppression, which leads to a lower incidence of infections and fever episodes (e.g., febrile neutropenia). Following the preferred infusion schedules of the present invention provides an objective response rate of greater than 10~ for patients suffering from epithelial ovarian carcinoma, and preferably an objective response rate of 14~ or greater for groups of at least 150 patients suffering from ovarian carcinoma.
The surprising discovery that paclitaxel could be safely administered via a short infusion (e.g., less than 6 hours and preferably over about 3 hours) means that it will now be possible to administer paclitaxel on an out-patient basis, saving patients the time and expense of yet another hospitalization while improving patient quality of life.
It has also been surprisingly discovered that lower paclitaxel dosages, such as about 135 mg/m2 can be administered via infusions lasting about 3 hours to about 28 hours, and still be antineoplastically effective.
EXP~RTM~TAL PROTOCOL
A multi-center, randomized comparative study of paclitaxel in patients suffering from ovarian carcinoma which had been pretreated with platinum was carried out. The high degree of success, greater than 10~ objective response, to treatment by patients suffering from drug refractory ovarian ~Q 86~-7~
carcinoma is truly astonishing, since responses to drug refractory ovarian carcinoma are extremely uncommon. While experimental data is provided herein for the successful treatment of ovarian carcinoma with paclitaxel using the present treatment protocol, it is to be understood that the treatment protocol can be used for the treatment of other forms of cancer with paclitaxel, such as melanoma, renal cell carcinoma, and other cancers which are treatable with paclitaxel. The present invention provides an improvement in the treatment of all types of cancer which can be treated with paclitaxel, since by use of the administration protocol of the present invention, lower toxicities and/or less time is required than that associated with prior art protocols for administering antineoplastically effective amounts of paclitaxel.
Patients with platinum pretreated recurrent ovarian cancer were randomized to a 135 or 175 mg/m2 dose arm, administered as a 3 hour or 24 hour IV infusion, which was repeated every three weeks. Responses were evaluated every two cycles. A11 four treatment groups were matched for age, performance status, previous treatments, and time from last platinum therapy. If a patient showed a complete response, CR, paclitaxel infusions were continued for four cycles post CR; patients showing a partial response, PR, had paclitaxel infusions continued until relapse, or treatment was stopped after four cycles post PR stabilization. For a stable disease, paclitaxel infusions were stopped after a maximum of 10 cycles or earlier if unacceptable toxicity was present; if disease progressed, PD, patients were taken off the study.

7 ~

The study included four arms as indicated below:

ARM INFUSION DURATION DOSAGE
A 24 hours 175 mg/m B 3 hours 175 mg/m C 24 hours 135 mg/m D 3 hours 135 mg/m Patients with ovarian carcinoma included in the study had been treated with and were unresponsive to a minimum of one but no more than two platinum-containing chemotherapy regimens, and were required to have shown progression during or relapse after their last regimen of chemotherapy. Patients were treated every 21 days if allowed by toxicity. Granulocyte count and platelet count were monitored; provided granulocyte count was at least 1500 cells/cmm and platelet count was at least 100,000 cells/cmm, paclitaxel was administered.

ADMINISTRATION QF PACLITAXEL
Prior to initiating an infusion, a resussitation/
emergency cart was placed outside the infusion room and remained there during the first hour of infusion. An emergency drug tray having all the necessary drugs, etc. was set up in the room in the event of an acute hypersensitivity reaction. Oxygen and suction equipment are provided at the bedside of each patient along with a 3-hour or 24-hour observation sheet (depending on the arm), and a fluid balance sheet.
Nitroglycerin tubing is required for the IV equipment because the Cremaphor EL (polyethoxylated castor oil) leaches plasticizer from regular IV tubing made of polyvinylchloride.

~ ~ 8 6 ~ ~ 4 .
Glass or polyolefin containers were used for storing all cremophor containing solutions. All paclitaxel-containing solutions were infused with in-line filtration using a microporous filter, preferably having a pore size not greater than 0.22 microns.
In order to minimize acute hypersensitivity reactions, patients were premedicated according to the protocol with dexamethasone or an e~uivalent (20 mg taken os at home, 12 hours and 6 hours prior to paclitaxel infusion).
Diphenhydramine (or equivalent antihistamine), 50 mg IV; and ranitidine (or equivalent H-2 receptor blocker), 50 mg IV, were provided 30 minutes prior to the paclitaxel infusion.
Paclitaxel was supplied by BRISTOL-MYERS SQUIBB as a concentrated sterile solution for IV administration. Each 5 ml vial contained 6 mg/ml paclitaxel in polyethoxylated castor oil (Cremaphor EL) 50~ in dehydrated alcohol, USP 50~. While an emulsion of paclitaxel in polyethoxylated caster oil in dehydrated alcohol is utilized as a vehicle in a preferred emodiment, it is contemplated that other pharmaceutically acceptable vehicles for paclitaxel may be used.
Paclitaxel was administered, after dilution in dextrose or saline solution, as a continuous infusion in two 500 mL
glass bottle of 5~ dextrose (D5W) or normal saline (NS) over 3 hours, or in two 500 mL glass bottles over 12 hours each for the 24-hour infusion. The first 18 mL of IV solution were infused at a fast rate (300 mL/hour) via the previously primed line to ensure that the paclitaxel had reached the end of the IV tubing. This procedure enables the accurate accessment of the patient receiving the paclitaxel.
A nurse remained with each patient for the first hour of each infusion. Temperature, pulse, blood pressure, and B

~ a ~ 6 ~-7 4 ._ respiration were taken preinfusion, every 15 minutes for the first hour, every 30 minutes for the next 2 hours, hourly for the next 4 hours, and every other hour until the end of the infusion (for patients receiving the 24-hour infusion). Other observations were also documented, such as rash/redness of the skin, hives, vomiting, nausea, and any other symptoms including neuropathy and skin rashes.
Another aspect of the present invention is that patients suffering from hypersensitivity reactions to paclitaxel infusions can be retreated with paclitaxel following management of the hypersensitivity reaction. In a preferred embodiment, patients experiencing hypersensitivity reactions were taken off paclitaxel. Hypersensitivity reactions were treated by administration of diphenhydramine (50 mg IV) with administration of epinephrine 0.35-0.5 cc s.c. every 15-20 minutes until reaction subsided or until a total of six dosages of epinephrine were given. IV fluids were administered when hypotension did not respond to epinephrine.
Likewise, if wheezing did not stop in response to epinephrine, 0.35 cc of nebulized albuterol solution was administered.
To prevent recurrent or ongoing allergy manifestation, 125 mg methylprednisolone was administered via IV.
In a preferred embodiment for rechallenging with paclitaxel patients who had experienced a hypersensitivity reaction (other than hypotension requiring pressor therapy, angioedema respiratory distress requiring bronchodilation therapy, and generalized urticaria), the following protocol was followed. A11 rechallenge patients received a prolonged 24-hour infusion, even if the hypersensitivity reaction occurred during a 3-hour infusion. Dosage reductions in subsequent treatment courses were mandated for all patients 2~ ~6~'74 having (1) a granulocyte count of less than 500 cells/cmm; (2) a platelet count of less than 50,000 cells/cmm for seven days or more; (3) febrile neutropenia; (4) infection; (5) hemmorage, (6) mucositis with vesiculation and/or ulcers; or (7) vomiting appearing despite anti-emetic premedication.
Paclitaxel treatment was discontinued in patients having (1) intolerable paresthesias and/or marked motor loss (neurological toxicity of World Health Organization, WHO, grade greater than 2); (2) symptomatic bradycardia or heart block of any degree or other arrhythmias; and (3) other major organ toxicity of WHO grade greater than 2.
If less than 75~ of a total paclitaxel dose was infused prior to a hypersensitivity reaction, retreatment preferably occurs within 72 hours of cessation of paclitaxel infusion, and the amount of paclitaxel used for retreatment equals the originally planned paclitaxel dose less the amount infused before the paclitaxel infusion was stopped due to the hypersensitivity reaction. A non-limiting embodiment of a preferred rechallenge procedure follows:
A. A steroid is given. For example, 8 mg dexamethasone is given intraveneously 24, 18, 12, and 6 hours prior to paclitaxel administration.
B. An antihistamine is given 30 minutes prior to paclitaxel, for example, 50 mg IV diphenhydramine.
C. An H2 receptor antagonist is given 30 minutes prior to paclitaxel infusion, for example, ranitidine 50 mg IV.
D. Paclitaxel is administered in the 24-hour infusion volume, but at one quarter the planned rate for a 24-hour IV
infusion during the first 6 hours. If no reaction is noted, the rate is increased to normal infusion speed. Thus, the entire infusion time is somewhat longer than 24 hours because .~

' 7 ~
of the slow infusion during the first 6 hours. Thus, by way of non-limiting example, total infusion time would be approximately 28.5 hours.
E. If a hypersensitivity reaction sufficient to discontinue the paclitaxel infusion recurs during the rechallenge procedure, the patient is taken off paclitaxel.
However, if a patient is successfully rechallenged without recurrence of a hypersensitivity reaction severe enough to require discontinuation of the infusion, the rechallenge procedure is continued.

EFFICACY AND SAFETY
In order to confirm the efficacy and safety of the paclitaxel administration method of the present invention, patients with histologically confirmed ovarian carcinoma, who had shown progression during or relapse after platinum-containing therapy, were administered paclitaxel according to one of the four previously described Arms.
Patients ranged between 18 and 75 years in age with an Eastern Cooperative Oncology Group, ECOG, performance status of 0, 1, or 2, and had to have recovered from all toxicities of previous treatment. Further, no prior chemotherapy or radiotherapy were allowed within four weeks prior to entry into the study (patients were also not entered into the study if there had been less than six weeks since prior mitomycin, nitrosureas, or high-dose carboplatin therapy). An adequate baseline bone marrow function was required, which was defined as a granulocyte count 2 2,000 cells/cmm and a platelet count 2 100, 000 cells/cmm. Adequate hepatic and renal functions were required with normal hepatic function defined as total billirubin 2 1. 5 times upper normal value and normal renal V

~ O ~ 6 ~-7 4 ' , ~
function defined as serum creatinine 2 1. 5 times upper normal value. Patients with abdominal adenocarcinoma, a previous history of atrial or ventricular arrhythmias, congestive heart failure, myocardial infarction within six months preceding randomization, complete bowel obstruction, a pre-existing neuropathy > WHO grade 2, active infection, prior allergic reaction to drugs containing Cremaphor, or other serious medical condition were excluded from the study.
Eligible patients were randomized according to a computerized randomization log. During therapy, hematology data (hemoglobin, white blood cell count, granulocytes and platelets) were collected, and toxicity assessments were continuously made. After each cycle, a physical history update was recorded, as well as tumor measurement, performance status (ECOG), hematology, chemistry (serum creatinine, billirubin, alkaline phosphatase, SGOT (AST), CA125) and a toxicity assessment. A quality of life assessment was also made after each cycle, and every two months until six months after treatment stopped.
The efficacy and safety of the improved method for administration of paclitaxel of the present invention was based on 159 patients randomized into the four treatment Arms previously described. Of the 159 patients, 157 patients received at least one dose of paclitaxel and were evaluated for both safety and efficacy.
528 courses of paclitaxel were given to 157 patients distributed over the 4 treatment Arms. The median number of courses received was 3 (range 1 - 8), which is equivalent to a median of 9 weeks on study. Overall 27 of 157 (17~) patients required a dose reduction, mainly for neutropenia with less than 500 cells/cmm for more than 7 days duration.

7 ~

.,.,_ Paclitaxel administration was discontinued for 2 patients during the second course. One of the 27 patients was retreated at a reduced dose level, and dosage was re-escalated after one course at a reduced dose level. Overall, 11~ of the paclitaxel courses were administered at reduced dosage.
Dose reduction was required in the 2 Arms using long term infusion more frequently than in Arms using the short term infusion. This is surprising considering that a more concentrated dosage of paclitaxel is given during the short infusion than during the long infusion.
The time between courses ranged from 17 to 49 days, with the median number of days between courses being 21. The following table lists the various premedications used in the various treatment Arms:

EXAMPLES OF PREMEDICATIONS USED

H-2 Receptor Blocker: Steroid: Antihistimine:
Ranitidine Dexamethasone Dyphenhydramine 25Famotidine Hydrocortisone Clemastine Cimetidine Prednisone Chlorpheniramine Chlorphen~m;ne Dimethindene maleate Promethazine In a preferred embodiment, at least one of each of the H-2 receptor blockers, steroids and antihistimines is utilized.

2 Q ~ 7 ~

' , By way of non-limiting examples, a combination of prednisone and hydrocortisone, or dexamethasone and hydrocortisone could be used in combination with at least one of the antihistamines and at least one of the H- 2 receptor blockers.
22 patients achieved an objective response (CR or PR).
Thus, the overall objective response rate is 14~ (22/157) for this study. However, 17 patients were not evaluated and another 5 patients were unevaluable. If these latter patients are excluded, the objective response rate is 16~ (22/135).
Further, 51 patients with stable disease were continued on paclitaxel and may later meet the criteria for an objective response (Note that an average of only three courses of paclitaxel yielded the present results). Thus, use of the present method for administration of paclitaxel produces at least a 14~ overall objective response rate for 157 patients.
This is an astonishing result, since all of the patients were considered drug refractory. It is also remarkable that 3 out of 46 (7~) of these patients who had progressed on previous platinum containing chemotherapy responded to paclitaxel.
20 Overall, responses to paclitaxel occurred in 13~ of patients (14/114) who were considered resistant to platinum therapy (i.e., progression on therapy or relapse within six months).
Further, 52~ of patients (24/46) with disease truly refractory to platinum, and 53~ of patients (16-36) with an early relapse after platinum, achieved a stabilization of their disease.
15~ of the platinum resistant patients who had received one previous regimen of platinum responded to paclitaxel, versus 10~ of the platinum resistant patients who had received two previous regimens of platinum, corresponding to 19 and 30 23 ~, respectively.

_ Of 159 patients, only one died of paclitaxel related toxicity (less than 1~).

HEMATOLOGIC TOXICITY
Another aspect of the present invention is the reduction in hematologic toxicity associated with the treatment of cancer with paclitaxel. The 157 patients who received paclitaxel had blood counts performed weekly. White blood cell (WBC) counts, absolute neutrophil count (ANC), platelet counts, and hemoglobin (Hb) concentration were the primary variables used to evaluate treatment related myelosuppression.
The World Health Orgination, WHO, grades of nadir for the entire study population of 157 patients during course 1 are presented in the table below. Leukopenia and neutropenia were the most frequent and severe hemotologic adverse effects observed during the first course of treatment. 53~ (83/157) of the patients had Grade III or IV neutropenia, while 39~
(61/157) had Grade III or IV leukopenia. Severe thrombocytopenia was observed in only two patients (1~) during course 1.
Of particular significance is that Grade IV neutropenia was reported almost five times more frequently in the patients treated with the 24-hour paclitaxel infusion than the patients treated with a 3-hour paclitaxel infusion.

B

Myelotoxicity Course 1 WHO Grade Per Treatment Arm (n=45) (n=29)(n=44) (n=39) (n=157) n (%) n (%)n (%) n (%) n (%) WBC Count 01 (2) 11 (38)4 (9) 15 (38) 31 (20) I5 (11) 6 (21)5 (11) 13 (33) 29 (18) II11 (25) 7 (24)11 (25) 7(18) 36 (23) III22 (49) 5 (17)20 (46) 3 (8) 50 (32) IV 6 (13) 0 - 4 (9) 1 (3) 11 (7) ANC Count 02 (5) 11 (38)6 (14) 18 (46) 37 (24) I1 (2) 2 (7)2 (15) 8 (21) 13 (8) II5 (11) 3 (10)7 (16) 8 (21) 23 (15) III8 (18) 6 (21)6 (14) 4 (10) 24 (15) IV28 (63) 7 (24)23 (52) 1 (3) 59 (38) Platelet Count 039 (87) 29 (100)43 (98) 36 (92) 147 (94) I 3 (7) 0 1 (2) 2 (5) 6 (4) II 1 (2) 0 0 1 (3) 2 (1) III 1 (2) 0 0 0 1 (1) IV 1 (2) 0 0 0 1 (1) As is clear from Table 2, grade 4 neutropenia was reported almost 5 times more frequently in patients treated with the 24-hour paclitaxel infusion compared to patients treated by a 3-hour infusion. 58~ (51/88) of the patients treated with the 24-hour paclitaxel infusion had grade 4 neutropenia in comparison to 12~ (8/68) of the patients treated by a 3-hour infusion. When the incidence of grade 3 and grade 4 are pooled, it is clear that severe leukopenia occurs more frequently in patients treated with a 24-hour paclitaxel infusion than with a 3-hour infusion.

~ n ~ ~ ~ 7 ~
,~", With reference to Table 3 below, analysis of median values for nadir count confirms the severity of paclitaxel induced neutropenia, especially in the two 24-hour treatment Arms.

Myelotoxicity Course 1 = Nadir Counts, Per Treatment Arm (n=45) (n=29) (n=44) (n=39) (n=157) WBC Count 20 - Median 1,600 3,300 1,900 3,700 2,400 - Range 300-7,300 1,400-13,900 500-7,800 980-6,850 300-13,900 ANC Count - Median 330 1,390 470 1,930 840 - Range 0-664 210-11,770 0-555 360-6,290 0-11,770 Platelet Count - Median 168,000 285,000 236,500 298,000 226,000 - Range 11,000 144,000 87,000 65,000 11,000 - 345,000 - 688,000 - 710,000 - 749,000 - 749,000 (1) Cell counts expressed in number of cells/cmm.

Further, anemia of any grade occurred in 47~ (26/55) of patients with normal baseline Hb values who had been treated with a paclitaxel long-term infusion, and anemia occurred in only 28~ of patients treated with the short-term infusion. In summary, neutropenia and leukopenia were the most frequent and severe hematologic adverse effects during the study period, with 63~ of the patients displaying severe neutropenia (WHO
Grade III and IV) during at least one course. The incidence of severe neutropenia in the long-term infusion versus the short-term infusion Arms was 85~ versus 32~. The incidents of severe neutropenia in the high dose Arms was 74~ (55/74) B

2 Q ~ 7 ~
versus 52~ (43/83) in the low dose Arms. Thus, it is clear that both reducing the dosage and the infusion time will lower hematologic toxicity; however, reducing the infusion to 3 hours from 24 hours appears to have a greater impact on reducing toxicity than reducing the paclitaxel dosage from about 175 mg/m2 to 135 mg/m2.
Those patients suffering from myelotoxicity can be treated with colony stimulating factors, CSFs. In a preferred - embodiment, patients are given granulocyte colony stimulating factors in an amount sufficient to be effective in either reducing myelotoxicity or increasing the rate of recovery from myelotoxicity. Preferably, the CSFs are provided in accordance with the method taught by Sarosy et al. in "Phase I Study of Taxol and Granulocyte Colony-Stimulating Factor In Patients With Refractory Ovarian Cancer," Journal of Clinical Oncology, Vol. 10, No. 7, (July, 1992) pp. 1165-1170. In a preferred embodiment, G-CSF (available from Amgen, Inc. of Thousand Oaks, CA) can be self-administered subcutaniously on a daily basis (thus allowing for treatment on an outpatient basis), with injections beginning 24 hours after the completion of a paclitaxel infusion. Preferably a G-CSF dose of about 10 ~g/kg/d is used, and G-CSF injection continues until there is convincing evidence of bone marrow recovery from paclitaxel-induced nadir. Convincing evidence of bone marrow recovery includes a white blood cell count of 10,000 cells/mm3 and a platelet count of more than 50,000/m~ . The use of G-CSF enables higher doses of paclitaxel to be used, and allows for certain patients suffering severe myelosuppression to be continued on paclitaxel treatment whereas, in the past, such patients may not have been allowed to continue on paclitaxel.

B

~n ~68-76~ ~
..._ HYPERSENSITIVITY REACTIONS
35~ (57/157) of the patients exhibited some type of hypersensitivity reaction. Only two hypersensitivity reactions (2 of 157 patients or 1~) were reported which required acute therapeutic intervention.
Table 4 below lists the number and percentage of patients demonstrating hypersensitivity reactions per treatment Arm and also provides the type of HSR per course.

Hyper~ensitivity Reactions (HSR) Per Treatment Arm n (%) n (~) n (~) n (~) n ~) Per Patient Number of patients analyzed 45 29 44 39 157 - Patients reporting a HSR 6 (36) 12 (41) 18 (41) 11 (28) 57 (36) Per Course Number of courses analyzed 163 92 141 132 528 - Courses with a reported HSR 46 (28) 23 (25) 44 (31) 23 (17) 136(26) Tvpe of HSR
Per Course - Flushing 32 16 40 15 103 - Rash 3 6 6 6 21 - Hypotension 10 1 1 1 13 - Dyspnea - - 3 2 5 - Edema face 1 1 - - 2 - Eye disorder - - - 2 2 - Pruritis - - - 2 2 - Headache - Arrhythmia - Hypertension - Tachychardia - Nausea/Vomiting - Chest pain (1) More than 1 sign and symptom may be experienced in each course.

.~
, 8 7 ~ ~

, The most frequent symptoms were flushing, mainly confined to the face, following by skin rash, hypotension, and dyspnea.
Of the two patients suffering severe hypersensitivity reactions, one patient was not rechallenged with paclitaxel.
The other patient suffering a severe hypersensitivity reaction received a 135 mg/m2 24-hour infusion first course of paclitaxel without experiencing a severe hypersensitivity reaction. Due to WHO Grade IV neutropenia for more than 7 days, the scheduled dose for course 2 was reduced by 20~.
During the second course, the delivery of less than 1 ml of paclitaxel infusion induced tachychardia and shortness of breath, requiring the infusion to be stopped. The patient developed an extreme general flush, with a heart rate of 150 per minute (regular) and the blood pressure was 150/100. The allergic reaction resolved completely after the administration of 50 mg benadryl IV and 35 mg adrenalin s.c. The infusion of paclitaxel was restarted after an interruption of 70 minutes at the regular infusion rate without further problems.
Another patient encountered a hypersensitivity reaction, which prompted the investigator to interrupt the infusion.
However, this incident did not qualify as a significant HSR.
During a first course, the patient received 135 mg/m2 during a 24-hour infusion. Course 1 was administered uneventfully.
Paclitaxel infusion during course 2 was interrupted after 1 minute (less than 1 mg of paclitaxel) due to dyspnea, flushing, and nausea, which was treated with 5 mg of chlorphenamine. The patient received another 250 mg hydrocortisone, and the paclitaxel infusion was recommenced 70 minutes after. The total infusion time was 28 hours, 15 minutes with no further occurrence of hypersensitivity ~ a 8 B ~ 7 6~
reaction. Course 3 was administered uneventfully with normal premedication.
Further examples of adverse reactions are discussed later.

PERIPHERAL NEUROPATHY
80 of 157 patients (51~) experienced some sign or symptom of peripheral neurotoxicity. The incidents in the high dose arms was 66~ (49/74), while the incidents in the low dose arms was 37~ (31/83). The incidents of peripheral neuropathy in the long-term (24-hour) infusion was 48~ (43/89), while the incidents of peripheral neuropathy in the short-term infusion arms was 54% (37/68).
Prior to paclitaxel treatment, 75~ (118/157) of patients were asymptomatic; of these, 44 patients (37~) developed some peripheral neuropathy symptoms. Overall, 54 of 157 patients (34~) developed, or had worse peripheral neuropathy symptoms, as can be seen by Table 5 below.
The substantial reduction in peripheral neurotoxicity symptoms (PNS) in patients receiving lower dosages of paclitaxel allows for more flexibility in treating patients, since lower paclitaxel dosages over longer infusion periods can be used for patients suffering from PNS while higher doses and/or shorter infusion periods can be used for patients not suffering from PNS.

B

20 86~ ~
.~., Peripheral Neuropathy Symptoms (PNS) and Paclitaxel Do~ing By Treatment Arm n = 45 n = 29 n = 44 n = 39 n = 157 n (~) n (~) n (~) n (~) n (~) Number of Patients Who Developed or Worsened PNS 20 (44) 18 (62) 7 (16) 8 (21) 54 (34) First Occurrence/
Worsening of PNS
- By Courses - Course 1 9 7 3 4 23/157 (15) - Course 2 8 9 2 _ 19/143 (13) - Course 3 1 1 1 1 4/95 (4) - Course 4 And more 2 1 1 3 7/69 (10) - By Cumulative Dose of Paclitaxel - ~ 200 mg/m2 9 7 3 4 23 - 200-401 mg/m2 8 9 3 1 22 - 401-600 mg/m2 2 1 1 2 4 - ~ 600 mg/m2 1 1 1 2 5 It is clear that peripheral neuropathy symptoms (PNS) are reduced when a dosage of about 135 mg/m2 paclitaxel is provided rather than a dosage of about 17 5 mg/m2.
Further examples of adverse drug reactions, and the procedures used to continue treatment follow.
Patient BB-2 was allocated to the 135 mg/m2~ 3 hour arm.
Shortly after beginning her second cycle of treatment, she developed asymptomatic bradyarrhythmia characterized by short periods of AV block or sinus pauses accompanied by some ventricular extrasystoles. She had some extrasystoles in B

cycle 1 as well. There was no prior cardiac history and subsequent investigations were normal. She had a pacemaker inserted and was retreated with a third cycle. The bradyarrhythmia appeared to have a definite relation to paclitaxel, and the patient recovered with treatment.
Patient BB-3 was allocated to the 175 mg/m2, 3 hour arm.
Shortly after beginning cycle 2, she developed a hypersensitivity reaction, characterized by generalized urticaria, diaphoresis, and dyspnea. The infusion was interrupted and she recovered quickly after treatment with epinephrine and antihistamine. She was retreated using the re-challenge amendment discussed above. The HSR appeared to be definitely related to paclitaxel, and the patient recovered with treatment.
Patient DF-l was randomized to the 135 mg/m2, 24 hour arm. During her first treatment, she had 2 episodes of asymptomatic bradycardia, during which the infusion was interrupted for durations lasting 3 and 5 minutes, respectively. She also developed a mild skin rash. However, the infusion was completed. The bradycardia appeared related to paclitaxel, and the patient recovered.
Patient IM-7 was allocated to the 135 mg/m2, 24 hour infusion, and experienced a non-significant HSR during cycle 2. Her first cycle of treatment had been uneventful; however, 5 minutes after the 2nd treatment began, she became flushed and dyspneic, with nausea (no vomiting). The infusion was stopped, 5 mg chlorpheniramine was given and symptoms rapidly resolved. The patient was re-treated according to the retreatment protocol discussed earlier with no problem, aside from minor facial rash. The HSR appeared to be related to paclitaxel.

~ Q ~ 4 Patient MP-7 was allocated to the 135 mg/m2, 24 hour infusion. About 30 minutes after cycle 1 began, she became hot, flushed, and slightly dyspneic (BP 114/80, pulse 112).
Paclitaxel was stopped, she was given diphenhydramine, and the reaction resolved immediately. The patient was re-treated according to the re-treatment protocol without event. Thus, the HSR appears related to paclitaxel.
Patient VA-30 was randomized to the 135 mg/m2, 24 hour arm. During her second treatment, she developed flushing and a sense of tightness in her throat. The infusion was stopped for a short while, and repeat doses of steroid, antihistamine and ranitidine were given. Paclitaxel was restarted with no further problems; this indicates that the HSR was caused by paclitaxel.
15The success of the use of the new paclitaxel infusion protocol of the present invention in the treatment of ovarian cancer makes it readily apparent that anti-neoplastically - effective dosages of paclitaxel can be infused over much shorter time periods than was previously believed possible, without inducing severe hypersensitivity reactions or inducing fatal anaphylactic shock. Thus, it is contemplated that the infusion protocol of the present invention may be utilized to treat solid tumors and leukemias, such as but not limited to lung cancer, breast cancers, and ovarian cancers. It is to be understood that treatment of different forms of cancer may require the adjustment of the paclitaxel dosage to have optimal efficacy.
The foregoing clearly establishes that paclitaxel is both safe and effective in the treatment of cancer, such as ovarian cancer, when administered according to the protocol of the present invention. In particular, by use of a 3-hour infusion fi ~ 7 ~
of about 135 mg/m2 paclitaxel, following premedication, a substantial reduction results in the frequency of myelotoxicity and neuropathy associated with the administration of paclitaxel to patients suffering from cancer. Further, patients who exhibit severe hypersensitivity reactions can be rechallenged with paclitaxel after treating the HSR symptoms by use of an infusion of about 24 hours or greater, preferably using a dosage of about 135 mg/m2 to about 175 mg/m2. Preferably, colony stimulation factors are administered to assist in ameliorating myelosuppression.
The use of lower dosages of paclitaxel to achieve antineoplastic results will allow for more patients to be treated with the present limited supply of paclitaxel.
Further, depending upon the toxicities noted in a patient during treatment with paclitaxel according to the present protocol, the duration of infusion can be extended or shortened, or the paclitaxel dosage can be reduced or increased, thus providing more flexibility in treating cancer with paclitaxel. Further, patients capable of handling higher doses of paclitaxel can be administered up to about 275 mg/m2;
should the patient encounter severe toxicity, such as a severe neuropathy, the protocol of the present invention allows for reducing the dosage.
From the above teachings, it is readily apparent that many modifications and variations of the present invention are possible. It is to be therefore understood that the invention may be practiced than as otherwise specifically described.

B

Claims (46)

1. Use of paclitaxel for the manufacture of an infusion preparation for effecting an antitumor response in a patient suffering from cancer, wherein said infusion preparation is formulated for administration of a dosage of from about 135 mg/m2 to about mg/m2 paclitaxel over a duration of less than 6 hours.
2. A use in accordance with claim 1, wherein said infusion preparation is formulated suitable for an infusion duration of about 3 hours.
3. A use in accordance with claim 1, wherein said infusion preparation is formulated for administration of a paclitaxel dosage of about 135 mg/m2 to about 175 mg/m2.
4. A use in accordance with claim 1, wherein said infusion preparation is formulated for administration of a paclitaxel dosage of about 135 mg/m2.
5. A use in accordance with claim 1, wherein said infusion preparation is useful in effecting an anticancer response in a patient suffering from solid tumors or leukemias.
6. A use in accordance with claim 1, wherein the infusion preparation is useful in effecting an antitumor response in a patient suffering from a cancer selected from the group consisting of ovarian, lung, and colon.
7. The use of paclitaxel for the manufacture of an infusion preparation for effecting an antitumor response in a patient suffering from ovarian cancer, wherein said preparation is formulated for administration to said patient at a dosage of about 135 mg/m2 paclitaxel over a duration of infusion of less than 6 hours.
8. The use of paclitaxel for the manufacture of an infusion preparation for effecting an antitumor response in a patient suffering from lung cancer, wherein said preparation is formulated for administration to said patient at a dosage of about 135 mg/m2 paclitaxel over a duration of infusion of less than 6 hours.
9. The use of paclitaxel for the manufacture of an infusion preparation for effecting an antitumor response in a patient suffering from colon cancer, wherein said preparation is formulated for administration to said patient at a dosage of about 135 mg/m2 paclitaxel over a duration of infusion of less than 6 hours.
10. The use of paclitaxel for the manufacture of an infusion preparation for repeat administration to patients suffering from cancer wherein the preparation is formulated such that it is suitable for administration in accordance with the use of claim 1, and suitable for repeat administration in accordance with the use of claim 1 within about 21 days following the prior administration.
11. The use of paclitaxel for the manufacture of an infusion preparation for effecting an antitumor response in a patient suffering from cancer wherein said preparation is formulated for administration of a paclitaxel dosage of from about 135 mg/m2 to about 175 mg/m2 over a duration of infusion of about 24 hours.
12. A use in accordance with claim 11, wherein said preparation is useful for effecting an anticancer response in patients suffering from solid tumors or leukemias.
13. A use in accordance with claim 11, wherein the preparation is formulated such that it is suitable for infusion of a paclitaxel dosage of about 135 mg/m2 to about 175 mg/m2.
14. A use in accordance with claim 11, wherein the preparation is formulated such that it is suitable for infusion of a paclitaxel dosage of about 135 mg/m2.
15. The use of paclitaxel for the manufacture of an infusion preparation for repeat administration for the treatment of cancer wherein the preparation is suitable for administration to a patient suffering from cancer consistent with the use of claim 9, and suitable for repeat administration in accordance with the use of claim 9 within about 21 days following the prior infusion.
16. A use in accordance with claim 11, wherein said cancer is selected from the group consisting of lung cancer, breast cancer, ovarian cancer, and colon cancer.
17. The use of paclitaxel for the manufacture of an infusion preparation for the treatment of ovarian cancer, wherein said preparation is formulated for effecting an infusion of about 135 mg/m2 paclitaxel over a period of about 24 hours to a patient suffering from ovarian cancer.
18. Use of paclitaxel at a dosage of from about 135 mg/m2 to about 175 mg/m2 over a duration of less than 6 hours for effecting an anti-tumour response in a patient suffering from cancer.
19. A use in accordance with claim 18, wherein the duration is over about 3 hours.
20. A use in accordance with claim 18, wherein said dosage is of about 135 mg/m2.
21. A use in accordance with claim 18, wherein said use is to effect an anti-cancer response in a patient suffering from solid tumours or leukemias.
22. A use in accordance with claim 18, wherein said use is to effect an anti-tumour response in a patient suffering from a cancer selected from the group consisting of ovarian, lung and colon.
23. Use of paclitaxel at a dosage of about 135 mg/m2 over a duration of less than 6 hours for effecting an anti-tumour response in a patient suffering from ovarian cancer.
24. Use of paclitaxel at a dosage of about 135 mg/m2 over a duration of less than 6 hours for effecting an anti-tumour response in a patient suffering from lung cancer.
25. Use of paclitaxel at a dosage of about 135 mg/m2 aver a duration of less than 6 hours for effecting an anti-tumour response in a patient suffering from colon cancer.
26. Use of paclitaxel at a first dosage of from about 135 mg/m2 to about 175 mg/m2 over a duration of less than 6 hours and at a second dosage identical to the first dosage, about 21 days following said first dosage, for effecting an anti-tumour response in a patient suffering from cancer.
27. Use of paclitaxel at a dosage of from about 135 mg/m2 to about 175 mg/m2 over a duration of about 24 hours for effecting an anti-tumour response in a patient suffering from cancer.
28. A use in accordance with claim 27 to effect an anti-cancer response in a patient suffering from solid tumours or leukemias.
29. A use in accordance with claim 27, wherein the dosage is of about 135 mg/m2.
30. Use of paclitaxel at a first dosage of about 135 mg/m2 over a duration of less than 6 hours and at a second dosage identical to the first dosage, about 21 days following said first dosage, for effecting an anti-tumour response in a patient suffering from cancer.
31. A use in accordance with claim 27, wherein said cancer is selected from the group consisting of lung cancer, breast cancer, ovarian cancer and colon cancer.
32. The use of paclitaxel as claimed in any one of claims 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 18, 19, 20, 21, 22, 23, 24, 25, 26 and 30 in which said paclitaxel is intended for use in a patient who has been premeditated with a medicament that reduces or eliminates hypersensitivity reactions.
33. The use of paclitaxel at a dosage of from 135 mg/m2 to 175 mg/m2 over a duration of 3 hours in an intravenous infusion for effecting an anti-tumour response in a patient suffering from cancer in which said paclitaxel is intended for use in a patient who has been premedicated with a medicament that reduces or eliminates hypersensitivity reactions.
34. A use in accordance with claim 33 to effect an anti-tumour response in a patient suffering from breast cancer.
35. A use in accordance with claim 33 wherein premedication is with a medicament selected from the group consisting of steroids, antihistamines, H2 receptor antagonists and combinations thereof.
36. The use in accordance with any of claims 33, 34 or 35 in which the dosage is 135 mg/m2.
37. The use in accordance with any of claims 33, 34 or 35 is when the dosage is 175 mg/m2.
38. The use of paclitaxel as claimed in any one of claims 14, 17 or 29 to reduce peripheral neurotoxicity symptoms in said patients.
39. An infusion preparation of paclitaxel for use at a dosage of 135 mg/m2 to mg/m2 over a duration of 3 hours for effecting an anti-tumour response in a patient suffering from cancer where the patient is premedicated with a medicament that reduces or eliminates hypersensitivity reactions.
40. An infusion preparation in accordance with claim 39 in which the dosage is mg/m2.
41. An infusion preparation in accordance with claim 39 is when the dosage is mg/m2.
42. An infusion preparation in accordance with any of claims 39, 40 or 41 wherein said premedication is with a medicament selected from the group consisting of steroids, antihistamines, H2 receptor antagonists and combinations thereof.
43. Use of paclitaxel for the manufacture of an infusion preparation for the treatment of cancer, in combination with an agent for the treatment of anaphylactic-like reactions and/or an agent for reducing myleotoxicity in accordance with an infusion protocol comprising an infusion duration not exceeding 6 hours and a paclitaxel dosage of from 135 mg/m2 to 175 mg/m2.
44. The use of paclitaxel in accordance with claim 43 wherein the infusion duration is 3 hours.
45. The use of paclitaxel in accordance with claim 43 wherein the dosage is mg/m2.
46. The use of paclitaxel in accordance with claim 43wherein the dosage is 175 mg/m2.
CA002086874A 1992-08-03 1993-01-07 Methods for administration of taxol Expired - Fee Related CA2086874E (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US92362892A 1992-08-03 1992-08-03
US07/923,628 1992-08-03

Publications (3)

Publication Number Publication Date
CA2086874A1 CA2086874A1 (en) 1994-02-04
CA2086874C CA2086874C (en) 1998-09-01
CA2086874E true CA2086874E (en) 2000-01-04

Family

ID=25448992

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002086874A Expired - Fee Related CA2086874E (en) 1992-08-03 1993-01-07 Methods for administration of taxol

Country Status (23)

Country Link
US (5) US5641803A (en)
EP (3) EP0783885A3 (en)
JP (1) JP2848760B2 (en)
AT (1) ATE152910T1 (en)
AU (3) AU3216893A (en)
BE (1) BE1006497A5 (en)
CA (1) CA2086874E (en)
CH (1) CH686868A5 (en)
DE (2) DE69310634T2 (en)
DK (2) DK65293A (en)
ES (2) ES2105926B1 (en)
FR (1) FR2694192B1 (en)
GB (1) GB9311740D0 (en)
GR (2) GR1001617B (en)
IE (1) IE80620B1 (en)
IL (1) IL105879A (en)
IT (1) IT1276041B1 (en)
MY (1) MY109862A (en)
NL (1) NL9301086A (en)
NZ (1) NZ245795A (en)
PT (1) PT101289A (en)
SE (1) SE513082C2 (en)
ZA (1) ZA93686B (en)

Families Citing this family (91)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6753006B1 (en) 1993-02-22 2004-06-22 American Bioscience, Inc. Paclitaxel-containing formulations
US20070117862A1 (en) * 1993-02-22 2007-05-24 Desai Neil P Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US5716981A (en) * 1993-07-19 1998-02-10 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
USRE38424E1 (en) * 1994-11-21 2004-02-10 The United States Of America As Represented By The Department Of Health And Human Services Method for treating taxol side-effects with G-CSF
US6441025B2 (en) * 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
PT932399E (en) 1996-03-12 2006-05-31 Pg Txl Co Lp PRO-PHOSPHOLIC CAPSULES
US20030157187A1 (en) * 1996-12-02 2003-08-21 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating or preventing inflammatory diseases
US6495579B1 (en) 1996-12-02 2002-12-17 Angiotech Pharmaceuticals, Inc. Method for treating multiple sclerosis
US6515016B2 (en) 1996-12-02 2003-02-04 Angiotech Pharmaceuticals, Inc. Composition and methods of paclitaxel for treating psoriasis
US6458373B1 (en) 1997-01-07 2002-10-01 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
US20030087954A1 (en) * 1997-01-07 2003-05-08 Sonus Pharmaceuticals, Inc. Method of treating bladder carcinoma using a Taxane/Tocopherol formulation
US20030105156A1 (en) * 1997-01-07 2003-06-05 Nagesh Palepu Method for administration of a taxane/tocopherol formulation to enhance taxane therapeutic utility
US6727280B2 (en) * 1997-01-07 2004-04-27 Sonus Pharmaceuticals, Inc. Method for treating colorectal carcinoma using a taxane/tocopherol formulation
GB9705903D0 (en) 1997-03-21 1997-05-07 Elliott Gillian D VP22 Proteins and uses thereof
WO1998051369A1 (en) 1997-05-13 1998-11-19 Edwards Stuart D Global medical treatment method and apparatus
CN100462066C (en) * 1997-06-27 2009-02-18 美国生物科学有限公司 Novel formulations of pharmacological agents, method for preparation thereof and method for use thereof
EP2272504A3 (en) 1997-06-27 2014-02-26 Abraxis BioScience, LLC Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
HUP9701554D0 (en) 1997-09-18 1997-11-28 Human Oltoanyagtermeloe Gyogys Pharmaceutical composition containing plazma proteins
AU1297899A (en) * 1997-10-31 1999-05-24 Walter Reed Army Institute Of Research Method of inhibiting side effects of pharmaceutical compositions containing amphiphilic vehicles or drug carrier molecules
US6683100B2 (en) 1999-01-19 2004-01-27 Novartis Ag Organic compounds
CA2320182C (en) * 1998-02-05 2008-11-25 Novartis Ag Compositions containing organic compounds
US6194181B1 (en) * 1998-02-19 2001-02-27 Novartis Ag Fermentative preparation process for and crystal forms of cytostatics
US7030155B2 (en) * 1998-06-05 2006-04-18 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
US6121029A (en) 1998-06-18 2000-09-19 Novartis Ag Genes for the biosynthesis of epothilones
US7314637B1 (en) 1999-06-29 2008-01-01 Neopharm, Inc. Method of administering liposomal encapsulated taxane
US6461637B1 (en) * 2000-09-01 2002-10-08 Neopharm, Inc. Method of administering liposomal encapsulated taxane
US6017948A (en) * 1998-10-30 2000-01-25 Supergen, Inc. Water-miscible pharmaceutical compositions
US6472420B2 (en) * 1998-11-12 2002-10-29 Cell Pathways, Inc. Method for treating a patient with neoplasia by treatment with a paclitaxel derivative
US6235776B1 (en) * 1998-11-12 2001-05-22 Cell Pathways, Inc. Method for treating a patient with neoplasia by treatment with a paclitaxel derivative
WO2000076556A2 (en) * 1999-06-11 2000-12-21 Neorx Corporation High dose radionuclide complexes for bone marrow suppression
TW422706B (en) 1999-07-01 2001-02-21 Wang Iz Rung An oil-in-water emulsion type of paclitaxel
US7094885B2 (en) * 1999-07-11 2006-08-22 Neorx Corporation Skeletal-targeted radiation to treat bone-associated pathologies
WO2001024763A2 (en) 1999-10-01 2001-04-12 Immunogen, Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
US20030054977A1 (en) * 1999-10-12 2003-03-20 Cell Therapeutics, Inc. Manufacture of polyglutamate-therapeutic agent conjugates
US6136846A (en) 1999-10-25 2000-10-24 Supergen, Inc. Formulation for paclitaxel
US6828346B2 (en) * 1999-10-25 2004-12-07 Supergen, Inc. Methods for administration of paclitaxel
JP2001247459A (en) 2000-03-03 2001-09-11 Oakland Uniservices Ltd Combination therapy for cancer
US20020077290A1 (en) * 2000-03-17 2002-06-20 Rama Bhatt Polyglutamic acid-camptothecin conjugates and methods of preparation
US6569459B2 (en) 2000-04-10 2003-05-27 Teva Pharmaceutical Industries, Ltd. Method of administration of paclitaxel-plasma protein formulation
WO2002009700A1 (en) 2000-07-28 2002-02-07 Cancer Research Technology Limited Cancer treatment by combination therapy
EP1390081A2 (en) 2001-01-08 2004-02-25 Neorx Corporation Therapeutic and diagnostic compounds, compositions, and methods
PT2314293T (en) 2001-01-16 2017-04-11 Vascular Therapies Llc Implantable device containing resorbable matrix material and rapamycin for preventing or treating vasuloproliferative diseases
US7115565B2 (en) 2001-01-18 2006-10-03 Pharmacia & Upjohn Company Chemotherapeutic microemulsion compositions of paclitaxel with improved oral bioavailability
US6548531B2 (en) 2001-02-09 2003-04-15 Hoffmann-La Roche Inc. Method for cancer therapy
US7666414B2 (en) * 2001-06-01 2010-02-23 Cornell Research Foundation, Inc. Methods for treating prostate cancer using modified antibodies to prostate-specific membrane antigen
US7514078B2 (en) * 2001-06-01 2009-04-07 Cornell Research Foundation, Inc. Methods of treating prostate cancer with anti-prostate specific membrane antigen antibodies
MXPA03010804A (en) * 2001-06-01 2004-11-22 Cornell Res Foundation Inc Modified antibodies to prostate-specific membrane antigen and uses thereof.
GB0121285D0 (en) 2001-09-03 2001-10-24 Cancer Res Ventures Ltd Anti-cancer combinations
US20030054042A1 (en) * 2001-09-14 2003-03-20 Elaine Liversidge Stabilization of chemical compounds using nanoparticulate formulations
EP1435965A4 (en) * 2001-09-26 2005-09-21 Intermune Inc Pharmaceutical compositions and methods for treating cancer
ES2625340T3 (en) * 2001-10-15 2017-07-19 Crititech, Inc. Compositions and methods for the administration of poorly water soluble drugs and treatment methods
IL163666A0 (en) 2002-02-22 2005-12-18 New River Pharmaceuticals Inc Active agent delivery systems and methods for protecting and administering active agents
WO2003080027A1 (en) * 2002-03-20 2003-10-02 Elan Pharma International, Ltd. Nanoparticulate compositions of angiogenesis inhibitors
US20080220075A1 (en) * 2002-03-20 2008-09-11 Elan Pharma International Ltd. Nanoparticulate compositions of angiogenesis inhibitors
GB2386836B (en) 2002-03-22 2006-07-26 Cancer Res Ventures Ltd Anti-cancer combinations
AU2003227610A1 (en) * 2002-04-12 2003-10-27 Multimmune Gmbh Non-toxic amounts of protein-aggregating substances stimulate hsp70 expression and function as anti-tumor agents
WO2003099323A1 (en) * 2002-05-10 2003-12-04 Agennix Incorporated Lactoferrin in the treatment of malignant neoplasms and other hyperproliferative diseases
CZ294371B6 (en) * 2002-06-10 2004-12-15 Pliva - Lachema, A. S. Stabilized pharmaceutical composition based on polyoxyethylated castor oil and process for preparing thereof
US20040047835A1 (en) * 2002-09-06 2004-03-11 Cell Therapeutics, Inc. Combinatorial drug therapy using polymer drug conjugates
GB2394658A (en) 2002-11-01 2004-05-05 Cancer Rec Tech Ltd Oral anti-cancer composition
EP1596825A2 (en) * 2003-02-03 2005-11-23 Neopharm, Inc. Stable sterile filterable liposomal encapsulated taxane and other antineoplastic drugs
EP1656124A1 (en) * 2003-03-12 2006-05-17 Cerenis Methods and compositions for the treatment of cancer
US7085886B2 (en) * 2003-05-28 2006-08-01 International Buisness Machines Corporation Autonomic power loss recovery for a multi-cluster storage sub-system
US20050208095A1 (en) * 2003-11-20 2005-09-22 Angiotech International Ag Polymer compositions and methods for their use
EP1610818A4 (en) * 2004-03-03 2007-09-19 Millennium Pharm Inc Modified antibodies to prostate-specific membrane antigen and uses thereof
US7989490B2 (en) * 2004-06-02 2011-08-02 Cordis Corporation Injectable formulations of taxanes for cad treatment
US8557861B2 (en) * 2004-09-28 2013-10-15 Mast Therapeutics, Inc. Low oil emulsion compositions for delivering taxoids and other insoluble drugs
GB0501192D0 (en) * 2005-01-20 2005-03-02 Resolution Chemicals Ltd Stable prostaglandin-containing compositions
EP1853250B1 (en) * 2005-02-18 2011-11-02 Abraxis BioScience, LLC Combinations and modes of administration of therapeutic agents and combination therapy
US8735394B2 (en) 2005-02-18 2014-05-27 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US20070003557A1 (en) * 2005-04-21 2007-01-04 Andres Forero Method for treating cancer using premedication
CA2601067A1 (en) 2005-05-04 2006-11-09 Medigene Ag Method of administering a cationic liposomal preparation comprising paclitaxel
TWI376239B (en) * 2006-02-01 2012-11-11 Andrew Xian Chen Vitamin e succinate stabilized pharmaceutical compositions, methods for the preparation and the use thereof
AU2007227466B2 (en) * 2006-03-16 2011-11-17 Bionumerik Pharmaceuticals, Inc. Anti-cancer activity augmentation compounds and formulations and methods of use thereof
CA2922029C (en) 2006-03-22 2017-11-28 Medigene Ag A combination of a cationic liposomal preparation comprising an antimitotic agent and a non-liposomal preparation comprising an antimitotic agent
CN101511372B (en) * 2006-07-07 2011-11-16 蒂尔坦制药有限公司 Anti-cancer therapy comprising an h2-blocker, at least one antiinflammatory agent and a cytotoxic agent
US8242165B2 (en) * 2006-10-26 2012-08-14 Creighton University Mucoadhesive nanoparticles for cancer treatment
CA2717181C (en) 2008-03-14 2013-10-15 Bionumerik Pharmaceuticals, Inc. Treatment methods and compositions for lung cancer, adenocarcinoma, and other medical conditions
CA2718233A1 (en) 2008-03-14 2009-09-17 Bionumerik Pharmaceuticals, Inc. Compositions and methods of use of compounds to increase cancer patient survival time
JP2012520314A (en) 2009-03-11 2012-09-06 アムビト ビオスシエンセス コルポラチオン Combination of indazolylaminopyrrolotriazine and taxane for cancer treatment
JP6242213B2 (en) 2010-03-29 2017-12-06 アブラクシス バイオサイエンス, エルエルシー Methods for improving the effectiveness of drug delivery and therapeutic agents
KR20130028728A (en) 2010-03-29 2013-03-19 아브락시스 바이오사이언스, 엘엘씨 Methods of treating cancer
KR20180049180A (en) 2010-06-04 2018-05-10 아브락시스 바이오사이언스, 엘엘씨 Methods of treatment of pancreatic cancer
KR20220025946A (en) 2014-03-21 2022-03-03 애브비 인코포레이티드 Anti-egfr antibodies and antibody drug conjugates
WO2017214335A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
CN109641962A (en) 2016-06-08 2019-04-16 艾伯维公司 Anti- B7-H3 antibody and antibody drug conjugates
JP2019522643A (en) 2016-06-08 2019-08-15 アッヴィ・インコーポレイテッド Anti-CD98 antibodies and antibody drug conjugates
BR112018075630A2 (en) 2016-06-08 2019-03-19 Abbvie Inc. anti-cd98 antibodies and antibody drug conjugates
JP2019526529A (en) 2016-06-08 2019-09-19 アッヴィ・インコーポレイテッド Anti-B7-H3 antibody and antibody drug conjugate
CN109562073B (en) 2016-09-29 2021-11-09 四川科伦药物研究院有限公司 Albumin pharmaceutical composition and preparation method thereof
UY39610A (en) 2021-01-20 2022-08-31 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2601675B1 (en) * 1986-07-17 1988-09-23 Rhone Poulenc Sante TAXOL DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FR2629818B1 (en) * 1988-04-06 1990-11-16 Centre Nat Rech Scient PROCESS FOR THE PREPARATION OF TAXOL
AU2005692A (en) * 1991-05-08 1992-12-21 United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Method for designing cancer treatment regimens and methods and pharmaceutical compositions for the treatment of cancer
FR2678833B1 (en) * 1991-07-08 1995-04-07 Rhone Poulenc Rorer Sa NEW PHARMACEUTICAL COMPOSITIONS BASED ON DERIVATIVES OF THE TAXANE CLASS.

Also Published As

Publication number Publication date
GR1001617B (en) 1994-07-29
EP0584001B1 (en) 1997-05-14
ZA93686B (en) 1993-09-06
IE930433A1 (en) 1994-02-09
ITMI931714A1 (en) 1995-01-29
EP0783886A2 (en) 1997-07-16
DE69310634D1 (en) 1997-06-19
DK65293A (en) 1994-02-04
AU3216893A (en) 1994-02-10
US5670537A (en) 1997-09-23
IL105879A (en) 1998-08-16
IE80620B1 (en) 1998-10-21
ES2105926A1 (en) 1997-10-16
DE4325927A1 (en) 1994-03-10
US6414014B1 (en) 2002-07-02
DE69310634T2 (en) 1998-01-02
ES2105926B1 (en) 1998-07-01
PT101289A (en) 1994-04-29
DE4325927B4 (en) 2004-04-15
SE9301951L (en) 1994-02-07
IT1276041B1 (en) 1997-10-24
SE513082C2 (en) 2000-07-03
CA2086874C (en) 1998-09-01
FR2694192A1 (en) 1994-02-04
BE1006497A5 (en) 1994-09-13
GB9311740D0 (en) 1993-07-21
AU641894B3 (en) 1993-09-30
AU704393C (en) 2002-07-25
FR2694192B1 (en) 1995-01-27
CA2086874A1 (en) 1994-02-04
DK65293D0 (en) 1993-06-04
EP0783886A3 (en) 1997-07-23
NL9301086A (en) 1994-03-01
US5665761A (en) 1997-09-09
ITMI931714A0 (en) 1993-07-29
EP0783885A2 (en) 1997-07-16
ES2101261T3 (en) 1997-07-01
SE9301951D0 (en) 1993-06-08
DK0584001T3 (en) 1997-08-25
US5641803A (en) 1997-06-24
EP0783885A3 (en) 1997-08-06
MY109862A (en) 1997-09-30
JP2848760B2 (en) 1999-01-20
GR3024415T3 (en) 1997-11-28
JPH07233064A (en) 1995-09-05
AU4207096A (en) 1996-04-26
ATE152910T1 (en) 1997-05-15
EP0584001A1 (en) 1994-02-23
NZ245795A (en) 1999-05-28
IL105879A0 (en) 1993-10-20
CH686868A5 (en) 1996-07-31
AU704393B2 (en) 1999-04-22
US5621001A (en) 1997-04-15

Similar Documents

Publication Publication Date Title
CA2086874E (en) Methods for administration of taxol
DE69233169T2 (en) Composition containing cisplatin and topotecan as an anti-tumor.
Tamura et al. Phase I study of paclitaxel by three‐hour infusion: hypotension just after infusion is one of the major dose‐limiting toxicities
MXPA01010138A (en) Docetaxel in combination with rhumab her2 for the treatment of cancers.
KR20160078987A (en) Cancer treatment with combination of plinabulin and taxane
EP2254570B1 (en) Combination comprising paclitaxel for treating ovarian cancer
EP1201247B1 (en) Treatment of metastatic renal cell carcinoma
WO2006124573A2 (en) Treatment of cancer with 2-deoxyglucose
GB2269319A (en) Compositions containing taxol
Dragnev et al. A dose escalation trial of biweekly docetaxel and gemcitabine with filgrastim or pegfilgrastim for the treatment of patients with advanced solid tumors
AU7179200A (en) Method of treatment of breast cancer
Phillips et al. Australasian multicentre phase II study of paclitaxel (Taxol*) in relapsed ovarian cancer
CN103889222B (en) Pharmaceutical composition containing paclitaxel milk surum acid esters
Riedel et al. Extravasation of paclitaxel (Taxol®)–clinical course
Boehnke The Evolving Role of Paclitaxel in Breast Cancer and the Ambulatory Care Setting
WO2009104152A1 (en) Combination treatment for ovarian cancer
WO2009104150A1 (en) Combination comprising bosentan for treating ovarian cancer

Legal Events

Date Code Title Description
EEER Examination request
NARE Reissued
MKLA Lapsed