Method of Administering Cationic Liposomes Comprising an Active Drug

Information

  • Patent Application
  • 20080063699
  • Publication Number
    20080063699
  • Date Filed
    October 15, 2004
    20 years ago
  • Date Published
    March 13, 2008
    16 years ago
Abstract
The present invention relates to the use of pharmaceutical preparations comprising paclitaxel for administration to a human patient in need thereof.
Description

FIGURE LEGENDS


FIG. 1: Tumor Volume after End of Treatment


Tumor size of L3.6pI pancreatic tumors 19 days after start of treatment. Treatment with 10% trehalose, paclitaxel, MBT-0206, Gemzar (gemcitabine) and the combination of both MBT-0206 and Gemzar started 8 days after tumor cell inoculation. Gemzar was applied i.p. at a dose of 100 mg/kg bw twice a week (Mon, Thu). paclitaxel and MBT-0206 were applied i.v. on a Mon, Wed, Fri schedule at a paclitaxel dose of 5 mg/kg bw. The combination group received both MBT-0206 and Gemzar with the respective schedule. Tumors were measured by palpation with a calliper on day 23 and 27. Mean±SEM; n=9 per group.



FIG. 2: Metastases after Therapy


Metastases at day 19 after start of treatment. Treatment with 10% trehalose, paclitaxel, MBT-0206, Gemzar (gemcitabine) and the combination of both MBT-0206 and Gemzar started at day 8 after tumor cell inoculation. Geinzar was applied i.p. at a dose of 100 mg/kg bw twice a week (Mon, Thu). paclitaxel and MBT-0206 were applied i.v. on a Mon, Wed, Fri schedule at a paclitaxel dose of 5 mg/kg bw. The combination group received both MBT-0206 and Gemzar with the respective schedule, n=9 per group.



FIG. 3-5: The growth inhibitory assay was performed in 24-well plates with each drug concentration tested in duplicate (n=2 wells). 4×104 cells per well were seeded into a 24-well plate and incubated over night. The following day, 10-11 concentrations of the respective drug formulation were added for 72 h to cover the range depicted in the respective graphs. Finally, the cell viability was determined by a standard MTT-assay measuring the activity of mitochondrial-dehydrogenases.



FIG. 3: Inhibitory Potential of MBT-0206 and paclitaxel against the highly drug-resistant uterus sarcoma line Mes-SADx-5MBT.



FIG. 4: Inhibitory Potential of MBT-0206 and paclitaxel against the moderatly drug-resistant uterus sarcoma line Mes-SA/Dx-5.



FIG. 5: Inhibitory Potential of MBT-0206 and paclitaxel against the drug-sensitive human uterus sarcoma line Mes-SA.



FIG. 6-7: The growth inhibitory assay was performed in 24-well plates with each drug concentration tested in duplicate (n=2 wells). 4×104 cells per well were seeded into a 24-well plate and incubated over night. The following day, 10-11 concentrations of the respective drug formulation were added for 72 h to cover the range depicted in the respective graphs. Finally, the cell viability was determined by a standard MTT-assay measuring the activity of mitochondrial dehydrogenases.



FIG. 6: Inhibitory Potential of MBT-0206 and paclitaxel against the highly drug-resistant murine colon carcinoma line Colon-26MBT.



FIG. 7: Inhibitory Potential of MBT-0206 and paclitaxel against the parental drug-sensitive murine colon carcinoma line Colon-26.



FIG. 8: Inhibitory Potential of MBT-0206 and paclitaxel against the drug-sensitive human endothelial line EA.hy926.


The growth inhibitory assay was performed in 24-well plates with each drug concentration tested in duplicate (n=2 wells). 4×104 cells per well were seeded into a 24-well plate and incubated over night. The following day, 11 concentrations of the respective drug formulation were added for 72 h to cover the range depicted in the respective graph. Finally, the cell viability was determined by a standard MTT-assay measuring the activity of mitochondrial dehydrogenases.



FIG. 9: In vitro inhibitory potential of paclitaxel against the sensitive parental line Mes-SA and the resistant derivative Mes-SA/Dx-5MBT. The growth inhibitory assay was performed in 24-well plates with each drug concentration tested in duplicate (n=2 wells). 4×104 cells per well were seeded into a 24-well plate and incubated over night. The following day, 10-11 concentrations of the respective drug formulation were added for 72 h to cover the range depicted in the respective graphs. Finally, the cell viability was determined by a standard MTT-assay measuring the activity of mitochondrial dehydrogenases.



FIG. 10: Inhibitory Potential of paclitaxel against the drug-resistant human dermal melanoma line Sk-Mel28. The growth inhibitory assay was performed in 24-well plates with each drug concentration tested in duplicate (n=2 wells). 4×104 cells per well were seeded into a 24-well plate and incubated over night. The following day, 11 concentrations of the respective drug formulation were added for 72 h to cover the range depicted in the respective graphs. Finally, the cell viability was determined by a standard MTT-assay measuring the activity of mitochondrial dehydrogenases.





The following examples should be illustrative only but are not meant to be limiting to the scope of the invention. Other generic and specific configurations will be apparent to those skilled in the art.


EXAMPLES
1. Human Therapy Treatment Protocol

This example is concerned with human treatment protocols using the formulations disclosed. Treatment will be of use preventing and/or treating various human diseases and disorders associated with enhanced angiogenic activity. It is considered to be particularly useful in anti-tumor therapy, for example, in treating patients with solid tumors and hematological malignancies or in therapy against a variety of chronic inflammatory diseases such as rheumatoid arthritis or psoriasis.


A feature of the invention is that several classes of diseases and/or abnormalities may be treated by directly targeting angiogenic epithelial cells without directly targeting the tissue or cells involved in the abnormality e.g., by inhibiting angiogenesis the blood supply to a tumor is cut off and the tumor is killed without directly targeting the tumor cells in any manner. Other classes of diseases and/or abnormalities may be treated by directly targeting angiogenic endothelial cells and by directly targeting the tissue or cells involved in the abnormality.


In an other application, drug resistant cells such as drug resistant cancer cells or highly proliferative synoviocytes in rheumatoid arthritis can be affected directly.


The various elements of conducting a clinical trial, including patient treatment and monitoring, will be known to those skilled in the art in light of the present disclosure.


For regulatory approval purposes, it is contemplated that patients chosen for a study would have failed to respond to at least one course of conventional therapy and would have objectively measurable disease as determined by physical examination, laboratory techniques, or radiographic procedures. Such patients would also have no history of cardiac or renal disease and any chemotherapy should be stopped at least 2 weeks before entry into the study.


Prior to application, the formulation can be reconstituted in an aqueous solution in the event that the formulation was freeze dried. As outlined above, the required application volume is calculated from the patient's body weight and the dose schedule.


The disclosed formulations may be administered over a short infusion time. The infusion given at any dose level should be dependent upon the toxicity achieved after each. Thus, if Grade II toxicity was reached after any single infusion, or at a particular period of time for a steady rate infusion, further doses should be withheld or the steady rate infusion stopped unless toxicity improved. Increasing doses should be administered to groups of patients until approximately 60% of patients showed unacceptable Grade III or IV toxicity in any category. Doses that are ⅔ of this value would be defined as the safe dose.


Physical examination, tumor measurements and laboratory tests should, of course, be performed before treatment and at intervals of about 3-4 weeks later. Laboratory tests should include complete blood cell counts, serum creatinine, creatine kinase, electrolytes, urea, nitrogen, SGOT, bilirubin, albumin and total serum protein.


Clinical responses may be defined by acceptable measure or changes in laboratory values e.g. tumor markers. For example, a complete response may be defined by the disappearance of all measurable disease for at least a month, whereas a partial response may be defined by a 50% or greater reduction.


All of the compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those skilled in the art that variations may be applied to the composition, methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.


Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by the FDA Office of Biologics standards.


The present invention includes a method of delivery of a pharmaceutically effective amount of the inventive formulation of an active agent to a target site such as an angiogenic vascular target site of a subject in need thereof. A “subject in need thereof” refers to a mammal, e.g. a human.


The route of administration preferably comprises peritoneal or parenteral administration.


For use with the present invention the “pharmacologically effective amount” of a compound administered to a subject in need thereof will vary depending on a wide range of factors. The amount of the compound will depend upon the size, age, sex, weight, and condition of the patient, as well as the potency of the substance being administered. Having indicated that there is considerable variability in terms of dosing, it is believed that those skilled in the art can, using the present disclosure, readily determine appropriate dosing by first administering extremely small amounts and incrementally increasing the dose until the desired results are obtained. Although the amount of the dose will vary greatly based on factors as described above, in general, the present invention makes it possible to administer substantially smaller amounts of any substance as compared with delivery systems which only target the pathologic tissue e.g., target the tumor cells themselves.


2. Mono-Therapy Protocols
















Study No.
Indication









CTLP01
Prostate Cancer



CTLP05
Gastro-Intestinal Cancer



CTLP07
Melanoma



CTLP09
Breast Cancer



CTLP10
Colorectal Cancer



CTLP15
Breast Cancer



CTLP16
Pancreatic Cancer










Dosing













Study No.
Dosages







CTLP01
0.0688 mg/kg, 0.275 mg/kg, 1.10 mg/kg, 1.65 mg/kg (mg



liposomal paclitaxel/kg)


CTLP05
0.275 mg/kg, 0.55 mg/kg, 1.10 mg/kg, 1.65 mg/kg (mg



liposomal paclitaxel/kg)


CTLP07
0.275 mg/kg, 0.55 mg/kg, 1.10 mg/kg (mg liposomal



paclitaxel/kg)


CTLP09
0.55 mg/kg, 1.10 mg/kg (mg liposomal paclitaxel/kg)


CTLP10
0.55 mg/kg (mg liposomal paclitaxel/kg)


CTLP15
25 mg/kg (mg liposomal paclitaxel/kg)


CTLP16
0.55-1.1 mg/kg (mg liposomal paclitaxel/kg)









Standard formulation liposomal paclitaxel 50 mol % DOTAP:47 mol % DOPC:3 mol % paclitaxel (MBT-0206=EndoTag 1)


Treatment Schedule Ongoing Completed Studies
















No.


Study No.
Schedule
of applications







CTLP01
3 times a week
N = 3


CTLP05
3 times a week with 3 weeks interval each
N = 6


CTLP09
3 times a week with 2 weeks interval each
N = 9


CTLP10
3 times a week with 2 weeks interval each
N = 9









Treatment Schedule Ongoing Studies
















No.


Study No.
Schedule
of applications







CTLP03
3 times a week
N = 3


CTLP04
weekly
N = 14


CTLP06
4-5 times a week with 1 week interval each
N = 14


CTLP07
3 times a week with 2-3 weeks interval each
N = 6-9


CTLP15
3 times a week with 2 weeks interval each
N = 24


CTLP16
3 times a week with 3 weeks interval each
N = 9









Efficacy

Response will be evaluated according to the WHO or RECIST criteria.


3. Combination Therapy Protocols
















Study No.
Indication









CTLP04
Lung Cancer



CTLP06
Colorectal or Gastric Cancer










Dosing
















Study No.
Dosages









CTLP04
liposomal paclitaxel, 1.5 mg/kg and Carboplatin




(2 mg/ml/min i.v. For 15 min once weekly)



CTLP06
liposomal paclitaxel, 0.5 or 1.0 mg/kg alone, 0.5




or 1.0 mg/kg liposomal paclitaxel and 5-fluorouracil




(2000 mg/m2).










Standard Formulation Liposomal Paclitaxel

50 mol % DOTAP:47 mol % DOPC:3 mol % paclitaxel Treatment schedule for liposomal paclitaxel in ongoing studies

















Study No.
Schedule
No. of applications









CTLP04
once weekly
N = 14



CTLP06
daily with one week
N = 14




terval each










A further planned study comprises administration of liposomal paclitaxel, e.g. 0.5 or 1.0 or 1.5 mg/kg and gemcitabine, e.g. 1000 mg/m2 once weekly for three weeks followed by one week without treatment, preferably for an interval of at least one year.


The treatment schedule for liposomal paclitaxel will be as described above for ongoing studies.


Efficacy

Response will be evaluated according to the WHO or RECIST criteria.


4. Case Report #1

Patient:

    • 49 years old patient with large therapy resistant recidivism of a mucoepidermoidal carcinoma of the larynx
    • metastases cervical, supraclavicular, axillar, mediastinal and pulmonal
    • 5 years after first tumor resection, neck tumor dissection and adjuvant radiotherapy
    • after repeated therapy of recidivism with multiple resections, plastic surgery, radiotherapy and chemotherapy


Dosing Schedule:

    • MBT-0206: 50/47/3 (DOTAP/DOPC/paclitaxel)
    • Application of 0.06, 0.25, 0.5 and 1.0 mg liposomal paclitaxel/kg bw, i.v.
    • One cycle of 3 times a week (on day 1, 3 and 5)


Results:

    • Good tolerance while monitoring cardiovascular, pulmonary and serological parameter during and after infusion
    • No signs of acute or chronic toxicity
    • Reduction of tumor blood circulation
    • Strongly reduced progression of tumor growth during 3 months


5. Case Report #2

One patient with liver cell carcinoma, who had disease progression after multiple chemotherapies, has been treated with MBT-0206.


Lyophilized MBT-0206 has been reconstituted with water for injection and a total infusion volume of 300400 ml MBT-0206 (equivalent to a dose of 1.0 mg liposomal paclitaxel/kg body weight) has been administered by central or peripheral intravenous infusion over a period of 2- 4 h. The infusion rate has been increased slowly up to a maximum speed of 2,5 ml/min. Premedication depended on the patient's sex, age, condition. In the specific case of the above mentioned patient it has been given dexamethasone and an antihistamine.


MBT-0206 has been administered once weekly with a dose escalation schedule, beginning with 2 times 0.25 mg liposomal paclitaxel/kg bw, 1 times 0.5 mg liposomal paclitaxel/kg bw) and than a consolidation dose of 19 times 1.0 mg liposomal paclitaxel/kg bw. This treatment is after 22 weekly administrations still ongoing and up to now no adverse drug reactions have been reported. Besides the favourable safety profile the last evaluation of tumor size, which has been performed by CT-Scan of the liver, showed stable disease.


6. Case Report #3

In another case a prostate cancer patient who became refractory to hormone therapy, has been treated with 1.0 mg liposomal paclitaxel/kg bw, 3 times weekly every third day under the same conditions of preparation and administration as described above. The premedication contained dexamethasone and antihistamines. The accumulated dose of liposomal paclitaxel for this patient in 7 days was 3.0 mg liposomal paclitaxel/kg bw.


7. Low Dosing Schedule With Liposomal Paclitaxel

Immortalised endothelial cells (EA.hy926) are seeded into 24-well plates (4×104 cells per well) and grown over night. The following day, 9 wells are treated for 1 h with the low dose of 51.2 ng/ml liposomal paclitaxel (60 nM) formulated as MBT-0206. In addition, 3 wells per formulation are treated with the high dose of 153.7 ng/ml (180 nM) liposomal paclitaxel formulated as MBT-0206 for 1 h and 3 wells remain untreated. Approximately 24 h later, 6 of the 9 low dose-treated wells are again treated with the same low doses of MBT-0206 for 1 h (i.e. 2× treatment groups). Again 24 h later, 3 of these 6 two times-treated wells are treated for the third time with 51.2 ng/ml paclitaxel formulated as MBT-0206 for 1 h (3× treatment groups). Approximately 96 h after this third treatment, the cell viability of all wells is quantitated. For this purpose, an assay which measures the activity of mitochopdrial dehydrogenases using the tetrazolium salt 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) is applied according to standard protocols (e.g. {Lindl, 1994 #28} with slight modifications).


The results demonstrate that the viability of cells treated for 3 times with a low dose of MBT-0206 is at least as strongly reduced as the viability of cells treated only one time with a high dose. Cells treated one time or two times with the low dose of MBT-0206 exhibit a somewhat increased viability which is, however, reduced in comparison to untreated cells.


Conclusion

Treatments with high doses of MBT-0206 can be replaced by using low doses at a higher frequency. There is a correlation between treatment density (no. of treatments per week) and treatment efficacy. Three weekly treatments with low doses were superior to 1 or 2 weekly treatments. This optimised dosing regimen potentially reduces toxic side effects caused by high dose treatments.


8. Anti-Tumor Efficacy of MBT-0206 in Combination with Gemcitabine (50 mg/kg) in L3.6pI Pancreatic Tumors
Animal Model



  • Species: male Balb/c nu/nu mice

  • Tumor model: Solid orthotopic L3.6pI pancreas tumor (highly mestastatic; Bruns C J, Harbison M T, Kuniyasu H, Eue I, Fidler I J. In vivo selection and characterization of metastatic variants from human pancreatic adenocarcinoma by using orthotopic implantation in nude mice. Neoplasia 1999; 1(1):50-62.)

  • Supplier: Charles River France



Treatment















Treatment start:
Day 7 after tumor inoculation


Last treatment:
Day 26 after tumor inoculation


Dose:
MBT-0206 with 5 mg paclitaxel/kg bw per application



paclitaxel at 5 mg paclitaxel/kg bw per application



Gemcitabine (Gemzar) at 50 mg/kg per application


Schedule:
MBT-0206, paclitaxel, trehalose: d 7, 10, 12, 14,



17, 19, 21, 24, 26



Gemcitabine: d 7, 11, 14, 18, 21, 25



Combination: combined mono treatments


Application
MBT-0206, paclitaxel, trehalose: i.v. bolus into tail vein


route:
Gemcitabine: i.p. bolus











Groups (n = 9; n = 2-5 at day 26, 28)
Treatment





Trehalose, paclitaxel, MBT-0206
d 7, 10, 12, 14, 17, 19, 21, 24, 26


Gemcitabine [50 mg/kg]
d 7, 11, 14, 18, 21, 25


MBT-0206 [5 mg/kg bW] +
d 7, 10, 12, 14, 17, 19, 21, 24, 26


Gemcitabine [50 mg/kg]
d 7, 11, 14, 18, 21, 25









The mortality was low before day 21: only 1 control and 1 MBT-0206 animal died. At day 24 the mortality increased for unknown reason: 3 MBT-0206, 1 Gemzar and 4 control animals were dead at day 24.


The body weight was not effected by either treatment, only the weight of control tumors decreased by 18% during the last week.


Monitored Parameters

Tumor volume palpated at day 10, 12, 14, 17, 19, 21, 24 after inoculation and at day 26, 28 after harvesting


Necrospy after harvest at day 26 and 28


Results

No effect of any treatment could be observed by palpation three days after begin of treatment. Strong anti-tumor effect was observed after one week and at day 24 by palpation, with the following ranking in efficacy: Gemzar-50≈paclitaxel <MBT-0206<MBT-0206+Gemzar. However, after harvest at day 26, the measured tumor volumes of all groups were clearly lower compared to day 24. This difference in size is most likely due to imprecise palpation before harvest. At day 24 tumor size is reduced to ˜30% by the mono treatments compared to the control group (n=2). The combination of MBT-0206+Gemzar resulted in the strongest reduction of the tumor size to 13%, which was significantly (p<0.05) more effective compared to paclitaxel, Gemzar and MBT-0206 alone. At day 28 the control group (n=2) is not shown because one of the two tumors was extremely small compared all other tumors (day 24, 26, 28) and thus considered as not representative. The tumors after the mono treatments showed a weak increase in tumor size compared to day 26 (paclitaxel: 536 mm3; MBT-0206: 392 mm3; Gemzar: 398 mm3), whereas the combination therapy led to a slight tumor regression between day 26 and 28 to 88 mm3.


These data show a strong anti-tumor efficacy of paclitaxel, Gemzar and MBT-0206 in this model. The anti-tumor action of MBT-0206 is slightly stronger than paclitaxel but similar to Gemzar. The combination of MBT-0206 and Gemzar shows an impressive anti-tumor efficacy.


9. Anti-Tumor Efficacy of MBT-0206 in Combination with Gemcitabine (100 mg/kg) in L3.6pI Pancreatic Tumors














Animal Model








Species:
male Bald/c nu/nu mice


Tumor model:
Solid orthotopic L3.6pl pancreas



tumor (highly mestastatic)


Supplier:
Charles River France







Treatment








Treatment start:
Day 8 after tumor inoculation (01.05.03)


Last treatment:
Day 26 after tumor inoculation (19.05.03)


Schedule:
MBT-0206, paclitaxel, trehalose: d 9, 12, 14, 16,



19, 21, 23, 26



Gemcitabine: d 8, 12, 15, 19, 22, 26



Combination: combined mono treatments











Groups (n = 9)
Treatment





Trehalose, paclitaxel, MBT-0206
d 9, 12, 14, 16, 19, 21, 23, 26


Gemcitabine [100 mg/kg]
d 8, 12, 15, 19, 22, 26


MBT-0206 [5 mg/kg bw] + Gemcitabine
d 9, 12, 14, 16, 19, 21, 23, 26


[100 mg/kg]
d 8, 12, 15, 19, 22, 26









Monitored Parameters

Tumor volume palpated at day 23, 26 after inoculation and after harvesting


Body weight from day 1, 7,12, 16, 19, 21, 23, 27


Necrospy after harvest at day 27


Results (See FIGS. 1 and 2)

Clear anti-tumor effect of all therapeutic treatments was observed at day 23 after tumor inoculation, with an prominent efficacy of the combination therapy (FIG. 1). Ranking of tumor inhibition: paclitaxel<MBT-0206=Gemzar-50<MBT-0206+Gemzar. Compared to the control group, the final tumor volumes were significantly reduced by MBT-0206 to 46% (p<0.05), by Gemzar to 47% (p<0.01) and by the combination therapy to 22% (p<0.01) at day 27. paclitaxel treatment reduced the final tumor volume to 68%, which was not significant. Interestingly, the efficacy of MBT-0206 and the combination therapy were more pronounced at day 23. Responsible for that might be the extended therapeutic interval during the weekend between day 23 and 27. These data reveal a clear anti-tumor efficacy of paclitaxel, Gemzar and MBT-0206 in this model. The anti-tumor action of MBT-0206 is slightly stronger than paclitaxel but similar to Gemzar. Both MBT-0206 and the combination with Gemzar inhibited the formation of metastases (FIG. 2). Liver metastases were absent only in the these two groups. Additionally, only in the combination group the lymph node metastases were rare. The data revealed that the combination of MBT-0206 and Gemzar enhances the anti-tumor efficacy of either mono-therapy.


The mortality was slightly increased in the treatment groups, particularly during combinatory treatment (4 mice died). No control animal died before harvest.


The body weight of control mice decreased by 18% during the last 11 days. During this period a transient decrease was also observed for the treated mice leading to a weight loss of 2% for paclitaxel, 12% for MBT-0206, 19% for the combination and 22% for Gemzar.


10. Killing of Paclitaxel Resistant Cells (e.g. Tumor Cell Lines)


To demonstrate the potential of MBT-0206 to directly kill tumors expressing (multi) drug resistance, two highly paclitaxel resistant mammalian tumor cell lines were investigated in vitro. These cell lines were selected by stepwise increasing the concentration of paclitaxel in the culture medium. Both cell lines have developed a high resistance level which is reflected by concentrations for 50% growth inhibition (IC50 value) for paclitaxel around 1 or 5 μM (867 or 5000 ng/ml). In both instances, MBT-0206 is clearly superior to paclitaxel in killing drug resistant tumor cells. In contrast, in drug-sensitve or low-resistant cell lines, MBT-0206 has a more or less identical killing potential to paclitaxel.


MBT-0206 and the Human Uterus Sarcoma Derived Cell Line Mes-SA and its Derivative Lines

The highly paclitaxel resistant derivative cell line Mes-SA/Dx-5MBT was selected with increasing paclitaxel concentrations from the commercially available line Mes-SA/Dx-5 (ATCC, {Harker, 1986 #29}). As shown in FIG. 3, it is highly resistant to paclitaxel indicated by the IC50 value of 867 ng/ml. Surprisingly, it is found that MBT-0206 is killing this cell line much more effectively, mirrored by the approximately 20-fold lower IC50 value. The commercially available line Mes-SA/Dx-5 which was selected from the parental line Mes-SA with doxorubicin ({Harker, 1986 #29}) expresses a low level of cross resistance for paclitaxel (compare FIGS. 3-5). The IC50 value for paclitaxel is approximately 7-fold lower than in Mes-SA/Dx-5MBT. Concomitantly, there is only a slight tendency of higher killing potential of MBT-0206 compared to paclitaxel in this cell line (FIG. 4). The parental line Mes-SA is highly sensitive for paclitaxel indicated by the low IC50 value of 5.5 ng/ml (FIG. 5). Against this drug-sensitive line, MBT-0206 has the same killing potential as paclitaxel. This is also true for all other paclitaxel-sensitive lines investigated so far. As example for this notion the results of treatments with MBT-0206 and paclitaxel of the immortalised endothelial line EA.hy926 are shown in FIG. 8.


MBT-0206 and the Murine Colon Carcinoma Derived Cell Line Colon-26

In a similar way to Mes-SA/Dx-5MBT, a highly paclitaxel resistant derivative line of the murine colon carcinoma line Colon-26 (Cell lines Service, Heidelberg) was established and called Colon-26MBT. The IC50 value for paclitaxel is approximately 5 μg/ml (FIG. 6). Again as in Mes-SA/Dx-5MBT, MBT-0206 had a clearly higher potential to inhibit the growth of this cell line. In this cell line, the IC50 values differ by a factor of 3. In line with the result shown for Mes-SA and EA.hy926 cells, the parental drug-sensitive line Colon-26 is equally sensitive for MBT-0206 and paclitaxel (FIG. 7).


Conclusion

In highly paclitaxel-resistant cell lines, MBT-0206 has a significantly higher killing potential as paclitaxel. In paclitaxel-sensitive lines, both paclitaxel formulations have a comparable efficacy. MBT-0206 may therefore be able to kill also (multi) drug resistant tumors directly in vitro and in vivo. It may, therefore, be a new approach to treat human tumors (or other diseases) which become unresponsive for paclitaxel.


11. Therapy of recurrent head and neck squamous-cell carcinoma

A safety evaluation of paclitaxel loaded cationic liposomes (MBT-0206) in patients with recurrent, therapy refractory head and neck squamous-cell carcinoma was carried out.


Study Design

    • Patients with recurrent, therapy refractory head and neck squamous-cell carcinoma
    • Measurable disease, defined as at least one lesion that can be accurately measured in at least on dimension with spiral CT/MRI scan
    • Karnofsky-performance index >60%
    • Life expectancy >4 months


Drug Administration

    • paclitaxel encapsulated in cationic liposomes was administered in 2 doses: 0.55 mg paclitaxel/kg and 1.10 mg paclitaxel/kg
    • After a screening period, the drug was intravenously injected on day 1, 3 and 5


Conclusions

During and after injection no signs of acute or chronic toxicity were observed: Vital and lab safety parameters remained nearly constant.


The results indicate that the dose and schedule suggested from preclinical toxicology studies was well tolerated by the patients.


Cationic liposomes selectively targeted endothelium of human head and neck squamous cell carcinoma. Laser Doppler flowmetry confirmed the antivascular mechanism of action of the therapy.


12. MBT-0206 (EndoTag 1) is Effective Against Paclitaxel-Resistant Tumors In Vivo

Two tumor cell lines which show high survival rates upon 72 h treatments with paclitaxel in vitro were used for mouse tumor models to investigate the potential of MBT-0206 in vivo. In these tumor models, it can be demonstrated that MBT-0206 is inhibiting tumor growth significantly more effective than paclitaxel.


1. A highly paclitaxel resistant derivative (Mes-SA/Dx-5MBT, IC50 0.87 μg/ml) was selected from the commercially available human uterus sarcoma derived cell Mes-SA/Dx-5 (ECACC). This moderately resistant line originates from the highly sensitive parental line Mes-SA. As shown in FIG. 9, the line Mes-SA/Dx-5MBT exhibits a ˜150-fold increased paclitaxel tolerance compared to the parental line Mes-SA as assessed by their respective IC50 values. For in vivo experiments, Mes-SA/Dx-5MBT cells were injected subcutaneously (s.c.) into NMRI nude mice. Treatments with MBT-0206 or paclitaxel were started at day 12 and extended until day 21 with 3 weekly doses (5 mg/kg b.w. paclitaxel). The mean tumor size of MBT-0206 treated animals was compared to the mean tumor size of paclitaxel treated animals 2 days after the last treatment (day 23 after start of treatments). The reduction induced by MBT-0206 compared to paclitaxel is given in % (see table below). As shown in the table below and FIG. 9, MBT-0206 was clearly more effective than paclitaxel in this tumor model in reducing the tumor growth by ˜⅓.


2. Concerning the human dermal melanoma line Sk-Mel28, already the commercially available line exhibits high paclitaxel resistance (IC50 5.8 μg/ml, FIG. 10) and no sensitive derivative line is available for comparison. In vivo, these cells were injected into SCID mice bearing human skin transplants between human skin transplant and mouse acceptor skin. Treatments were started on day 17 after tumor cell injection and extended to day 28 with doses (12.5 mg/kg b.w.) every second day. The therapeutic potential of MBT-0206 and paclitaxel were compared on day 7 after start of treatments (see table below). In this tumor model, MBT-0206 nearly completely blocked tumor development whereas paclitaxel was completely ineffective. Upon treatment with MBT-0206, tumors only marginally grew to about 1/10 the size of paclitaxel (paclitaxel)-treated tumors.











TABLE







No. of animals



Tumor reduction induced by
in group MBT-0206


Name of Tumor
MBT-0206 vs. paclitaxel (%)
or paclitaxel







Mes-SA/Dx-5MBT
35.5
4/4


Sk-Mel28
90.0
6/5










Mes-SA/Dx-5MBT tumor model: tumor volumes on day 23 after start of treatment (9 treatments, i.e. 3 per week, from day 12 to day 31 after tumor cell injection)


Sk-Mel tumor model: tumor volumes on day 7 after start of treatment (6 treatments very other day, i.e. from day 17 to day 28 after tumor cell injection).


REFERENCE LIST



  • 1. Rowinsky, E. K., and R. C. Donehower. 1995. paclitaxel (paclitaxel). N Engl J Med 332:1004-1014.

  • 2. Awada, A. 2002. New cytotoxic agents and molecular-targeted therapies in the treatment of metastatic breast cancer. Forum (Genova) 12:4-15.

  • 3. Seidman, A. D. 2003. Monotherapy options in the management of metastatic breast cancer. Semin Oncol 30:6-10.

  • 4. Romanini, A., L. Tanganefli, F. Carnino, A. Fanucchi, R. Lionetto, S. Pastorino, S. Cosio, A. Gadducci, and P. F. Conte. 2003. First-line chemotherapy with epidoxorubicin, paclitaxel, and carboplatin for the treatment of advanced epithelial ovarian cancer patients. Gynecol Oncol 89:354-359.

  • 5. Blom, R., N. Palm, and E. Simonsen. 1996. paclitaxel (paclitaxel) monotherapy in the treatment of progressive and recurrent ovarian carcinoma after platinum-based chemotherapy. Acta Oncol 35:733-736.

  • 6. Modi, S., K. S. Panageas, E. T. Duck, A. Bach, N. Weinstock, J. Dougherty, L. Cramer, C. Hudis, L. Norton, and A. Seidman. 2002. Prospective exploratory analysis of the association between tumor response, quality of life, and expenditures among patients receiving paclitaxel monotherapy for refractory metastatic breast cancer. J Clin Oncol 20:3665-3673.

  • 7. Ozols, R. F., B. N. Bundy, B. E. Greer, J. M. Fowler, D. Clarke-Pearson, R. A. Burger, R. S. Mannel, K. DeGeest, E. M. Hartenbach, and R. Baergen. 2003. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol 21:3194-3200.

  • 8. Vogelstein, B., E. R. Fearon, S. R. Hamilton, S.E. Kern, A. C. Preisinger, M. Leppert, Y. Nakamura, R. White, A. M. Smits, and J. L. Bos. 1988. Genetic alterations during colorectal-tumor development. N Engl J Med 319:525-532.

  • 9. Kerbel, R. S. 1991. Inhibition of tumor angiogenesis as a strategy to circumvent acquired resistance to anti-cancer therapeutic agents. Bioessays 13:31-36.

  • 10. Schünemann, Possinger, Scheidel, and Willich. 1999. Gynäkologische Malignome. Zuckschwerdt GmbH, Germering/München.

  • 11. Heidemann, E., B. Steinke, and H. D. Waller. 1997. Therapieschemata Onkologie und Hämatologie. Urban & Schwarzenberg, München.

  • 12. Heinemann, V. 2003. Role of gemcitabine in the treatment of advanced and metastatic breast cancer. Oncology 64:191-206.

  • 13. Thigpen, J. T., J. A. Blessing, G. Olt, S. S. Lentz, and J. Bell. 2003. Cisplatin as second-line therapy in ovarian carcinoma treated initially with single-agent paclitaxel: a Gynecologic Oncology Group study. Gynecol Oncol 90:581-586.

  • 14. Kuenen, B. C., L. Rosen, E. F. Smit, M. R. Parson, M. Levi, R. Ruijter, H. Huisman, M. A. Kedde, P. Noordhuis, W. J. van der Vijgh, G. J. Peters, G. F. Cropp, P. Scigalla, K. Hoekman, H. M. Pinedo, and G. Giaccone. 2002. Dose-finding and pharmacokinetic study of cisplatin, gemcitabine, and SU5416 in patients with solid tumors. J Clin Oncol 20:1657-1667.

  • 15. Sledge, G. W., Jr. 2003. Gemcitabine combined with paclitaxel or paclitaxel/trastuzumab in metastatic breast cancer. Semin Oncol 30:19-21.

  • 16. Reck, M., J. von Pawel, H. N. Macha, E. Kaukel, K. M. Deppermann, R. Bonnet, K. Ulm, S. Hessler, and U. Gatzemeier. 2003. Randomized phase III trial of paclitaxel, etoposide, and carboplatin versus carboplatin, etoposide, and vincristine in patients with small-cell lung cancer. J Natl Cancer Inst 95:1118-1127.

  • 17. Zimpfer-Rechner, C., U. Hofmann, R. Figl, J. C. Becker, U. Trefzer, I. Keller, A. Hauschild, and D. Schadendorf. 2003. Randomized phase II study of weekly paclitaxel versus paclitaxel and carboplafin as second-line therapy in disseminated melanoma: a multicentre trial of the Dermatologic Co-operative Oncology Group (DeCOG). Melanoma Res 13:531-536.

  • 18. Sledge, G. W., D. Neuberg, P. Bernardo, J. N. Ingle, S. Martino, E. K. Rowinsky, and W. C. Wood. 2003. Phase III trial of doxorubicin, paclitaxel, and the combination of doxorubicin and paclitaxel as front-line chemotherapy for metastatic breast cancer: an intergroup trial (E1193). J Clin Oncol 21:588-592.

  • 19. Nobmann, S., B. Bauer, and G. Fricker. 2001. Ivermectin excretion by isolated functionally intact brain endothelial capillaries. Br J Pharmacol 132:722-728.

  • 20. Thomas, H., and H. M. Coley. 2003. Overcoming multidrug resistance in cancer: an update on the clinical strategy of inhibiting p-glycoprotein. Cancer Control 10:159-165.

  • 21. Harker, W. G., and B. I. Sikic. 1985. Multidrug (pleiotropic) resistance in doxorubicin-selected variants of the human sarcoma cell line MES-SA. Cancer Res 45:4091-4096.

  • 22. Fellner, S., B. Bauer, D. S. Miller, M. Schaffrik, M. Fankhanel, T. Spruss, G. Bernhardt, C. Graeff, L. Farber, H. Gschaidmeier, A. Buschauer, and G. Fricker. 2002. Transport of paclitaxel (paclitaxel) across the blood-brain barrier in vitro and in vivo. J Clin Invest 110:1309-1318.

  • 23. Kiesewetter, D. O., E. M. Jagoda, C. H. Kao, Y. Ma, L. Ravasi, K. Shimoji, L. P. Szajek, and W. C. Eckelman. 2003. Fluoro-, bromo-, and iodo paclitaxel derivatives: synthesis and biological evaluation. Nucl Med Biol 30:11-24.

  • 24. Kohler, S., and W. D. Stein. 2003. Optimizing chemotherapy by measuring reversal of P-glycoprotein activity in plasma membrane vesicles. Biotechnol Bioeng 81:507-517.

  • 25. Leonard, G. D., O. Polgar, and S. E. Bates. 2002. ABC transporters and inhibitors: new targets, new agents. Curr Opin Investig Drugs 3:1652-1659.

  • 26. Agrawal, M., J. Abraham, F. M. Balis, M. Edgerly, W. D. Stein, S. Bates, T. Fojo, and C. C. Chen. 2003. Increased 99mTc-sestamibi accumulation in normal liver and drug-resistant tumors after the administration of the glycoprotein inhibitor, XR9576. Clin Cancer Res 9:650-656.

  • 27. Callies, S., D. P. de Alwis, A. Harris, P. Vasey, J. H. Beijnen, J. H. Schellens, M. Burgess, and L. Aarons. 2003. A population pharmacokinetic model for paclitaxel in the presence of a novel P-gp modulator, Zosuquidar Trihydrochloride (LY335979). Br J Clin Pharmacol 56:46-56.

  • 28. Lindl, T., and J. Bauer. 1994. Zell- und Gewebekultur. Gustav Fischer Verlag, Stuttgart.

  • 29. Harker, W. G., D. Bauer, B. B. Etiz, R. A. Newman, and B. I. Sikic. 1986. Verapamil-mediated sensitization of doxorubicin-selected pleiotropic resistance in human sarcoma cells: selectivity for drugs which produce DNA scission. Cancer Res 46:2369-2373.


Claims
  • 1. Use of a cationic liposomal preparation comprising at least one cationic lipid from about 30 mole % to about 99.9 mole %, paclitaxel in an amount of at least about 0.1 mole % and at least one neutral and/or anionic lipid from about 0 mole % to about 70 mole % for manufacturing a pharmaceutical composition for administering to a human patient in need thereof at a monthly dose of about 0.25 mg up to about 60 mg of paclitaxel/kg body weight of said patient.
  • 2. The use of claim 1, wherein said monthly dose is about 0.5 mg up to about 30 mg paclitaxel/kg body weight.
  • 3. The use of claim 2, wherein said monthly dose is about 1.0 mg up to about 15 mg paclitaxel/kg body weight.
  • 4. The use of claim 2, wherein said monthly dose is about 1 to about 7.5 mg/paclitaxel/kg body weight.
  • 5. The use of claim 1, wherein said monthly dose is about 20 to about 60 mg/paclitaxel/kg body weight.
  • 6. The use of claim 1, wherein administering said cationic liposomal preparation is at least once a time daily.
  • 7. The use of claim 1, wherein administering said cationic liposomal preparation is a plurality of times during a month period, each of said times being separated by an interval of between one day and 3 weeks.
  • 8. The use of claim 1, wherein administering said cationic liposomal preparation is (i) at least 3 times, especially 3-5 times in a first week, followed by an interval of 1-3weeks without administration, and optionally one or several repeats of this protocol,(ii) once in a first week followed by an interval of at least one week, especially 1-3weeks, without administration, and optionally one or several repeats of this protocol,(iii) once in a week for one week or several successive weeks, or(iv) a combination of (i), (ii) and/or (iii).
  • 9. Use of a cationic liposomal preparation comprising at least one cationic lipid from about 30 mole % to about 99.9 mole %, paclitaxel in an amount of at least about 0.1 mole % and at least one neutral and/or anionic lipid from about 0 mole % to about 70 mole % for manufacturing a pharmaceutical composition for simultaneous, separate, or sequential combination therapy with a jointly effective dose of at least one further active agent and/or heat and/or radiation and/or cryotherapy.
  • 10. The use of claim 9, wherein the composition is for simultaneous combination therapy with a jointly effective dose of at least one further active agent.
  • 11. The use of claim 1, wherein said cationic liposomal preparation comprises paclitaxel in an amount of at least about 2 mole % to about 8 mole %.
  • 12. The use of claim 1, wherein said cationic liposomal preparation comprises paclitaxel in an amount of about 2.5 mole % to about 3.5 mole %.
  • 13. The use of claim 1, wherein said cationic liposomal preparation comprises 50:47:3 mole % of DOTAP, DOPC and paclitaxel.
  • 14. The use of claim 1, wherein said cationic liposomal preparation comprises substantially no paclitaxel crystals.
  • 15. The use of claim 1 for treating an angiogenesis-associated condition.
  • 16. The use of claim 15 for treating wound healing, cancer, an inflammatory disease or a chronic inflammatory disease such as rheumatoid arthritis, dermatitis, psoriasis or endometriosis.
  • 17. Use of a cationic liposomal preparation comprising at least one cationic lipid from about 30 mole % to about 99.9 mole %, an active agent in an amount of at least about 0.1 mole % and at least one neutral and/or anionic lipid from about 0 mole % to about 70 mole % for manufacturing a pharmaceutical composition for the prevention or treatment of disorders associated with and/or accompanied by the occurrence of drug resistant cells, e.g. for the prevention or treatment of drug-resistant tumors.
  • 18 The use of claim 17 as a second or third line treatment, particularly for cancer.
  • 19. The use of claim 17, wherein said cationic liposomal preparation comprises 50:47:3 mole % of DOTAP, DOPC and paclitaxel.
  • 20. Use of a cationic liposomal preparation comprising at least one cationic lipid from about 30 mole % to about 99.9 mole %, an active agent in an amount of at least about 0.1 mole % and at least one neutral and/or anionic lipid from about 0 mole % to about 70 mole % for manufacturing a pharmaceutical composition for the prevention or treatment of metastasis formation, e.g. onset and/or progression, particularly associated with and/or accompanied by a tumor disorder.
  • 21. The use of claim 20 for manufacturing a pharmaceutical composition for the prevention or treatment of liver metastasis formation.
  • 22. Use of a cationic liposomal preparation comprising at least one cationic lipid from about 30 mole % to about 99.9 mole %, an active agent in an amount of at least about 0.1 mole % and at least one neutral and/or anionic lipid from about 0 mole % to about 70 mole % for manufacturing a pharmaceutical composition for simultaneous, separate, or sequential combination therapy with a jointly effective dose of at least one further active agent and/or heat and/or radiation and/or cryotherapy against metastasis onset and/or progression, e.g. associated with and/or accompanied by the tumors.
  • 23. The use of claim 22, wherein the composition is for simultaneous combination therapy with a jointly effective dose of at least one further active agent.
  • 24. The use of claim 17, wherein said active agent is selected from a cytotoxic or cytostatic substance such as an anti-tumor or an anti-endothelial cell active substance, a chemotherapeutic agent or an immunological active substance.
  • 25. The use of claim 20, wherein said cationic liposomal preparation comprises 50:47:3 mole % of DOTAP, DOPC and paclitaxel.
  • 26. The use of claim 20, wherein said active agent is selected from a taxane, a camptothecin, a statin, a depsipeptide, thalidomide, other agents interacting with microtubuli such as discodermolide, laulimalide, isolaulimalide, eleutherobin, Sarcodictyin A and B, and in a most preferred embodiment it is selected from paclitaxel, docetaxel, camptothecin or any derivative thereof.
  • 27. The use of claim 9, wherein said further active agent is an anti-endothelial cell active substance, an anti-tumor active substance, a chemotherapeutic agent, an immunological active substance, a compound that reduces or eliminates hypersensitivity reactions or a chemosensitizer.
  • 28. The use of claim 9, wherein said further active agent is selected from antineoplastic agents especially antimitotic agents like paclitaxel, alkylating agents especially platinum containing compounds like cisplatin, carboplatin, DNA topoisomerase inhibiting agents like camptothecin or doxorubicin, RNA / DNA antimetabolites, especially 5-fluorouracil or gemcitabine and other compounds having antitumor activity.
  • 29. The use of claim 27, wherein said compound that reduces or eliminates hypersensitivity reactions is selected from the group comprising steroids, antihistamines, H2 receptor antagonists, and combinations thereof in a sufficient amount to prevent fatal anaphylactic reactions.
  • 30. The use of claim 28, wherein said compound is selected from the group comprising Ranitidine, Dexamethasone, Diphenhydramine, Famotidine, Hydrocortisone, Clemastine, Cimetidine, Prednisolone, Chlorpheniramine, Chlorphenamine, Dimethindene maleate, and Promethazine.
  • 31. The use of claim 27, wherein said chemosensitzier is selected from the group comprising cell cycle modulators, substances that revert a drug resistance like verapamil, vasoactive substances like anti-hypertensive drugs, substances that modify interactions of cationic liposomes with blood components like protamine.
  • 32. The use of claim 1 for the treatment of cancer, especially pancreatic cancer, inoperable pancreatic cancer, gastro-intestinal cancer, lung cancer, colorectal or gastric cancer, breast cancer, prostate cancer and melanoma.
  • 33. The use of claim 1, wherein said cationic liposomal preparation comprises liposomes having an average particle diameter from about 25 nm to about 500 nm, preferably about 100 nm to about 300 nm.
  • 34. The use of claim 1, wherein said cationic liposomal preparation is administered systemically, preferably intravenously.
  • 35. Use of a cationic liposomal preparation comprising at least one cationic lipid from about 30 mole % to about 99.9 mole %, paclitaxel in an amount of at least about 0.1 mole % and at least one neutral and/or anionic lipid from about 0 mole % to about 70 mole % for manufacturing a pharmaceutical composition for administering to a human patient in need thereof at a monthly dose of about 9 mg up to about 2337 mg of paclitaxel/m2 body surface of said human patient.
Priority Claims (2)
Number Date Country Kind
03023539.4 Oct 2003 EP regional
04000361.8 Jan 2004 EP regional
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/EP04/11655 10/15/2004 WO 00 9/28/2007