WO2001026684A2 - Method for treating hypoxic cells - Google Patents

Method for treating hypoxic cells Download PDF

Info

Publication number
WO2001026684A2
WO2001026684A2 PCT/EP2000/010422 EP0010422W WO0126684A2 WO 2001026684 A2 WO2001026684 A2 WO 2001026684A2 EP 0010422 W EP0010422 W EP 0010422W WO 0126684 A2 WO0126684 A2 WO 0126684A2
Authority
WO
WIPO (PCT)
Prior art keywords
process according
cancer
cells
hypoxic
chosen
Prior art date
Application number
PCT/EP2000/010422
Other languages
French (fr)
Other versions
WO2001026684A3 (en
Inventor
Constance E. Medlen
Original Assignee
Aventis Pharma S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aventis Pharma S.A. filed Critical Aventis Pharma S.A.
Priority to AU13864/01A priority Critical patent/AU1386401A/en
Publication of WO2001026684A2 publication Critical patent/WO2001026684A2/en
Publication of WO2001026684A3 publication Critical patent/WO2001026684A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0038Radiosensitizing, i.e. administration of pharmaceutical agents that enhance the effect of radiotherapy

Definitions

  • This invention relates to the use of SN-38 to potentiate the cytotoxic effect of radiation on hypoxic cell populations.
  • chemotherapeutic drugs are known to be able to enhance the cytotoxicity of ionizing radiation.
  • Widely used chemotherapeutic agents including 5-fluorouracil, etoposide, ad amycin, vinblastine, mytomycin C, cisplatin, bleomycin, and paclitaxel, have all been shown to mediate radiosensitization effects via different mechanisms.
  • Camptothecin belongs to a group of anticancer agents with a unique mechanism of action. It inhibits topoisomerase I through the formation of a stable topoisomerase l-DNA cleavable complex. It is a potent cytotoxic agent that is active against a number of experimental tumors. However, some clinical trials with camptothecin have been terminated owing to a severe toxicity profile.
  • Irinotecan also known as CPT-11 (7-ethyl-10-[4-(1- piperidino)-1 -piperidino]carbonyloxycamptothecin) is a semisynthesized derivative of camptothecin. It has broad antitumor activities and has demonstrated good results in clinical trials against lung, colorectal, cervical, and ovarian carcinomas. Irinotecan is converted by the enzyme carboxylesterase to a more active anti- tumor compound, SN-38 (7-ethyl-10-hydroxycamptothecin), a metabolite of CPT-11 , which has been reported to be 200 to 1000 times more potent than irinotecan (Kawato et al.. Cancer Res.
  • hypoxic cells in solid tumors limit the therapeutic efficacy of radiotherapy of tumors, including carcinomas of the larynx and pharynx
  • Hypoxia refers to a condition whereby body tissues are obtaining insufficient oxygen, for reasons including lack of sufficient blood supply.
  • One aspect of the invention relates to a process for treating cancers wherein at least a portion of the cancer cells are hypoxic, including digestive tract cancers such as, for example, colorectal, gastrointestinal, laryngeal, pharyngeal, and esophageal cancers; and solid tumors such as carcinomas, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and suprotentorial PNEUT, said process comprising administering to a patient in need thereof a radiosensitizing effective amount of SN-38, followed by treatment with radiation.
  • Another aspect of the invention is a process for sensitizing hypoxic cells to radiation, comprising administering to said hypoxic cells a radiosensitizing effective amount of SN-38.
  • An additional aspect of the invention is a process for potentiating the cytotoxic effect of radiation on a carcinoma cell line under hypoxic conditions, said process comprising administering to said cell line a potentiating effective amount of SN-38.
  • Still another aspect of the invention is a process for inhibiting hypoxic cell growth comprising administering to a patient in need thereof an effective amount of SN-38.
  • Yet another aspect of the invention is a process for determining the radiosensitizing effect of a compound under hypoxic conditions, said process comprising providing a modular incubator chamber having at least one layer of wax cast in an amount effective to simulate living tissue ; inserting into said modulator incubator chamber at least one container containing : (A) a number of cells upon which the radiosensitizing potential of a compound will be tested ; and (B) the compound for which the radiosensitizing effect is to be determined into the modular incubator chamber ; creating hypoxic conditions in said modular incubator chamber ; sealing said modular incubator chamber ; and subjecting the modular incubator chamber to irradiation.
  • Figure 1 shows survival curves of WH03 cells after a 6 day exposure to irinotecan (Fig. 1A) and SN-38 (Fig. 1 B).
  • WH03 is an esophageal cancer cell line.
  • Each point of Figure 1 represents the mean of three experiments with the standard error.
  • Figure 2 shows growth fractions of WH03 cells following irradiation under aerobic ( ⁇ ) and hypoxic ( ⁇ ) conditions.
  • Figure 3 shows growth fractions of WH03 cells following irradiation under hypoxic conditions in the presence of 3.1 :M irinotecan (T). The control irradiation response under hypoxia ( ⁇ ) is shown for comparison. Each point represents the mean of those three experiments with the standard error.
  • Figure 4 shows growth fractions of WH03 cells following irradiation under hypoxic conditions in the presence of 0.045 nM SN-38 (T) or 0.92 nM SN-38 (•).
  • T 0.045 nM SN-38
  • 0.92 nM SN-38
  • Figure 5 shows a modular incubator chamber having a layer of wax cast on the inside and the outside of the chamber.
  • the test tubes are embedded in holes cut into a piece of polystyrene, which is about 5 cm thick and the approximate diameter of the chamber.
  • the polystyrene rests on the top of the inside layer of the wax. This chamber allows for the determination of the radiosensitizing effect of a compound under aerobic or hypoxic conditions.
  • Figures 6A and 6B show the cytotoxic effects of irinotecan and SN-38 on WH03 cells without irradiation.
  • One aspect of the invention relates to the cytotoxicity and radiosensitizing potential of the topoisomerase I modulator SN-38 on digestive tract cancers such as, for example, colorectal, gastrointestinal, laryngeal, pharyngeal, and esophageal cancers; and solid tumors such as carcinomas, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and suprotentorial PNEUT. It has now been found that SN-38 is 1000 times more active than irinotecan against the human carcinoma cell line WH03. This is in agreement with Kawato et al., Cancer Res.
  • SN-38 The active metabolite of irinotecan, SN-38, induces hypoxia-selective radiosensitization of a rapidly proliferating human esophageal cancer cell line at submicromolar concentrations.
  • SN-38 may be clinically useful in combination with radiation in hypoxic conditions in the treatment of cancers such as digestive tract cancers, for example, colorectal, gastrointestinal, laryngeal, pharyngeal, and esophageal cancers; and solid tumors such as carcinomas, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and suprotentorial PNEUT.
  • a cytotoxic effect of SN- 38 may be administered to a patient without radiation treatment.
  • the present inventor has found that SN-38 significantly controls cell proliferation in hypoxic cell populations even without radiation co-therapy. For example, it has been found that SN-38 exerts greater than 25% control over a hypoxic WH03 cell population at a concentration as low as 0.046 nM (See Fig. 6).
  • SN-38 may be administered to a patient in an amount effective to induce a cytotoxic effect, which amount may be readliy determined through routine experimentation by one of ordinary skill in the art. For example, SN-38 may be administered to a patient in an amount ranging from 100 to 1000 mg/m 2 in order to induce a cytotoxic effect.
  • a combination therapy comprising SN- 38 and radiation may be useful for treating digestive tract cancers such as, for example, laryngeal cancer, pharyngeal cancer, esophageal cancer, colorectal cancer, and gastrointestinal cancer.
  • an effective amount of at least one additional chemotherapeutic agent may be administered with a radiosensitizing or cytotoxic effective amount of SN-38.
  • Suitable chemotherapeutic agents include any agent known to be useful for the treatment of solid tumors and cancer generally, and such agents may include, for example, 5-fluorouracil, etoposide, adriamycin, vinblastine, mytomycin C, cisplatin, bieomycin, and paclitaxel.
  • the additional chemotherapeutic agent can be administered simultaneously or sequentially with SN-38 by the same or different routes of administration.
  • the additional chemotherapeutic agent can be administered in an amount effective to treat cancer, including an amount effective to potentiate the cytotoxic or radiosensitizing amount of SN-38.
  • hypoxic cell radiosensitizer potent enough to radiosensitize hypoxic cells at clinically acceptable doses.
  • various procedures have been utilized to achieve an in vitro hypoxic atmosphere (Zeman et al., Radiat. Res. 122:72-76, (1990); Hentosh, Analvt. Biochem. 210:249-252, (1993)). These methods were mostly time consuming and were not suitable for high output.
  • the present inventor has found that such regularly encountered problems can be eliminated by the use of a standard modular incubator chamber having a layer of wax equivalent to an amount of soft tissue cast on the inside and/or outside of the modular incubator chamber.
  • Multiple test tubes containing monolayers of proliferating cells may be grouped together for each radiation dosage. In this way, it is possible to perform various drug treatments within any one experiment with an oxygen enhancement ratio (OER) of 2.1.
  • OER oxygen enhancement ratio
  • Cell cultures may be plated in containers such as 5 ml glass tubes at densities of, for example, 600 cells per tube, and incubated for 24 hours at 37°C in a humidified atmosphere of 5% CO2. Solutions of either irinotecan or SN-38 are then added to the tubes before radiation. The tubes are then placed into a modular incubator chamber (Billups-
  • cell cultures are placed in an incubator at 37°C and 5% CO2 for a further period of time, for example 4 hours, after which the medium containing the drugs is replaced with complete medium and the tubes incubated for a further period of time, for example, 6 days. Cell survival may then be assessed using the MTT assay.
  • the compounds and/or compositions administered according to the invention can be administered orally and/or parenterally, such as intravenously or intraperitoneally. According to a certain embodiment, the compounds and/or compositions are administered via hepatic arterial infusion.
  • compositions for oral administration comprise tablets, pills, powders, or granules.
  • the active product according to the invention is mixed with one or more inert diluents, such as sucrose, lactose, or starch.
  • these compositions can comprise substances other than diluents, for example, a lubricant such as magnesium stearate.
  • compositions for oral administration can be used as liquid compositions for oral administration.
  • inert diluents such as water or liquid paraffin
  • compositions can also comprise substances other than diluents, for example, wetting, sweetening or flavoring products.
  • compositions for parenteral administration according to the invention can be aqueous or nonaqueous sterile solutions, suspensions, or emulsions.
  • Propylene glycol, a polyethylene glycol, plant oils, such as olive oil, or injectable organic esters, for example, ethyl oleate can be used as a solvent or vehicle.
  • These compositions can also contain adjuvants, such as wetting agents, emulsifiers, and dispersing agents. Sterilization can be carried out in many ways, for example, using a bacteriological filter, by incorporating sterilizing agents into the composition, or by heating.
  • These compositions for parenteral use can also be prepared in the form of sterile solid compositions which can be dissolved at the time of use in sterile water or any other injectable sterile medium.
  • the doses used to carry out the methods according to the invention are those which allow a maximum therapeutic response in terms of radiosensitization and cytotoxicity.
  • the doses vary according to the form of administration, the particular product selected, and the personal characteristics of the individual to be treated. In general, the doses are those which are therapeutically effective for the treatment of disorders due to abnormal cell proliferation, and in particular a cytostatic treatment.
  • the products according to the invention can be administered as often and for as long as necessary to obtain the desired therapeutic effect.
  • the doses range from 100 to 1000 mg/m 2 .
  • the therapeutic dose is approximately 80 mg/m 2 /day for 5 consecutive days, or 30 mg/m /day for 14 consecutive days. It is understood that, in order to select the most appropriate dosage, the route of administration, the patient's weight, general state of health, age, and all the factors which may influence the effectiveness of the treatment will have to be taken into account. In general, the doctor will make this determination based on such factors relevant to the individual to be treated. The example which follows is given by way of non-limiting illustration of the present invention.
  • the following example demonstrates the potential of irinotecan and SN-38 in enhancing the cytotoxic effects of radiation on a human esophageal carcinoma cell line under both aerobic and hypoxic conditions.
  • irinotecan and SN-38 supplied by Aventis Pharma S.A., were solubilized in dimethylsulfoxide at a concentration of 2 mg/ml and diluted in medium to the required concentration immediately before use.
  • WH03 human esophageal carcinoma cell line
  • MEM Minimum Essential Medium
  • FCS heat inactivated fetal calf serum
  • the chamber was placed on a shaker incubator for 30 minutes, continuously passing a mixture of 5% C ⁇ 2 /95% N 2 through said chamber before sealing.
  • the cells were then incubated for a further 1 h at 37°C to deplete residual oxygen by cellular and respiratory metabolism before irradiation (1-10 Gy).
  • irradiation performed at room temperature using an 8 MV photon beam, cell cultures were placed in an incubator at 37°C and 5% CO2 for a further 4 hours after which the medium containing the drugs was replaced with complete medium and the tubes incubated for a further 6 days. Cell survival was assessed using the MTT assay.
  • Figure 1 which shows the cell survival curves of WH03 monolayer cultures exposed to either irinotecan or SN-38 for 6 days as a function of the untreated control cells
  • WH03 cells were only marginally sensitive to irinotecan (activity between 1.5 and 2.5 :M) (Figure 1A)
  • SN-38 possessed activity against WH03 cells at concentration of between 0.003 and 0.012 :M ( Figure 1 B).
  • the dose modifying factor for irinotecan at 3.1 :M was 1.5 and for SN-38 at 0.046 and 0.092 nM were 1.6 and 2.1 respectively (Table 1 ).
  • Table 1 The dose modifying factor for irinotecan at 3.1 :M was 1.5 and for SN-38 at 0.046 and 0.092 nM were 1.6 and 2.1 respectively (Table 1 ).
  • Mean inactivation doses are calculated from the response of WH03 cells following treatment with radiation and/or different drugs under aerobic or hypoxic conditions.
  • the dose modifying factor is stated as the ratio of mean inactivation doses.

Abstract

The present invention relates to the cytotoxic and radiosensitizing potential of SN-38 (7-ethyl-10-hydroxycamptothecin), a metabolite of CPT-11. SN-38 has been found to be useful for sensitizing hypoxic cells, such as hypoxic cancer cells, to the effects of radiation.

Description

METHOD FOR TREATING HYPOXIC CELLS
BACKGROUND OF THE INVENTION
Field of the Invention
This invention relates to the use of SN-38 to potentiate the cytotoxic effect of radiation on hypoxic cell populations.
Discussion of the Related Art
The combination of chemotherapy and radiation therapy has become the treatment of choice for a number of advanced human malignancies. Although systemic chemotherapy ideally may control metastatic disease, radiation therapy provides an effective local control at primary tumor sites. A number of chemotherapeutic drugs are known to be able to enhance the cytotoxicity of ionizing radiation. Widely used chemotherapeutic agents, including 5-fluorouracil, etoposide, ad amycin, vinblastine, mytomycin C, cisplatin, bleomycin, and paclitaxel, have all been shown to mediate radiosensitization effects via different mechanisms. However, the potential excessive toxicities imposed by combined modality treatments of normal tissues have dictated the achievable doses and limited their potential efficacies in eradicating cancers. The development of new radiosensitizers that can selectively enhance the cytotoxicity of ionizing radiation in cancer cells remains a challenge in the art of oncology. Camptothecin belongs to a group of anticancer agents with a unique mechanism of action. It inhibits topoisomerase I through the formation of a stable topoisomerase l-DNA cleavable complex. It is a potent cytotoxic agent that is active against a number of experimental tumors. However, some clinical trials with camptothecin have been terminated owing to a severe toxicity profile. Irinotecan, also known as CPT-11 (7-ethyl-10-[4-(1- piperidino)-1 -piperidino]carbonyloxycamptothecin) is a semisynthesized derivative of camptothecin. It has broad antitumor activities and has demonstrated good results in clinical trials against lung, colorectal, cervical, and ovarian carcinomas. Irinotecan is converted by the enzyme carboxylesterase to a more active anti- tumor compound, SN-38 (7-ethyl-10-hydroxycamptothecin), a metabolite of CPT-11 , which has been reported to be 200 to 1000 times more potent than irinotecan (Kawato et al.. Cancer Res. 51 :4187-4191. (1991)). Both irinotecan and SN-38 potentiate the cytotoxic effect of radiation on various carcinoma cell lines under aerobic conditions ((Omura et al., Radiother. Oncol. 43:197- 201 , (1997) ; Sasai et al., Int. J. Radiat. Oncol. Biol. Phvs. 42:785-788, (1998); Chen et al., Cancer Res. 57:1529-1536, (1997)).
Hypoxic cells in solid tumors limit the therapeutic efficacy of radiotherapy of tumors, including carcinomas of the larynx and pharynx
(Overgaard et al., Int. J. Radiat. Oncol. Biol. Phvs. 12:515-521 , (1986)). The identification of drugs that act preferentially on hypoxic cells by sensitizing them to radiation is a challenge to researchers in this field.
SUMMARY OF THE INVENTION Hypoxia refers to a condition whereby body tissues are obtaining insufficient oxygen, for reasons including lack of sufficient blood supply.
One aspect of the invention relates to a process for treating cancers wherein at least a portion of the cancer cells are hypoxic, including digestive tract cancers such as, for example, colorectal, gastrointestinal, laryngeal, pharyngeal, and esophageal cancers; and solid tumors such as carcinomas, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and suprotentorial PNEUT, said process comprising administering to a patient in need thereof a radiosensitizing effective amount of SN-38, followed by treatment with radiation. Another aspect of the invention is a process for sensitizing hypoxic cells to radiation, comprising administering to said hypoxic cells a radiosensitizing effective amount of SN-38.
An additional aspect of the invention is a process for potentiating the cytotoxic effect of radiation on a carcinoma cell line under hypoxic conditions, said process comprising administering to said cell line a potentiating effective amount of SN-38. Still another aspect of the invention is a process for inhibiting hypoxic cell growth comprising administering to a patient in need thereof an effective amount of SN-38.
Yet another aspect of the invention is a process for determining the radiosensitizing effect of a compound under hypoxic conditions, said process comprising providing a modular incubator chamber having at least one layer of wax cast in an amount effective to simulate living tissue ; inserting into said modulator incubator chamber at least one container containing : (A) a number of cells upon which the radiosensitizing potential of a compound will be tested ; and (B) the compound for which the radiosensitizing effect is to be determined into the modular incubator chamber ; creating hypoxic conditions in said modular incubator chamber ; sealing said modular incubator chamber ; and subjecting the modular incubator chamber to irradiation.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows survival curves of WH03 cells after a 6 day exposure to irinotecan (Fig. 1A) and SN-38 (Fig. 1 B). WH03 is an esophageal cancer cell line. Each point of Figure 1 represents the mean of three experiments with the standard error.
Figure 2 shows growth fractions of WH03 cells following irradiation under aerobic (■) and hypoxic (▼) conditions.
Figure 3 shows growth fractions of WH03 cells following irradiation under hypoxic conditions in the presence of 3.1 :M irinotecan (T). The control irradiation response under hypoxia (■) is shown for comparison. Each point represents the mean of those three experiments with the standard error.
Figure 4 shows growth fractions of WH03 cells following irradiation under hypoxic conditions in the presence of 0.045 nM SN-38 (T) or 0.92 nM SN-38 (•). The control irradiation response under hypoxia (■) is shown for comparison. Each point represents the mean of these three experiments with the standard error.
Figure 5 shows a modular incubator chamber having a layer of wax cast on the inside and the outside of the chamber. The test tubes are embedded in holes cut into a piece of polystyrene, which is about 5 cm thick and the approximate diameter of the chamber. The polystyrene rests on the top of the inside layer of the wax. This chamber allows for the determination of the radiosensitizing effect of a compound under aerobic or hypoxic conditions.
Figures 6A and 6B show the cytotoxic effects of irinotecan and SN-38 on WH03 cells without irradiation.
DETAILED DESCRIPTION OF THE INVENTION
One aspect of the invention relates to the cytotoxicity and radiosensitizing potential of the topoisomerase I modulator SN-38 on digestive tract cancers such as, for example, colorectal, gastrointestinal, laryngeal, pharyngeal, and esophageal cancers; and solid tumors such as carcinomas, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and suprotentorial PNEUT. It has now been found that SN-38 is 1000 times more active than irinotecan against the human carcinoma cell line WH03. This is in agreement with Kawato et al., Cancer Res. 51 :4187-4191 , (1991 ), who reported SN-38 to be anywhere between 200 and 1000 times more potent than irinotecan. Irinotecan and SN-38, at subtoxic concentrations, did not sensitize proliferating WH03 cells under aerobic conditions. This is consistent with the findings of Falk et al., Int. J. Radiat. Biol. 61 :749-757, (1992), and Omuru et al., Radiother. Oncol. 43:197-201 (1997), who reported that the radiosensitizing effects of camptothecin and SN-38 are restricted to Go phase cells and thus are ineffective against rapidly proliferating tumor populations. However, it has now been found that SN-38, but not irinotecan, sensitizes proliferating WH03 cells to radiation under hypoxic conditions. A concentration of SN-38 as low as 0.046 nM increased the radiation-induced cell lethality significantly. The combined effect of radiation and SN-38 under hypoxic conditions has not previously been described.
The active metabolite of irinotecan, SN-38, induces hypoxia-selective radiosensitization of a rapidly proliferating human esophageal cancer cell line at submicromolar concentrations. Thus, SN-38 may be clinically useful in combination with radiation in hypoxic conditions in the treatment of cancers such as digestive tract cancers, for example, colorectal, gastrointestinal, laryngeal, pharyngeal, and esophageal cancers; and solid tumors such as carcinomas, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and suprotentorial PNEUT.
According to one embodiment of the invention, a cytotoxic effect of SN- 38 may be administered to a patient without radiation treatment. Surprisingly, the present inventor has found that SN-38 significantly controls cell proliferation in hypoxic cell populations even without radiation co-therapy. For example, it has been found that SN-38 exerts greater than 25% control over a hypoxic WH03 cell population at a concentration as low as 0.046 nM (See Fig. 6). SN-38 may be administered to a patient in an amount effective to induce a cytotoxic effect, which amount may be readliy determined through routine experimentation by one of ordinary skill in the art. For example, SN-38 may be administered to a patient in an amount ranging from 100 to 1000 mg/m2 in order to induce a cytotoxic effect.
According to one embodiment, a combination therapy comprising SN- 38 and radiation may be useful for treating digestive tract cancers such as, for example, laryngeal cancer, pharyngeal cancer, esophageal cancer, colorectal cancer, and gastrointestinal cancer.
According to another embodiment of the invention, an effective amount of at least one additional chemotherapeutic agent may be administered with a radiosensitizing or cytotoxic effective amount of SN-38. Suitable chemotherapeutic agents include any agent known to be useful for the treatment of solid tumors and cancer generally, and such agents may include, for example, 5-fluorouracil, etoposide, adriamycin, vinblastine, mytomycin C, cisplatin, bieomycin, and paclitaxel. The additional chemotherapeutic agent can be administered simultaneously or sequentially with SN-38 by the same or different routes of administration. The additional chemotherapeutic agent can be administered in an amount effective to treat cancer, including an amount effective to potentiate the cytotoxic or radiosensitizing amount of SN-38.
Over the past 20 years, there has been considerable interest in developing a hypoxic cell radiosensitizer potent enough to radiosensitize hypoxic cells at clinically acceptable doses. For these purposes various procedures have been utilized to achieve an in vitro hypoxic atmosphere (Zeman et al., Radiat. Res. 122:72-76, (1990); Hentosh, Analvt. Biochem. 210:249-252, (1993)). These methods were mostly time consuming and were not suitable for high output.
Surprisingly, the present inventor has found that such regularly encountered problems can be eliminated by the use of a standard modular incubator chamber having a layer of wax equivalent to an amount of soft tissue cast on the inside and/or outside of the modular incubator chamber. Multiple test tubes containing monolayers of proliferating cells may be grouped together for each radiation dosage. In this way, it is possible to perform various drug treatments within any one experiment with an oxygen enhancement ratio (OER) of 2.1.
Cell cultures may be plated in containers such as 5 ml glass tubes at densities of, for example, 600 cells per tube, and incubated for 24 hours at 37°C in a humidified atmosphere of 5% CO2. Solutions of either irinotecan or SN-38 are then added to the tubes before radiation. The tubes are then placed into a modular incubator chamber (Billups-
Rothenburg Inc., Del Mar, California, USA) with a wax build-up equivalent to approximately 2 cm tissue (8 MV dmaχ) cast into the bottom of the chamber. The chamber may then be placed on a shaker incubator for 30 minutes while continuously passing either a mixture of 5% Cθ2/95% air for aerobic conditions, or 5% Cθ2/95% N for hypoxic conditions, through said chamber before sealing. The cells may then incubated for a further period of time, for example 1 hour, at approximately 37°C, in order to deplete residual oxygen by cellular and respiratory metabolism before irradiation (1 -10 Gy).
Immediately after irradiation, performed at room temperature using an 8 MV photon beam, cell cultures are placed in an incubator at 37°C and 5% CO2 for a further period of time, for example 4 hours, after which the medium containing the drugs is replaced with complete medium and the tubes incubated for a further period of time, for example, 6 days. Cell survival may then be assessed using the MTT assay.
The compounds and/or compositions administered according to the invention can be administered orally and/or parenterally, such as intravenously or intraperitoneally. According to a certain embodiment, the compounds and/or compositions are administered via hepatic arterial infusion.
The compositions for oral administration comprise tablets, pills, powders, or granules. In these compositions, the active product according to the invention is mixed with one or more inert diluents, such as sucrose, lactose, or starch. These compositions can comprise substances other than diluents, for example, a lubricant such as magnesium stearate.
Pharmaceutically acceptable emulsions, solutions, suspensions, syrups, or elixirs containing inert diluents, such as water or liquid paraffin, can be used as liquid compositions for oral administration. These compositions can also comprise substances other than diluents, for example, wetting, sweetening or flavoring products.
The compositions for parenteral administration according to the invention can be aqueous or nonaqueous sterile solutions, suspensions, or emulsions. Propylene glycol, a polyethylene glycol, plant oils, such as olive oil, or injectable organic esters, for example, ethyl oleate, can be used as a solvent or vehicle. These compositions can also contain adjuvants, such as wetting agents, emulsifiers, and dispersing agents. Sterilization can be carried out in many ways, for example, using a bacteriological filter, by incorporating sterilizing agents into the composition, or by heating. These compositions for parenteral use can also be prepared in the form of sterile solid compositions which can be dissolved at the time of use in sterile water or any other injectable sterile medium.
The doses used to carry out the methods according to the invention are those which allow a maximum therapeutic response in terms of radiosensitization and cytotoxicity. The doses vary according to the form of administration, the particular product selected, and the personal characteristics of the individual to be treated. In general, the doses are those which are therapeutically effective for the treatment of disorders due to abnormal cell proliferation, and in particular a cytostatic treatment. The products according to the invention can be administered as often and for as long as necessary to obtain the desired therapeutic effect.
Generally, in humans, the doses range from 100 to 1000 mg/m2. In a certain embodiment, the therapeutic dose is approximately 80 mg/m2/day for 5 consecutive days, or 30 mg/m /day for 14 consecutive days. It is understood that, in order to select the most appropriate dosage, the route of administration, the patient's weight, general state of health, age, and all the factors which may influence the effectiveness of the treatment will have to be taken into account. In general, the doctor will make this determination based on such factors relevant to the individual to be treated. The example which follows is given by way of non-limiting illustration of the present invention.
EXAMPLE
The following example demonstrates the potential of irinotecan and SN-38 in enhancing the cytotoxic effects of radiation on a human esophageal carcinoma cell line under both aerobic and hypoxic conditions.
Both irinotecan and SN-38, supplied by Aventis Pharma S.A., were solubilized in dimethylsulfoxide at a concentration of 2 mg/ml and diluted in medium to the required concentration immediately before use.
WH03 (human esophageal carcinoma cell line) was maintained in a mixture of MEM (Minimal Essential Medium) and HamsF12 supplemented with 10% heat inactivated fetal calf serum (FCS) and 0.1 mg/ml of penicillin and streptomycin.
This was performed using a metabolic assay based on the reactivity of MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide) (Sigma Diagnostics Inc.) with viable cells (Twentyman et al., Radiother. Oncol. 43:197-201 (1992)). Cells were seeded at 3 x 103/well in 96-well microtiter plates in a final volume of 200:1 complete medium in the presence or absence of different concentrations of either irinotecan or SN-38.
Cell cultures were plated in 5 ml glass tubes at densities of 600 cells per tube and incubated for 24 hours at 37°C in a humidified atmosphere of 5% C02. Solutions of either irinotecan (3.1 :M) or SN-38 (0.046 nM and 0.092 nM) were added to the tubes before radiation. The concentrations of the experimental compounds used caused 25% or less inhibition of cell growth under the experimental conditions used without the effect of radiation. The tubes were placed into a modular incubator chamber (Billups-Rothenburg Inc., Del Mar, California, USA) with a wax build-up equivalent to 2 cm tissue (8 MV dmax) cast into the bottom of the chamber. The chamber was placed on a shaker incubator for 30 minutes, continuously passing a mixture of 5% Cθ2/95% N2 through said chamber before sealing. The cells were then incubated for a further 1 h at 37°C to deplete residual oxygen by cellular and respiratory metabolism before irradiation (1-10 Gy). Immediately after irradiation, performed at room temperature using an 8 MV photon beam, cell cultures were placed in an incubator at 37°C and 5% CO2 for a further 4 hours after which the medium containing the drugs was replaced with complete medium and the tubes incubated for a further 6 days. Cell survival was assessed using the MTT assay.
Results
The cell growth fraction (S), relative to that of control samples, for cells treated with a dose (D) of radiation, was fitted to a linear-quadratic equation as loge= -AxD - BxD2 - C. In cases where one of the inactivation parameters
(A or B) was found to be negative, it was set to zero and the other parameter re-estimated. Overall cellular responses were expressed in terms of the mean inactivation dose calculated from the respective inactivation parameters (Kellerer et al., Encvclop. Med. Radiol. Ill 1 :42, (1972)). This parameter quantifies radiosensitivity in a one-dimensional manner and is proportional to the area under the dose-response curve. As the mean inactivation dose is determined predominantly by the response of cells within the first order of magnitude, it is particularly suitable for use with the current set of data where cell growth in most measurements was not less than 10% of control samples. Furthermore, the ratio of mean inactivation dose values obtained with the MTT assay yield dose modifying factors similar to those obtained using conventional colony forming methods (Stabbert et al., Strahlenther. Onkol. 10:567-572, (1996)).
As displayed in Figure 1 , which shows the cell survival curves of WH03 monolayer cultures exposed to either irinotecan or SN-38 for 6 days as a function of the untreated control cells, WH03 cells were only marginally sensitive to irinotecan (activity between 1.5 and 2.5 :M) (Figure 1A), whereas SN-38 possessed activity against WH03 cells at concentration of between 0.003 and 0.012 :M (Figure 1 B).
Exposure of WH03 cells to hypoxic conditions greatly reduced the sensitivity of WH03 cells for radiation (Figure 2). The mean oxygen enhancement ratio for these experiments was 2.1 (Table 1 ).
Irinotecan at 3.1 :M and SN-38 at 0.046 and 0.092 nM had no effect on the sensitivity of WH03 cells for radiation under aerobic conditions (results not shown). However, under hypoxic conditions, SN-38, but not irinotecan, was able to sensitize WH03 cells to radiation at 0.092 nM (Figures 3 and 4). An additive effect was observed between radiation and either SN-38 at 0.046 nM or irinotecan at 3.1 :M (Figures 3 and 4).
The dose modifying factor for irinotecan at 3.1 :M was 1.5 and for SN-38 at 0.046 and 0.092 nM were 1.6 and 2.1 respectively (Table 1 ). Table 1
Mean inactivation doses are calculated from the response of WH03 cells following treatment with radiation and/or different drugs under aerobic or hypoxic conditions. The dose modifying factor is stated as the ratio of mean inactivation doses.
Figure imgf000012_0001
The present invention may be embodied in other specific forms without departing from its spirit or essential characteristics. The described embodiments are to be considered in all respects as illustrative only and not restrictive. The scope of the invention is, therefore, indicated by the appended claims, rather than by the foregoing description. All changes which come within the meaning and range of equivalency of the claims are to be embraced within their scope.

Claims

1. A process for treating solid tumors wherein at least a portion of the tumor cells are hypoxic, said process comprising administering to a patient in need thereof a radiosensitizing effective amount of SN-38, followed by treatment with radiation.
2. The process according to claim 1 , wherein the tumor cells are chosen from digestive tract cancer cells.
3. The process according to claim 2, wherein the digestive tract cancer is chosen from laryngeal cancer, pharyngeal cancer, esophageal cancer, colorectal cancer, and gastrointestinal cancer.
4. The process according to claim 1 , wherein the tumor cells are chosen from carcinomas, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and suprotentorial PNEUT.
5. The process according to claim 1 , wherein the tumor cells are esophageal cancer cells.
6. The process according to claim 1 , wherein the SN-38 is administered in an amount ranging from 100 to 1000 mg/m2.
7. The process according to claim 1 , wherein the SN-38 is administered in an amount of 80 mg/m2/day for 5 consecutive days.
8. The process according to claim 1 , wherein the SN-38 is administered in an amount of 30 mg/m2/day for 14 consecutive days.
9. A process for sensitizing hypoxic cells to radiation, comprising administering to said hypoxic cells a radiosensitizing effective amount of SN-38.
10. The process according to claim 9, wherein said hypoxic cells are cancer cells.
11. The process according to claim 9, wherein the cancer cells are chosen from digestive tract cancer cells.
12. The process according to claim 11 , wherein the digestive tract cancer is chosen from laryngeal cancer, pharyngeal cancer, esophageal cancer, colorectal cancer, and gastrointestinal cancer.
13. The process according to claim 9, wherein the hypoxic cells are cells chosen from carcinomas, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and suprotentorial PNEUT.
14. The process according to claim 9, wherein said hypoxic cells are esophageal cancer cells.
15. A process for potentiating the cytotoxic effect of radiation on a carcinoma cell line under hypoxic conditions, said process comprising administering to said cell line a potentiating effective amount of SN-38.
16. The process according to claim 15, wherein the SN-38 is administered at a concentration ranging from 0.04 to 1.00 nM.
17. The process for inhibiting hypoxic cell growth comprising administering to a patient in need thereof an effective amount of SN-38.
18. The process according to claim 17, wherein the hypoxic cell growth is a cancerous cell growth.
19. The process according to claim 17, wherein the hypoxic cell growth is chosen from digestive tract cancer cells.
20. The process according to claim 19, wherein the digestive tract cancer is chosen from laryngeal cancer, pharyngeal cancer, esophageal cancer, colorectal cancer, and gastrointestinal cancer.
21. The process according to claim 17, wherein the hypoxic cell growth is chosen from carcinomas, neuroblastoma, medulloblastoma, rhabdomyosarcoma, and suprotentorial PNEUT.
22. The process according to claim 17, wherein the SN-38 is administered in an amount ranging from 100 to 1000 mg/m2.
23. A process for determining the radiosensitizing effect of a compound, said process comprising providing a modular incubator chamber containing at least one layer of wax in an amount effective to simulate living tissue ; inserting into said modulator incubator chamber at least one container containing :
(A) a number of cells upon which the radiosensitizing potential of a compound will be tested; and
(B) the compound for which the radiosensitizing effect is to be determined into the modular incubator chamber ; sealing said modular incubator chambe r; and subjecting the modular incubator chamber to irradiation.
24. The process according to claim 23, wherein prior to sealing the modular incubator chamber hypoxic conditions are created by passing through said chamber a flow of gas comprising at least one gas chosen from air, carbon dioxide, nitrogen, and a mixture thereof.
25. The process according to claim 23, wherein prior to sealing the modular incubator chamber aerobic conditions are created by passing through said chamber a flow of gas comprising at least one gas chosen from air, carbon dioxide, nitrogen, and a mixture thereof.
26. The process according to claim 24, wherein the flow of gas through said chamber is maintained continuously for a period of time prior to said chamber being sealed.
27. The process according to claim 25, wherein the flow of gas through said chamber is maintained continuously for a period of time prior to said chamber being sealed.
28. The process according to claim 23, wherein the container is a test tube.
PCT/EP2000/010422 1999-10-14 2000-10-13 Method for treating hypoxic cells WO2001026684A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU13864/01A AU1386401A (en) 1999-10-14 2000-10-13 Method for treating hypoxic cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US15937699P 1999-10-14 1999-10-14
US60/159,376 1999-10-14

Publications (2)

Publication Number Publication Date
WO2001026684A2 true WO2001026684A2 (en) 2001-04-19
WO2001026684A3 WO2001026684A3 (en) 2001-12-27

Family

ID=22572346

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2000/010422 WO2001026684A2 (en) 1999-10-14 2000-10-13 Method for treating hypoxic cells

Country Status (2)

Country Link
AU (1) AU1386401A (en)
WO (1) WO2001026684A2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0030016A2 (en) * 1979-11-28 1981-06-10 The Wellcome Foundation Limited Method of preserving monolayers of tissue culture cells, and tissue culture cells so preserved
WO2001032210A2 (en) * 1999-10-29 2001-05-10 Pharmacyclics, Inc. Compositions for treating atheroma and neoplastic tissue

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0030016A2 (en) * 1979-11-28 1981-06-10 The Wellcome Foundation Limited Method of preserving monolayers of tissue culture cells, and tissue culture cells so preserved
WO2001032210A2 (en) * 1999-10-29 2001-05-10 Pharmacyclics, Inc. Compositions for treating atheroma and neoplastic tissue

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
EDER, J. PAUL ET AL: "Irinotecan and radiation in vitro and in vivo" INT. J. ONCOL. (1997), 11(6), 1235-1240 , XP001024436 *
MARIYA, YASUSHI ET AL: "Combined treatment effects of irradiation and SN - 38, an active metabolite of camptothecin derivative irinotecan, on the human tumor cell line PECA 4197" HIROSAKI IGAKU (1999), 51(1), 10-14 , XP001024429 *
OKISHIO, K. ET AL: "Irinotecan (CPT-11) enhances the radiosensitivity of lung cance cells in vitro" CELL. PHARMACOL. (1996), 3(4), 247-252 , XP001024397 *
OMURA, MOTOKO ET AL: "SN - 38, a metabolite of the camptothecin derivative CPT-11, potentiates the cytotoxic effect of radiation in human colon adenocarcinoma cells grown as spheroids" RADIOTHER. ONCOL. (1997), 43(2), 197-201 , XP001024384 cited in the application *
SASAI, K. ET AL: "Effects of SN - 38 (an active metabolite of CPT-11) on responses of human and rodent cells to irradiation" INT. J. RADIAT. ONCOL., BIOL., PHYS. (1998), 42(4), 785-788 , XP001024385 cited in the application *
XIE, XUEJUN ET AL: "P53 status plays no role in radiosensitizing effects of SN - 38, a camptothecin derivative" CANCER CHEMOTHER. PHARMACOL. (2000), 45(5), 362-368 , XP001024382 *

Also Published As

Publication number Publication date
AU1386401A (en) 2001-04-23
WO2001026684A3 (en) 2001-12-27

Similar Documents

Publication Publication Date Title
Chaplin et al. The effect of hydralazine on the tumor cytotoxicity of the hypoxic cell cytotoxin RSU-1069: evidence for therapeutic gain
JP2681147B2 (en) Remedy for cancerous metastasis
Wallner et al. Effect of cisplatin resistance on cellular radiation response
Saunders et al. The clinical testing of Ro 03-8799—pharmacokinetics, toxicology, tissue and tumor concentrations
Kim et al. Potentiation of radiation effects on two murine tumors by lonidamine
TWI222863B (en) Synergistic pharmaceutical compositions comprising anthracycline derivatives and anticancer agents
EP1478355B1 (en) Antitumor compositions containing taxane derivatives
Rockwell Cytotoxicities of mitomycin C and X rays to aerobic and hypoxic cells in vitro
KR20080030559A (en) Pharmaceutical compositions comprising of arsenous acid, its sodium salt and its derivatives intended for the treatment of urogenital cancer and its metastasis
Horsman et al. The potentiation of radiation damage by nicotinamide in the SCCVII tumour in vivo
CN109152839A (en) Purposes of the bipolarity trans-carotenoid together with chemotherapy and radiotherapy in treating cancer
Henk et al. Treatment of head and neck cancer with CHART and nimorazole: phase II study
Yang et al. Advanced nanomedicines for the regulation of cancer metabolism
Collingridge et al. Pentoxifylline improves the oxygenation and radiation response of BA1112 rat rhabdomyosarcomas and EMT6 mouse mammary carcinomas
TW200908976A (en) Cancer treatment combination therapy comprising vinflunine and trastuzumab
US6281223B1 (en) Radioenhanced camptothecin derivative cancer treatments
Potmesil et al. Growth inhibition of human cancer metastases by camptothecins in newly developed xenograft models
Robins et al. Phase I trial of lonidamine with whole body hyperthermia in advanced cancer
Teicher et al. Effect of Fluosol-DA® on the response of intracranial 9L tumors to X rays and BCNU
Chaplin Postirradiation modification of tumor blood flow: a method to increase the effectiveness of chemical radiosensitizers
WO2001026684A2 (en) Method for treating hypoxic cells
JP2020045329A (en) Composition comprising hydrogen gas for suppression or prevention of cancer metastasis
CN108635581A (en) A kind of composition and its application in the drug for preparing treatment tumour
Pollack et al. The efficacy of tamoxifen as an antiproliferative agent in vitro for benign and malignant pediatric glial tumors
Franchin et al. Simultaneous radiochemotherapy in the treatment of inoperable, locally advanced head and neck cancers. A single‐institution study

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)