CA2494209A1 - Radiosensitizer formulations and methods for use - Google Patents

Radiosensitizer formulations and methods for use Download PDF

Info

Publication number
CA2494209A1
CA2494209A1 CA 2494209 CA2494209A CA2494209A1 CA 2494209 A1 CA2494209 A1 CA 2494209A1 CA 2494209 CA2494209 CA 2494209 CA 2494209 A CA2494209 A CA 2494209A CA 2494209 A1 CA2494209 A1 CA 2494209A1
Authority
CA
Canada
Prior art keywords
cancer
radiosensitizer
radiotherapy
poly
administering
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA 2494209
Other languages
French (fr)
Inventor
Richard H. Matthews
Nuran Ercal
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2494209A1 publication Critical patent/CA2494209A1/en
Abandoned legal-status Critical Current

Links

Abstract

The present invention provides radiosensitizer compositions, in controlled-release formulations or other acceptable formulations, particularly nitrohistidine radiosensitizer compositions, which may be administered by any suitable means including oral, intravenous, arterial infusion, intraperitoneal, intramuscular, subcutaneous, surgical, and topical.
Optionally, radiosensitizer compositions may be formulated with other agents, including chemotherapy agents and agents that provide a synergistic radiosensitizing effect. Methods of potentiating radiotherapy cancer treatment of cancers in humans, particularly of astrocytomas, are also presented, wherein a radiosensitizer composition is administered and radiotherapy is directed to the site of the tumor. Chemotherapy regimens may also be used as adjuvant therapy.

Description

Attorney Docket: 2967/lOICA
Radiosensitizer Formulations and Methods for Use Cross Reference to Related Anulications Technical Field and Background Art The present invention relates to radiosensitizers based on histidine derivatives for use as a radiosensitizer in the treatment of cancers including lung carcinomas, breast cancers, prostate cancers, cancers of the cervix and endometrium, neuroblastomas, lymphomas, gliomas, melanomas, squamous cell carcinomas, sarcomas, adenocarcinomas, astrocytomas, to head-and-neck tumors, and GI tumors. More particularly, the invention relates to nitrohistidine, alone or in combination with other agents, prepared in slow-release formulations or normal release formulations, for the treatment of the brain tumor glioblastoma multiforme.
Histidine is one of 20 naturally occurring amino acids serving as subunits for 15 proteins. It is often present at the catalytic active site of enzymes.
Histidine is one of the nutritionally essential amino acids for mammalian cells, meaning that it cannot be synthesized by mammals, so it must be ingested and then taken up by cells to sustain protein synthesis. Rapidly-growing cells and tissues must be effective at taking up histidine from the environment, and cancer cells, in particular, usually have several very effective amino acid 20 transport systems to aid them in competing for amino acids with other cells and tissues.
Previous studies of the active transport of histidine in marine ascites tumor cells (1, 2, 3) indicate that there are two major amino acid transport systems for the uptake of histidine, and most cancer cells necessarily have both systems to permit their continued and rapid growth.
Previous studies have also indicated that nitroimidazoles have some capacity to act as 25 radiosensitizers. Examples include metronidazole (Flagyl), misonidazole, RO-07-0554, RO-I 1-3696, RO-03-8799 (Pimonidazole), SR-2508 (Etanidazole) and RSU-1069, shown in Figure 1. These compounds are sometimes referred to as "true radiosensitizers"
in that they can apparently substitute for oxygen in "fixing" radiation-induced damage of DNA, making it non-reversible and lethal.
Hypoxic cells are difficult to kill with ionizing radiation compared to normal cells because ionizing radiation requires oxygen to "fix" a lesion transiently induced in DNA by the ionizing radiation in order to kill the cell, and hypoxic cells have little to no oxygen exposure, particularly deep within a tumor. Various attempts to overcome the problem of killing hypoxic cells have been made over the past years. Administration of hyperbaric oxygen or carbogen (a mixture of oxygen and carbon dioxide) has proved problematic, even dangerous, in clinical application, and resulted in mixed results.
A limitation of the nitroimidazole radiosensitizers is that although a number have been found to be effective radiosensitizers in vitro, they have limited practical effectiveness in vivo because they are not concentrated by cancers. Only 5 of 38 clinical trials for the most extensively studied of the nitroimidazoles, misonidazole, suggested any clinical benefit for misonidazole as a radiosensitizer, probably because the compound exhibits poor concentration in tumors.
Summary of the Invention In a first embodiment of the invention there is provided a radiosensitizer composition comprising an effective amount of 4(S)-nitrohistidine, represented by the following formula (I):

COOH
CH
H2N~ ,N

HN
(I).
The radiosensitizer formula (I) may be racemic 4(5)-nitrohistidine or substantially optically pure 4(5)-vitro-L-histidine, or any pharmaceutically acceptable salt thereof, and 3o may optionally comprise one or more agents. These other agents may be buthionine
2 2967/lOlCA
sulfoximine, which is an inhibitor of glutathione, N-(phosphonacetyl)-z-aspartic acid (PALA), which is an inhibitor of 1,-aspartate transcarbamylase, a chemotherapeutic agent, or any combination thereof.
In some embodiments, the chemotherapeutic agent is a nitrosourea agent, cisplatin, carboplatin (CBDCA), bleomycin, doxorubicin, methotrexate, cyclophosphamide, gemcitabine, treosulfan, 5-fluorouracil, dacarbazine, temozolomide, 9-nitrocamptothecin, vincristine, fotemustine, lomustine, a cytokine, an interferon, or any combination thereof Other embodiments in accordance with the present invention provide a radiosensitizer composition as described above, further comprising a biomodulator to compound. The biomodulator compound may be a controlled-release compound, such as a slow-release compound, and may be a biodegradable polymer. In specific embodiments, the biodegradable polymer is selected from the group consisting of a homopolymer of lactic acid; a homopolymer of glycolic acid; a copolymer of poly-D,1,,-lactic acid and glycolic acid;
a water-insoluble peptide salt of a luteinizing hormone-releasing hormone (LHRI~ analogue;
15 a poly(phosphoester); a bis(p-carboxyphenoxy)propane (CPP) with sebacic acid copolymer;
a polyanhydrides polymer; poly(lactide)-co-glycolide)polyethylene glycol copolymers; and an ethylene-vinyl acetate copolymer.
Another embodiment provides a method of potentiating radiotherapy cancer treatment comprising administering to a patient in need thereof a therapeutically effective 2o amount of a composition comprising a radiosensitizer of the formula:
COOH
H2N~.CH ~N
25 ~ N02 HN
(n and directing radiotherapy at a prescribed dosage to a tumor. Still another 3o embodiment provides a method of potentiating radiotherapy cancer treatment by administering an effective amount of a composition comprising the radiosensitizer of formula (I), or any pharmaceutically acceptable salt thereof, wherein the composition further 2967/lOlCA
comprises one or more agents, and the agents may be buthionine sulfoximine, a nitrosourea agent, N-(phophonylacetyl)-L-aspartic acid (PALA), a chemotherapeutic agent, or any combination thereof. In particular embodiments of the method for potentiating radiotherapy cancer treatment, the chemotherapeutic agent is a nitrosourea agent, cisplatin, carboplatin (CBDCA), bleomycin, doxorubicin, methotrexate, cyclophosphamide, gemcitabine, treosulfan, 5-fluorouracil, dacarbazine, temozolomide, 9-nitrocamptothecin, vincristine, fotemustine, lomustine, a cytokine, an interferon, or any combination thereof.
Alternatively, specific embodiments provide a method of potentiating radiotherapy cancer treatment as described, further comprising administering chemotherapy after directing radiotherapy. In 1 o some embodiments, the method further comprises administering chemotherapy by administering a nitrosourea agent, cisplatin, carboplatin (CBDCA), bleomycin, doxorubicin, methotrexate, cyclophosphamide, gemcitabine, treosulfan, 5-fluorouracil, dacarbazine, temozolomide, 9-nitrocamptothecin, vincristine, fotemustine, lomustine, a cytokine, an interferon, or any combination thereof.
In other specific embodiments, there is provided a method of potentiating radiotherapy cancer treatment by administering an effective amount of a radiosensitizer composition of formula (I) or any pharmaceutically acceptable salt thereof, and directing radiotherapy at a prescribed dosage to a locus of cancer, wherein the method also comprises administering chemotherapy before directing radiotherapy. In some embodiments, 2o administering chemotherapy before directing radiotherapy includes administering a nitrosourea agent, cisplatin, carboplatin (CBDCA), bleomycin, doxorubicin, methotrexate, cyclophosphamide, gemcitabine, treosulfan, 5-fluorouracil, dacarbazine, temozolomide, 9-nitrocamptothecin, vincristine, fotemustine, lomustine, a cytokine, an interferon, or any combination thereof.
Alternative embodiments provide a method of potentiating radiotherapy cancer treatment as described, further comprising, in administering, providing the composition in a slow-release formulation, and alternatively, wherein the slow-release formulation is administered by any suitable means including oral, intravenous, arterial infusion, intraperitoneal, intramuscular, subcutaneous, surgical, and topical.
3o In accordance with particular embodiments of these methods, the slow-release formulation comprises a biodegradable polymer, and the biodegradable polymer is selected from the group consisting of a homopolymer of lactic acid; a homopolymer of glycolic acid;
a copolymer of poly-D,t,,-lactic acid and glycolic acid; a water-insoluble peptide salt of a luteinizing hormone-releasing hormone (LHRII) analogue; a poly(phosphoester);
a bis(p-carboxyphenoxy)propane (CPP) with sebacic acid copolymer; a polyanhydrides polymer;
poly(lactide)-co-glycolide)polyethylene glycol copolymers; and an ethylene-vinyl acetate copolymer.
In some specific embodiments of the method, the slow-release formulation releases the radiosensitizer over a period of four or more weeks, alternatively over a period of one week or more, or alternatively over a period of 24 hours or more. The cancers to be treated to include any of a brain cancer, a lung cancer, a head-and-neck cancer, a GI
cancer, a breast cancer, a prostate cancer, a lymphoma, a sarcoma, a melanoma, a cancer of the cervix or endometrium, a bladder cancer, a renal cancer, a liver cancer, or an ocular cancer. In particular embodiments, the brain cancer is an astrocytoma, and more particularly, is glioblastoma multiforme; the lung cancer is either a small cell lung carcinoma or a non small cell lung carcinoma; and the head-and-neck cancer is squamous cell carcinoma or adenocarcinoma.
More particular embodiments of the method of potentiating radiotherapy cancer treatment further comprise administering daily doses of the radiosensitizer throughout the course of treatment as an alternative to administering the radiosensitizer in a slow-release formulation.
Brief Description of the Drawings The foregoing features of the invention will be more readily understood by reference to the following detailed description, taken with reference to the accompanying drawings, in which:
Fig. 1 shows prior art compounds categorized as nitroimidazole radiosensitizers.
Fig. 2A. is an in vitro study of nitrohistidine on Chinese hamster ovary (CHO) cells.

Fig. 2B is an in vitro study of nitrohistidine compared to misonidazole on CHO
cells.
Fig. 3 is an in vitro study of nitrohistidine in CHO cells after pretreatment with hypoxic conditions.
Fig. 4 is an in vivo Growth Retardation Study of Lewis lung carcinoma in rats.
Fig. 5 is a tumor size assessment plot from the Growth Retardation Study of Fig. 4, showing tumor size as a function of time (in days).
to Fig. 6 shows in vitro synergism of buthionine sulfoximine and nitrohistidine as radiosensitizers in CHO cells.
Detailed Description of Specific Embodiments Definitions. As used in this description and the accompanying claims, the following 15 terms shall have the meanings indicated, unless the context otherwise requires:
"Potentiate" means, in the context of this application to enhance or increase the effect of, for example, a drug, or to promote or strengthen, for example, a biochemical or physiological action or effect.
"Synergism" means, in the context of this application, increased activity with the two 20 or more compounds over what is observed with either compound individually and what would be observed if the activities were merely additive.
"Slow-release compound" means, in the context of this application, a compound that, in conjunction with an active ingredient, releases the active ingredient to the surroundings in a controlled, non-instant manner, time-dependent manner. The slow release of the active 25 agent over time may mean release over several hours, release over several days, or release over several weeks or longer.
"Biomodulator" means, in the context of this application, a compound or agent that is capable of modulating the release of a drug, or the activity of a drug action, by either increasing or decreasing activity of the drug or radiosensitizer, or increasing or decreasing 3o the release of the drug to a desired location.

"Biodegradable polymer" means, in the context of this application, a polymer that can be degraded and metabolized by existing enzymes in the organism to which the biodegradable biopolymer is administered, with little or no side effects or toxicity.
Given the limited ability of ionizing radiation to kill hypoxic cancer cells because of the need for oxygen to "fix" the lesions in the DNA to make them lethal, a radiosensitizer capable of overcoming hypoxic resistance that is well-concentrated by cancer cells, that can be delivered to the tumor cells and released over time, and that has limited to no toxic side effects would be desirable, to augment radiation therapy.
to Although 4(5)-nitrohistidine and substantially pure 4(5)-nitro-I,-histidine were previously synthesized (see Trout and Brossi et al., respectively), the researchers focused on the structural importance of histidine in determining its biological function (i.e., in protein synthesis - Trout) and on the efficacy of nitrohistidine as an anti-protozoal agent relative to the known anti-protozoal nitroimidazole agents, which turned out to be zero (Brossi et al.).
Nitrohistidine has not been explored previously as a radiosensitizer.
Because histidine is an essential amino acid, and taken up in rapidly growing cells, it was hypothesized that nitrohistidine may likewise be taken up by rapidly growing cells, and that it may also act as a radiosensitizer, given its structural similarities to nitroimidazoles.
Therefore, using the synthetic routes previously published by Trout and Brossi et al. (Trout, J
2o Med Chem (1972) 15:1259-1262; and Brossi et al., JMed Chem (1973) 16: 705-707, both of which are incorporated by reference herein), 4(5)-nitrohisitidine and substantially optically pure 4(5)-vitro-L-histidine were synthesized and used in studies developed to assess radiosensitizer capability and efficiency.
The two neutral amino acid transport systems that transport histidine into cells that have been studied in animal model tumor systems such as in Ehrlich ascites cells or S37 cells have points of interaction with amino acids in general that include the amino group, the carboxyl group, and the side chain. The nature of the side chain affects which of the two transport systems a substrate neutral amino acid will utilize, but more variability is allowed at the side chain point of interaction than at the amino and carboxyl points of interaction.
3o The best treatment for patients with refractive tumors such as lung tumors, head-and neck tumors, GI tumors, and inoperable brain tumors like recurrent high-grade gliomas, 2967/lOlCA
including anaplastic astrocytoma and glioblastoma multiforme, often remains open to debate, and has not significantly increased the survival rate of patients with some of the worst tumors for the past 25 years, in spite of advances in surgery techniques, chemotherapy treatments, and radiotherapy and imaging techniques. Radiosensitizers have been utilized to increase the response of patients to radiotherapy, with dubious success reported using halogenated pyrimidine analogs such as bromdeoxyuridine (BrdUrd) and iododeoxyuridine (IdUrd) when treating patients with high-grade gliomas. Somewhat better success has been shown using such radiosensitizers for treatment of GI and head-and-neck tumors, promising greater enhancement with other tumors, given the right conditions.
to In particular, the use of slow-release devices and slow-release or controlled-release formulations for combined delivery of the radiosensitizer with a biodegradable polymer capable of modulating delivery of the radiosensitizer at the locus of the tumor, or able to modulate delivery of a radiosensitizer that concentrates at a tumor site, have begun to be investigated. Such delivery means, coupled with an effective radiosensitizer that concentrates in tumor cells or can be delivered to the tumor location, represents a welcome chance to make significant advances in the war against the most deadly tumors.
Recently, successful reports for treating an intracranial rat glioma model implanted with a radiosensitizer and biomodulator drug coupled with radiotherapy, proved effective at treating such cancers (see Int J Radiat Oncol Biol Phys (2004) 58: 519-527, incorporated by 2o reference herein), statistically increasing the 180-day survival of rats with C6 tumor to 83%
when the radiosensitizer was delivered using a biomodulator intracranial implant.
Astrocytomas are tumors that arise from astrocytes cells, part of the supportive, neuroglial tissue of the brain. Astrocytomas account for about half of all primary tumors of the brain and spinal cord. Glioblastomas are fast growing astrocytomas that contain areas of dead tumor cells (necrosis). Glioblastoma multiforme (GBM), the most common type of grade IV tumor, is also the most malignant and tends to invade adjacent brain tissue and spread throughout the central nervous system. Variants of GBM include gliosarcomas and giant cell glioblastomas.
Glioblastomas occur most often in the cerebrum, especially in the frontal and 3o temporal lobes of the brain and represent about 30% of all primary brain tumors and about 50% of the astrocytomas. Glioblastoma is more common in older adults and affects more 2967/lOICA
men that women. They are very difficult to treat and no significant advancements in their treatment have occurred in the past 25 years. Without therapy, patients with GBM uniformly die within 3 months. Patients treated with optimal therapy - to day this includes surgical resection, radiation therapy, and chemotherapy, have a median survival of approximately one s year.
Uveal melanoma is the most common primary intraocular malignant tumor. Despite the ability to make an accurate diagnosis and availability of various primary treatments, mortality from this disease has remained unchanged for a number of reasons, primarily because of hematogenous metasases, mainly to the liver, which are often highly resistant to to chemotherapy. Moreover, evidence suggests that if local treatment of the uveal melanoma fails, there is an increased incidence of metastases. Mediaa survival times of between S and 7 months are not uncommon after chemotherapy of metastasized uveal melanoma.
Example 1: In Vitro Study of Nitrohistidine as a Radiosensitizer 15 An in vitro study using Chinese hamster ovary (CHO) cells was carried out using F12 medium with fetal calf serum for growth of the CHO cells. A 9 MeV electron beam, generated by a Varian Clinac 1800 instrument, was used to irradiate the Petri dishes, at a dose of 300 cGy, and a concentration of 1 to 4 mM nitrohistidine was added alone, or in combination with the radiation dose. Results of this study are shown in Figure 2A. As can 2o be seen, controls with nitrohistidine alone at all 4 concentrations did not exhibit any toxicity, as seen in the constant levels of cells remaining at all 4 concentrations of nitrohistidine (exhibited along the Y-axis as the S. fraction - SF - the surviving fraction of cells, as a log scale from 10 to 1, with 1 being the number of cells present at time zero). In contrast, in the presence of ionizing radiation (Vitro + XRT) it can be seen that the S.
fraction value 2s decreased from just under 1 to just under 0.6 at all four concentrations tested, indicating that this concentration range is an effective concentration range for radiosensitization, and that nitrohistidine may be a promising radiosensitizer for use for cancer treatment in vivo in conjunction with radiation therapy. Additional studies found that 2 mM
nitrohistidine was nearly as effective as 5 mM (data not shown), and that in vitro efficacy of nitrohistidine as a 3o radiosensitizer was similar to that of misonidazole when both were administered at 2 mM
concentration (See Fig. 2B).

2967/lOlCA
Example 2: In Vitro Study of Nitrohistidine as a Radiosensitizer Under Hypoxic Conditions As in Example 1, CHO cells were grown in F12 medium supplemented with fetal calf serum. However, before ionizing radiation treatment, the CHO cells underwent a 30-minute s pre-incubations in hypoxic conditions, in the presence or absence of 2 mM
nitrohistidine. A
9 MeV electron beam, as in Example 1 above, was used to deliver the radiation doses ranging from 0 to 10 Gy. Figure 3 shows the S. Fraction plotted against radiation dose, in Gy, at 0, 2, 4, 6, 8, and 10 Gy for radiation alone (XRT, indicated with -~-) and radiation plus 2 mM nitrohistidine (XRT + Nitrohistidine, indicated with $-).
l0 As can be seen in Figure 3, the cells exposed to 2 mM nitrohistidine exhibited a radiation dose-dependent depletion of the S. Fraction consistently over that observed with just radiation alone, down to about 0.006 at 10 Gy. The calculated enhancement ratio, at the 0.1 level SF value, equivalent to the killing power of the nitrohistidine plus radiation over treatment with radiation alone (determined using the radiation dosage and radiation plus 1 s nitrohistidine dosages required to effect 90% killing of the tumor cells) was determined to be 1.38.
Example 3: In vitro Syne~ism of Buthionine Sulfoximine and Nitrohistidine Chinese hamster ovary cells were grown on F 12 medium supplemented with fetal 2o calf serum. Cells were treated with dosages of radiation from 0 to 10 Gy, using a 9 MeV
electron beam as above, either alone or in the presence of nitrohistidine, buthionine sulfoximine, or nitrohistidine plus buthionine sulfoximine together. When present, the buthionine sulfoximine and nitrohistidine were added at 2 mM concentration. As can be seen in Figure 4, results with cells treated with dosages of radiation from 0 to 10 Gy alone 2s (XRT, indicated by -~-), or in the presence of nitrohistidine (XRT + NHis, indicated by -f-), buthionine sulfoximine (XRT + BSO, indicated by -~-), or nitrohistidine plus buthionine sulfoximine together (XRT + NHis + BSO, indicated by -e-) show that the radiation plus NHis plus BSO exhibited an enhanced effect relative to radiation alone or with either the NHis or BSO singly. S. fractions remaining for the combined dual drug/radiation treatment 3o indicate an initial lag time, before continued depletion of cells surviving, down to less than 0.01 SF at 10 Gy in the presence of both NHis and BSO. T'he enhancement ratio calculated to for the combined treatment was greater than two over that of any other treatment combination.
Example 4: In Vivo Growth Retardation Experiment 1 With Lewis Lung Carcinoma Groups of 5-6 mice (C-57) were subcutaneously implanted in the hind leg with Lewis lung carcinoma cells and the cells were permitted to grow until a measurable tumor was evident. Radiosensitizers were administered using i.p. injection at 2 mM
concentration, and then treated with a single 500 cGy dose administered with a 9 MeV electron beam and tumor size measured daily for a period of 9 days. Control mice were implanted with tumor cells to and left untreated.
As can be seen in Figure 5, tumor size grew from about 10-12 mm at day 1 to about 23 mm at day 9 for the control group (indicated by = ), to only about 20 mm for the mice treated with radiation along (XRT, indicated by -o-) and to only about 18 mm for mice treated with radiation and NHis (NHis + XRT, indicated by -~-). This indicates a growth 15 retardation of about 22% compared to the control group, and a growth retardation of about 10% compared to the group treated with radiation alone.
Similar results with a second group of mice are shown in Figure 6, but in this group, the growth retardation of NHis + XRT is more almost 35% compared to the XRT
treatment alone (the tumor was reduced from about 23 mm to about 15 mm at day 9) and about 29%
2o compared to the control or NHis alone (the tumor was reduced from about 21 mm to about 15 mm at day 9).
Examine 5: In Yivo Growth Retardation Experiment 2 With Lewis Lung Carcinoma Another growth retardation study was done in mice implanted with Lewis lung 25 carcinoma comparing NHis plus radiation treatment to misonidazole plus radiation treatment.
In this study, misonidazone plus radiation-treated mice lagged in tumor growth initially, but then grew again parallel to the growth rate of the contron mice, indicating that after an initial response, misonidazole plus radiation had no ability to retard tumor growth.
In contrast, mice treated with NHis plus radiation showed a plateau in tumor growth after 9-10 days, 3o indicating that tumor growth had effectively ceased. Evidence of this plateau can also be seen in Figure 5, above.

These results indicate that nitrohistidine plus radiation, in vivo, is a more potent and effective radiosensitizer than misonidazole, presumably due to the concentration of NHis in tumor cells through uptake with the cells' amino acid transport system.
s Example 6: Intracranial Rat Model RadiosensitizerlRadiotherapy Study A. Tissue culture studies Rat glioma cell line C6/LacZ is obtained from American Tissue Culture Collection (ATCC). These cells constitutively express the LacZ reporter gene. Cell lines are maintained in Dulbecco's modified Eagle's medium, supplemented with 15% heat-1o inactivated fetal calf serum and 0.2 mM glutamine, 50 g/mL neomycin, and 100 wg/mL
streptomycin. Irradiation of cells is done in the log phase growth, and the cells are then dispersed by trypsinization, suspended in drug-free medium and irradiated in suspension using approximately a 9 MeV electron beam of X-rays generated by a Varian Clinac 1800 inshvment at a dose rate of up to approximately 2-10 Gy/min, as required.
15 After irradiation, cells at each dose level are plated, plates are fixed and stained at about 7 to 9 days after plating, and colonies of >50 cells are scored.
Survival curves are fitted to the data points using the linear-quadratic model. For measurement of the effect of incorporation of the radiosensitizer, cells in exponential growth are cultured for 72 hr before irradiation in medium adjusted to between about 10'3 tol0'S M with respect to the 2o radiosensitizer. Other drugs, such as buthionine sulfoximine, can also be added to the medium at the same time as the radiosensitizer, at 72 hr before irradiation, to examine any possible synergistic effects.
B. In vitro drug release 2s In vitro drug release is measured as described in published reports, such as Int J
Radiat Oncol Bio Phys ( 1997) 39: 497-504, the contents of which are incorporated by reference herein. Polymer rods, beads, or matrices containing labeled (i.e., radiolabeled, fluorescence-labeled, etc.) or non-labeled radiosensitizer, and optionally additional drug(s), are placed in phosphate-buffered saline at 37°C. Solutions are collected daily, and fresh 3o phosphate-buffered saline is added to the container, as needed. Solutions are analyzed for radiosensitizer by ultraviolet light, or other means, depending on the nature of the 2967/lOlCA
radiosensitizer and whether a label is used to detect the radiosensitizer.
These studies are used to help determine appropriate doses and efficacy of potential radiosensitizers and combinations of radiosensitizers with additional agent(s).
C. Intracranial Implant of C6 Tumors The C6 cell line was developed from a rat glioblastoma induced in randomly bred Wistar rats by N-nitrosomethylurea administration (see JNeurosurg (1971) 34:310-323), and transplantation of cultured C6 cells into the brain of rats by a stereotaxic procedure produces intracerebral tumors representing, as close as possible, the characteristics of spontaneous to gliomas, with concomitant good reproducibility. Cells cultured as described above, in A, are harvested by trypsinization, pelleted by centrifugation, and resuspended for intracranial implant. For tumor implantation, anesthetized rats are placed in a stereotactic head frame and a small frontal craniectomy is drilled about 2.5 mm from the midline and about 1.0 mm anterior to the bregma. Glioma cells (5x105/50 N,L) are implanted stereotactically to a depth 15 of about 4 mm below the craniectomy, using a Hamilton syringe. The craniectomy is resealed with bone wax and the scalp closed.
D. Druglpolymer implants Suitable biodegradable polymer materials include a homopolymer of lactic acid;
a 2o homopolymer of glycolic acid; a copolymer of poly-v,l.,-lactic acid and glycolic acid; a water-insoluble peptide salt of a luteinizing hormone-releasing hormone (LHRH) analogue; a poly(phosphoester); a bis(p-carboxyphenoxy)propane (CPP) with sebacic acid copolymer; a polyanhydrides polymer; poly(lactide)-co-glycolide)polyethylene glycol copolymers; and an ethylene-vinyl acetate copolymer. The biodegradable polymers are synthesized as described 25 in the literature according to published procedures (see, for example, J
Biomed Mat Res (1985)19:941-955 (CPP-sebacic acid copolymer); US Patent No. 6,376,644 (poly(phosphoester)); US Patent No. 6,699,504 (LHRH analogs); US Patent No.
6,201,072 (poly(lactide)co-glycolide)polyethylene glycol copolymers); and US Patent No.
5,792,477 (poly-D,1,; lactic acid and glycolic acid copolymers). Preparations of drug-polymer implants 3o as then prepared, according to established procedures. Briefly, the drug and polymer are mixed, by grinding, or other mechanical means to create a homogeneous mixture, heated to melt the polymer, and extruded to create rods, beads, or other drug-polymer matrix implants, as desired. The drug-polymer implants are then cooled, and stored, dessicated, until needed.
E. Drug Administration The polymer implants, containing 5-25% wt/wt radiosensitizer and optionally, additional agent, are implanted near the tumor through the original hole where the tumor was implanted, using fine forceps to place the polymer/drug implant on top of the site of the tumor implant. Implantation is approximately 8-15 days after tumor implantation, as desired.
Implantation can also be done through injection with a syringe, if the polymer matrix is in to the appropriate form, for example, microbeads.
F. External Beam Radiotherapy Rats are anesthetized and tumors irradiated using approximately a 9 MeV
electron beam of X-rays generated by a Varian Clinac 1800 instrument at a dose rate of up to 15 approximately 2-10 Gy/min, as required. Single-dose hemibrain radiotherapy is done using a 18-mm diameter circular AP field on day 12-15 after tumor implant, as desired, and 2-3 days after implant of the polymer/drug matrix.
G. Tumor Growth Assessment 2o After tumor implant, rats are examined daily for behavioral and neurological signs of tumor growth. Signs include decreased alertness, passivity, poor grooming, irritability, fearfulness, and neurological deficits such as focal motor deficits and gait disturbance.
Animals are killed when 4 of these signs appear, indicating increasing intracranial pressure as the tumor size the behavioral and neurological symptoms worsen. At 14 days, tumor 25 volume will be approximately 20-30 mm3, and at 16-17 days after implant, the earliest at which extraneous signs of tumor growth are typically observed, tumor volume is approximately 30-40 mm3. As the tumor continues to grow, most rats will show symptoms by about 20-23 after implant, and be killed. Rats from all experiments are autopsied at the time of death and the tumor size noted, and the presence of aggressive tumor growth verified.
H. Statistical analysis 2967/lOlCA
Survival can be plotted in various ways, including on a Kaplan-Meier survival curve, allowing survival data to be compared by a log-rank test. Statistical analysis can then be done using any statistical package software.
Example 7: Human Radiosensitizer/Radiotherapy Study in Astroc omas A. Tissue culture studies To screen for effective doses and drug combinations for potentiation of radiotherapy in human cancers, human cancer cell lines of interest, particularly glioblastomas, obtainable l0 from American Tissue Culture Collection (ATCC) are selected. The various cell lines are maintained in Dulbecco's modified Eagle's medium or an equivalent, supplemented with 10-15% heat-inactivated fetal calf serum, as needed, and 0.2 mM glutamine, 50 g/mL neomycin, and 100 wg/mL streptomycin or other antibiotics, as described for culturing the particular cell line of interest. Irradiation of cells is done in the log phase growth, and the cells are then 15 dispersed by trypsinization, suspended in drug-free medium and irradiated in suspension using approximately a 9 MeV electron beam of X-rays generated by a Varian Clinac 1800 instrument at a dose rate of up to approximately 2-10 Gy/min, as required.
After irradiation, cells at each dose level are plated, plates are fixed and stained at about 7 to 9 days after plating, and colonies of >50 cells are scored.
Survival curves are 20 fitted to the data points using the linear-quadratic model. For measurement of the effect of incorporation of the radiosensitizer, cells in exponential growth are cultured for 72 hr before irradiation in medium adjusted to between about 10-3 tol0'S M with respect to the radiosensitizer. Other drugs, such as buthionine sulfoximine, an inhibitor of glutathione and rl-(phosphonacetyl)-1.-aspartic acid (PALA), an inhibitor of L-aspartate transcarbamylase, 25 can also be added to the medium at the same time as the radiosensitizer, at 72 hr before irradiation, to examine any possible synergistic effects.
B. In vitro drug release In vitro drug release is measured as described in published reports, such as Int J
30 Radiat Oncol Bio Phys (1997) 39: 497-504, the contents of which are incorporated by reference herein. Polymer rods, beads, or matrices containing labeled (i.e., radiolabeled, 2967/lOICA
fluorescence-labeled, etc.) or non-labeled radiosensitizer, and optionally additional drug(s), are placed in phosphate-buffered saline at 37°C. Solutions are collected daily, and fresh phosphate-buffered saline is added to the container, as needed. Solutions are analyzed for radiosensitizer by ultraviolet light, or other means, depending on the nature of the radiosensitizer and whether a label is used to detect the radiosensitizer.
These studies are used to help determine appropriate doses and efficacy of potential radiosensitizers and combinations of radiosensitizers with additional agent(s).
C. Druglpolymer implants to One means for administering the radiosensitizer formulations in accordance with the present invention is to use suitable biodegradable polymer materials to prepare an implant to be placed surgically at the site of the tumor. Suitable biodegradable polymer materials include a homopolymer of lactic acid; a homopolymer of glycolic acid; a copolymer of poly-~,1,,-lactic acid and glycolic acid; a water-insoluble peptide salt of a luteinizing hormone-15 releasing hormone (LHRH) analogue; a poly(phosphoester); a bis(p-carboxyphenoxy)propane (CPP) with sebacic acid copolymer; a polyanhydrides polymer;
poly(lactide)-co-glycolide)polyethylene glycol copolymers; and an ethylene-vinyl acetate copolymer. The biodegradable polymers are synthesized as described in the literature according to published procedures (see, for example, JBiomed Mat Res (1985)19:941-955 20 (CPP-sebacic acid copolymer); US Patent No. 6,376,644 (poly(phosphoester));
US Patent No. 6,699,504 (LHRH analogs); US Patent No. 6,201,072 (poly(lactide)co-glycolide)polyethylene glycol copolymers); and US Patent No. 5,792,477 (poly-D,1,,-lactic acid and glycolic acid copolymers). Preparations of drug-polymer implants as then prepared, according to established procedures. Briefly, the drug and polymer are mixed, by grinding, 25 or other mechanical means to create a homogeneous mixture, heated to melt the polymer, and extruded to create rods, beads, or other drug-polymer matrix implants, as desired. The drug-polymer implants are then cooled, and stored, dessicated, until needed.
D. Drug Administration 3o The polymer implants, containing 5-25% wt/wt radiosensitizer and optionally, additional agent, are placed near the tumor, if the tumor is accessible, using endoscope-type injection guns, microinjection, a syringe, or surgical implantation. Delivered dosages range from between about 0.5 mM to about 4 mM daily, whether released from a slow-release polymer matrix or administered daily. Accessibility of the tumor, tumor size and type, and the type of radiosensitizer formulation desired, such as a slow-release formulation that is able to deliver the drug over a period of weeks or a formulation that delivers the drug over a period of days or even hours, will determined the choice of the specific polymer material and the appropriate form of the polymer matrix to be used in administering the radiosensitizer, for example, microbeads, rods, etc. The radiosensitizer, and optionally other agents, are administered in a slow-release formulation or other acceptable formulation by any suitable l0 means including oral, intravenous, arterial infusion, intraperitoneal, intramuscular, subcutaneous, surgical, and topical.
E. External Beam Radiotherapy The subject is anesthetized and tumors irradiated using approximately a 9 MeV
15 electron beam of X-rays generated by a Varian Clinac 1800 instrument at a dose rate of up to approximately 2-10 Gy/min, as required. For brain tumors, single-dose hemibrain radiotherapy is done using a 18-mm diameter circular AP field approximately 1 hour to 2-3 days after implant of the polymer/drug matrix, depending on the release formulation used.
2o F. Tumor GrowthlRetardation Assessment Patients are examined daily for behavioral and neurological signs of tumor regression or growth. Neurological symptoms and signs affecting patients with glioblastomas can be either general or focal, and reflect the location of the tumor. Nonetheless, patients are observed for changes/improvements in the following general symptoms:
headaches, nausea 25 and vomiting, personality changes such as irntability, and malaise, changes in mental capacity and concentration ability, and slowing of cognitive function. Focal signs of the tumor are also observed for change/improvement, including hemiparesis, changes in sensory perceptions and visual ability, and aphasia. Other signs that can be observed are increase/decrease in frequency of seizures. Tumor size is also assessed using standard MRI
3o techniques with and without contrast, and CT scans. On CT scans, glioblastomas usually appear as irregularly shaped hypodense lesions with a peripheral ring-like zone of contrast enhancement and a penumbra of cerebral edema.
MRI with and without contrast shows lesions that are typically enhancing ring observed on Tl-weighted images and a broad surrounding zone of edema apparent on T2-weighted images. The central hypodense core represents necrosis, the contrast-enhancing ring is composed of highly dense neoplastic cells with abnormal vessels permeable to contrast agents, and the peripheral zone of non-enhancing low attenuation is vasogenic edema containing varying numbers of invasive tumor cells. Several pathological studies have clearly shown that the area of enhancement does not represent the outer tumor border to because infiltrating glioma cells can be identified easily within, and occasionally beyond, a 2-cm margin.
Positron emission tomography (PET) scans and magnetic resonance (MR) spectroscopy can be helpful to identify and characterize glioblastomas in difficult cases, such as those associated with radiation necrosis or hemorrhage. On PET scans, increased regional glucose metabolism closely correlates with cellularity and reduced survival.
MR
spectroscopy demonstrates an increase in the choline-to-creatine peak ratio, an increased lactate peak, and decreased N acetylaspartate (NAA) peak in areas with glioblastomas. All of these characteristics can be observed for changes/improvements during and after the course of treatment to assess efficacy and indicate whether changes in dosage and/or 2o administration means are indicated.
G. Statistical analysis Survival and tumor size can be plotted in various ways, including on a Kaplan-Meier survival curve, allowing survival data to be compared by a log-rank test.
Statistical analysis can then be done using any statistical package software.
Example 8: Radiosensitizer Potentiation of Radiotherapy in Humans With Glioblastomas Au~~mented by Chemotherapy_ Methods for potentiating cancer radiotherapy treatment using radiosensitizers can also be done with patients that are, or have been, additionally treated with standard chemotherapy. As described in Example 7 above, patients are administered a radiosensitizer formulation and then treated with radiotherapy and chemotherapy regimen.
Carmustine (bischloroethylnitrosourea - i.e. BCNU) and cis-platinum (cisplatin) are the primary chemotherapeutic agents for treatment of malignant gliomas. Data from the University of California at San Francisco indicate that, for the treatment of glioblastomas, surgery followed by radiation therapy leads to 1-, 3-, and 5-year survival rates of 44%, 6%, and 0%, respectively. By comparison, surgery followed by radiation and chemotherapy using nitrosourea-based regimens resulted in 1-, 3-, and 5-year survival rates of 46%, 18%, and 18%, respectively.
to In this embodiment, chemotherapy agents can be formulated with the radiosensitizer to be administered in a controlled-release formulation over hours, days, or weeks.
Alternatively, the chemotherapy agents may be administered independently of the radiosensitizer using standard chemotherapy formulations and administration routes, such as by i.v. or oral routes.
is Tumor size and response to treatment is assessed as described in Example 7 above.
For tumors other than glioblastomas, other chemotherapeutic agents routinely used for treatment of such cancers, may be formulated for administration in combination with, or independent from, the radiosensitizer.

Claims (38)

What is claimed is:
1. A radiosensitizer composition comprising an effective amount of 4(5)-nitrohistidine, represented by the following formula (I):
or any pharmaceutically acceptable salt thereof.
2. The radiosensitizer composition according to claim 1, wherein the 4(5)-nitrohistidine is non-racemic, substantially optically pure 4(5)-nitro-L-histidine.
3. A radiosensitizer composition according to claim 1, further comprising one or more agents.
4. A radiosensitizer composition according to claim 3, wherein the one or more agents is buthionine sulfoximine, N-(phophonylacetyl)-L-aspartic acid (PALA), a chemotherapeutic agent, or any combination thereof.
5. A radiosensitizer composition according to claim 3, wherein the chemotherapeutic agent is a nitrosourea agent, cisplatin, carboplatin (CBDCA), bleomycin, doxorubicin, methotrexate, cyclophosphamide, gemcitabine, treosulfan, 5-fluorouracil, dacarbazine, temozolomide, 9-nitrocamptothecin, vincristine, fotemustine, lomustine, a cytokine, an interferon, or any combination thereof
6. A radiosensitizer composition according to any of claims 1, further comprising a biomodulator compound.
7. The radiosensitizer composition according to claim 6, wherein the biomodulator compound is a slow-release compound.
8. The radiosensitizer composition according to claim 7, wherein the slow-release compound is a biodegradable polymer.
9. The radiosensitizer composition according to claim 8, wherein the biodegradable polymer is selected from the group consisting of a homopolymer of lactic acid;
a homopolymer of glycolic acid; a copolymer of poly-D,L,-lactic acid and glycolic acid;
a water-insoluble peptide salt of a luteinizing hormone-releasing hormone (LHRH) analogue; a poly(phosphoester); a bis(p-carboxyphenoxy)propane (CPP) with sebacic acid copolymer; a polyanhydrides polymer; poly(lactide)-co-glycolide)polyethylene glycol copolymers; and an ethylene-vinyl acetate copolymer.
10. A radiosensitizer composition comprising an effective amount of 4(5)-nitrohistidine, represented by the following formula (I):
or any pharmaceutically acceptable salt thereof, and a slow-release biodegradable polymer.
11. A radiosensitizer composition according to claim 10, further comprising buthionine sulfoximine, a nitrosourea agent, N-(phophonylacetyl)-L-aspartic acid (PALA), a chemotherapeutic agent, or any combination thereof.
12. A radiosensitizer composition according to claim 11, wherein the chemotherapeutic agent is a nitrosourea agent, cis-platin, carboplatin (CBDCA), bleomycin, doxorubicin, methotrexate, cyclophosphamide, gemcitabine, treosulfan, S-fluorouracil, dacarbazine, temozolomide, 9-nitrocamptothecin, vincristine, fotemustine, lomustine, a cytokine, an interferon, or any combination thereof
13. The radiosensitizer composition according to claim 10, wherein the biodegradable polymer is selected from the group consisting of a homopolymer of lactic acid; a homopolymer of glycolic acid; a copolymer of poly-D,L,-lactic acid and glycolic acid; a water-insoluble peptide salt of a luteinizing hormone-releasing hormone (LHRH) analogue; a poly(phosphoester); a bis(p-carboxyphenoxy)propane (CPP) with sebacic acid copolymer; a polyanhydrides polymer; poly(lactide)-co-glycolide)polyethylene glycol copolymers; and an ethylene-vinyl acetate copolymer.
14. The radiosensitizer composition according to claim 13 wherein the 4(5)-nitrohistidine is non-racemic, substantially optically pure 4(5)-nitro-L-histidine.
15. A method of potentiating radiotherapy cancer treatment comprising:
administering to a patient in need thereof a therapeutically effective amount of a composition comprising a radiosensitizer of the formula:
or any pharmaceutically acceptable salt thereof; and directing radiotherapy at a prescribed dosage to a locus of cancer.
16. A method of potentiating radiotherapy cancer treatment according to claim 15, wherein the composition further comprises one or more agents.
17. A method of potentiating radiotherapy cancer treatment according to claim 16, wherein the one or more agents is buthionine sulfoximine, a nitrosourea agent, N-(phophonylacetyl)-L-aspartic acid (PALA), a chemotherapeutic agent, or any combination thereof.
18. A method of potentiating radiotherapy cancer treatment according to claim 17, wherein the chemotherapeutic agent is a nitrosourea agent, cisplatin, carboplatin (CBDCA), bleomycin, doxorubicin, methotrexate, cyclophosphamide, gemcitabine, treosulfan, 5-fluorouracil, dacarbazine, temozolomide, 9-nitrocamptothecin, vincristine, fotemustine, lomustine, a cytokine, an interferon, or any combination thereof.
19. A method of potentiating radiotherapy cancer treatment according to claim 17, further comprising:
administering chemotherapy after directing radiotherapy.
20. A method of potentiating radiotherapy cancer treatment according to claim 19, wherein administering chemotherapy includes administering a nitrosourea agent, cisplatin, carboplatin (CBDCA), bleomycin, doxorubicin, methotrexate, cyclophosphamide, gemcitabine, treosulfan, 5-fluorouracil, dacarbazine, temozolomide, 9-nitrocamptothecin, vincristine, fotemustine, lomustine, a cytokine, an interferon, or any combination thereof.
21. A method of potentiating radiotherapy cancer treatment according to claim 17, further comprising:
administering chemotherapy before directing radiotherapy.
22. A method of potentiating radiotherapy cancer treatment according to claim 21, wherein administering chemotherapy includes administering a nitrosourea agent, cisplatin, carboplatin (CBDCA), bleomycin, doxorubicin, methotrexate, cyclophosphamide, gemcitabine, treosulfan, 5-fluorouracil, dacarbazine, temozolomide, 9-nitrocamptothecin, vincristine, fotemustine, lomustine, a cytokine, an interferon, or any combination thereof.
23. A method of potentiating radiotherapy cancer treatment according to claim 15, further comprising, in administering, providing the composition in a slow-release formulation.
24. A method of potentiating radiotherapy cancer treatment according to claim 23, further comprising administering the slow-release formulation by any suitable means including oral, intravenous, arterial infusion, intraperitoneal, intramuscular, subcutaneous, surgical, and topical.
25. A method according to claim 23, wherein the slow-release formulation comprises a biodegradable polymer.
26. A method according to claim 25, wherein the biodegradable polymer is selected from the group consisting of a homopolymer of lactic acid; a homopolymer of glycolic acid; a copolymer of poly-D,L,-lactic acid and glycolic acid; a water-insoluble peptide salt of a luteinizing hormone-releasing hormone (LHRH) analogue;
a poly(phosphoester); a bis(p-carboxyphenoxy)propane (CPP) with sebacic acid copolymer; a polyanhydrides polymer; poly(lactide)-co-glycolide)polyethylene glycol copolymers; and an ethylene-vinyl acetate copolymer.
27. A method according to claim 23, wherein the slow-release formulation releases the radiosensitizer over a period of four or more weeks.
28. A method according to claim 23, wherein the slow-release formulation releases the radiosensitizer over a period of one week or more.
29. A method according to claim 23, wherein the slow-release formulation releases the radiosensitizer over a period of 24 hours or more.
30. A method according to claim 23 wherein the cancer is any of a brain cancer, a lung cancer, a head-and-neck cancer, a GI cancer, a breast cancer, a prostate cancer, a lymphoma, a sarcoma, a melanoma, a cancer of the cervix or endometrium, a bladder cancer, a renal cancer, a liver cancer, or an ocular cancer.
31. A method according to claim 30 wherein the brain cancer is an astrocytoma.
32. A method according to claim 31 wherein the astrocytoma is glioblastoma multiforme.
33. A method according to claim 30 wherein the lung cancer is either a small cell lung carcinoma or a non small cell lung carcinoma.
34. A method of potentiating radiotherapy cancer treatment according to claim 15, further comprising, administering daily doses of the radiosensitizer throughout the course of treatment.
35. A method according to claim 34 wherein the cancer is any of a brain cancer, a lung cancer, a head-and-neck cancer, a GI cancer, a breast cancer, a lymphoma, a melanoma, a prostate cancer, a bladder cancer, a kidney cancer, a liver cancer, a sarcoma, or an ocular cancer.
36. A method according to claim 34 wherein the brain cancer is the astrocytoma glioblastoma multiforme, and the method further comprises administering the radiosensitizer formulation daily by any suitable means including oral, intravenous, arterial infusion, intramuscular, intraperitoneal, subcutaneous, surgical, and topical.
37. A method according to claim 34, wherein the head-and-neck cancer is Squamous Cell Carcinoma or Adenocarcinoma.
38. A method according to claim 34, wherein the lung cancer is a small cell lung carcinoma or a non small cell lung carcinoma.
CA 2494209 2005-01-21 2005-01-25 Radiosensitizer formulations and methods for use Abandoned CA2494209A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US64563005P 2005-01-21 2005-01-21
US60/645,630 2005-01-21

Publications (1)

Publication Number Publication Date
CA2494209A1 true CA2494209A1 (en) 2006-07-21

Family

ID=36693835

Family Applications (1)

Application Number Title Priority Date Filing Date
CA 2494209 Abandoned CA2494209A1 (en) 2005-01-21 2005-01-25 Radiosensitizer formulations and methods for use

Country Status (1)

Country Link
CA (1) CA2494209A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113801276A (en) * 2020-06-12 2021-12-17 北京化工大学 Nano-medicine for tumor radiotherapy and chemotherapy synergistic treatment and preparation method and application thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113801276A (en) * 2020-06-12 2021-12-17 北京化工大学 Nano-medicine for tumor radiotherapy and chemotherapy synergistic treatment and preparation method and application thereof

Similar Documents

Publication Publication Date Title
Sipos et al. Optimizing interstitial delivery of BCNU from controlled release polymers for the treatment of brain tumors
RU2270003C2 (en) Paclitaxel-base medicinal agent stabilized with albumin for treatment of solid tumor
Wang et al. Intratumoral delivery of bortezomib: impact on survival in an intracranial glioma tumor model
US11779644B2 (en) Sensitizing cells to proton radiation
US6391911B1 (en) Coadministration of lucanthone and radiation for treatment of cancer
AU768868B2 (en) Synergistic tumorcidal response induced by histamine
JP2015057409A (en) Combinations comprising macitentan for treatment of glioblastoma multiforme
US7608612B2 (en) Radiosensitizer formulations and methods for use
JP2011506467A (en) Method of treatment of melanoma with alpha thymosin peptide combined with antineoplastic heat shock apoptosis activator (HSAA)
WO2007140280A1 (en) Anti-cancer composition and method for using the same
Hasegawa et al. Effect of hydralazine on the blood flow in tumors and normal tissues in rats
Teicher et al. Effect of Fluosol-DA® on the response of intracranial 9L tumors to X rays and BCNU
CA2494209A1 (en) Radiosensitizer formulations and methods for use
Osami et al. Local chemotherapy with slowly-releasing anticancer drug-polymers for malignant brain tumors
CA1330422C (en) Reduction of neurotoxic effects of cytotoxic agents
Wahlberg et al. Polymeric controlled-release amsacrine chemotherapy in an experimental glioma model
US20080038376A1 (en) Anti-cancer composition and method for using the same
US5407925A (en) Regional chemotherapy within the central nervous system with 4-hydroperoxycyclophosphamide
CN110177552A (en) For adjusting the composition of PD-1 signal transduction
CN109674788B (en) Application of carboxyamidotriazole and IDO1 inhibitor combination in resisting tumors
US20050095196A1 (en) Method for using potassium channel agonists for delivering a medicant to an abnormal brain region and/or a malignant tumor
Wu et al. Section on tumors: Young Investigator Award: Local release of carboplatin via an Alzet mini-osmotic pump prolongs survival in a rat brainstem tumor model
Walker et al. Combination and individual antitumor effects of hyperthermia, cisplatin, and selected dithiocarbamates
CN100500220C (en) Anti entity tumour medicinal composition containing blood vessel inhibitor
CA2149731A1 (en) Specialized antibody treatment for brain neoplasm

Legal Events

Date Code Title Description
EEER Examination request
FZDE Dead

Effective date: 20121116