[go: up one dir, main page]

WO2007060012A2 - Use of l-phenylalanine conjugated to an emitting isotope for therapy of hormone dependent carcinoma - Google Patents

Use of l-phenylalanine conjugated to an emitting isotope for therapy of hormone dependent carcinoma Download PDF

Info

Publication number
WO2007060012A2
WO2007060012A2 PCT/EP2006/011368 EP2006011368W WO2007060012A2 WO 2007060012 A2 WO2007060012 A2 WO 2007060012A2 EP 2006011368 W EP2006011368 W EP 2006011368W WO 2007060012 A2 WO2007060012 A2 WO 2007060012A2
Authority
WO
WIPO (PCT)
Prior art keywords
phenylalanine
carcinoma
conjugated
therapy
iodine
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.)
Ceased
Application number
PCT/EP2006/011368
Other languages
French (fr)
Other versions
WO2007060012A3 (en
Inventor
Samuel Samnick
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 WO2007060012A2 publication Critical patent/WO2007060012A2/en
Publication of WO2007060012A3 publication Critical patent/WO2007060012A3/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/088Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins conjugates with carriers being peptides, polyamino acids or proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma
  • the present invention provides a use of a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine- 211 for the preparation of a pharmaceutical composition for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma. Furthermore, the invention provides a method for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma and a method for the monitoring of the progress of such treatment.
  • PC prostate cancer
  • androgen- deprivation therapies radiotherapy and chemotherapy [6 - 8].
  • these therapies are effective for patients with localized and androgen-responsive PCs, they have demonstrated limited curative effect against advanced metastatic and androgen-resistant states of PCs, and no demonstrable survival benefit has been demonstrated in randomized studies [8, 9].
  • Various new treatment modalities are currently being developed, including second-line hormonal therapy, immunotherapy, radical prostectomy, gene therapy, dendritic cell vaccination therapy, antisense therapy, growth factor inhibition, vitamin D, and biphosphonates.
  • second-line hormonal therapy including second-line hormonal therapy, immunotherapy, radical prostectomy, gene therapy, dendritic cell vaccination therapy, antisense therapy, growth factor inhibition, vitamin D, and biphosphonates.
  • Cytotoxic chemotherapy using anthracyclines or melphalan as well as newer drugs such like paclitaxel or docetaxel has been shown to be of clinical benefit [13].
  • pilot studies using these chemotherapy regimens to treat advanced stage cancers revealed high rates of congestive cardiac failure [18 % to 20 %], and toxic effects on bone marrow and other organs, limiting clinical application [14].
  • none of the treatments currently used has shown a significant survival benefit in patients with metastatic, hormone- refractory prostate cancer or metastatic breast cancer as yet. The median survival time for these patients remains in the range of 1 to 3 years [9, 12].
  • a cytostatically active drug with a favorable tolerability profile, overcoming cellular detoxification strategies, and ideally conveying additional anti-tumor activity, therefore, would be desirable as an alternative for maintenance or induction therapy in tumor, to improve tolerability and efficacy of existing chemo- and radiotherapy regimens.
  • the technical problem underlying the present invention is to provide means and methods for an improved treatment of hormone dependent carcinoma and hormone- refractory and/or metastatic disease.
  • the solution to this technical problem is achieved by the embodiments characterized in the claims.
  • the present invention relates to the use of a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 for the preparation of a pharmaceutical composition for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma, wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma.
  • hormone dependent carcinoma describes in the context of the present invention any human cancer which is histologically derived from tissues of the primary or secondary male or female sexual organs, and the functional regulation of which is controlled primarily by sexual hormones of the steroid class, typically androgens (e.g. testosterone) or gestagens (e.g. progesterone) or estrogens (e.g .estradiol).
  • sexual hormones of the steroid class typically androgens (e.g. testosterone) or gestagens (e.g. progesterone) or estrogens (e.g .estradiol).
  • hormone availability e.g. tamoxifen, an anti-estrogen.
  • anti-hormon agents e.g. tamoxifen, an anti-estrogen
  • most tumors later progress to more malignant hormone-independent and/or metastatic forms. These forms may be resistant to many current therapies.
  • carcinoma derived from hormone dependent carcinoma defines according to the invention such malignant hormone-independent and/or metastatic forms.
  • the carcinoma to be treated by the use or method of the invention are selected from the group consisting of prostate carcinoma and mamma carcinoma.
  • prostate carcinoma describes a type of hormone dependent cancer of the human prostate gland which is initially characterized by androgen-dependence, and which does typically progress to an androgen-resistant state. Such a carcinoma may demonstrate resistance to many chemotherapeutic agents that are diverse both in structure and mechanism of action.
  • mamma carcinoma is understood in the context of the invention as a synonym for "breast cancer”.
  • breast cancer describes a further type of hormone dependent cancer of the human mammary glands which is also characterized by progress to therapy resistant metastatic state.
  • An indication to treat a subject according to the invention can be derived by the diagnosis of minimal residual disease, preferably early solid tumor, advanced solid tumor or metastatic solid tumor, which is characterized by the local and non-local recurrence of the tumor caused by the survival of single cells.
  • a L-phenylalanine is conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine- 211.
  • Exemplary for the compounds of the invention and in particular as a proof of principle for IPA-131 , orphan status was granted by the EMEA on April 11 , 2006 under the EU designation number EU/3/06/363.
  • alpha-, beta- or Auger-electron emitting isotope defines in the context of the present invention radioactive istopes, characterised by the emission of different particles (rays) formed during radioactive decay or by nuclear transition processes.
  • An alpha emitting isotope is defined as a radioactive nuclide emitting alpha particles, corresponding to a helium nucleus consisting of two protons and two neutrons.
  • a beta emitting isotope is defined as a nuclide emitting fast nuclear electrons (negatrons) formed during radioactive decay.
  • An Auger-electron emitting isotope is defined as a nuclide emitting low energy nuclear electrons, formed by nuclear electron capture or internal transition processes. The maximum path lengths of these particles are in a range from 10 nm to 12 mm.
  • X is bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 or astatine-211 linked to L-phenylalanine at the 3- (meta-) or 4- (para-) position within the aromatic ring.
  • Ri is H, alkyl group, amino acid, peptide, protein or an other residue known to facilitate or improve tumor targeting.
  • R 2 is OH, amino acid, or an other residue known to facilitate or improve tumor targeting.
  • Preferred conjugates according to the formula I are those in which X is a bromine-77, bromine-82, iodine-125, iodine-131 , or astatine-211 linked to L-phenylalanine at the para- position of the aryl group, while Ri is H and R 2 is OH.
  • the conjugates According to the physical half life of the radionuclide conjugated to the L- phenylalanine, also the conjugates have a corresponding half life of 16.2 h for bromine-76, 57.04 h for bromine-77, 35.3 h for bromine-82, 4.17 d for iodine-124, 59.41 d for iodine-125, 8.02 d for iodine-131 or 7.21 h astatine-211 labelled L- phenylalanine, respectively.
  • the halogen isotope may e.g.
  • n.c.a. non-carrier- added conjugation
  • c.a. carrier-added conjugation
  • an endoradiotherapeutic agent defines in the context of the present invention an agent which comprises at least one type of radioactive isotopes.
  • Such agent is to be administered to a subject in the need thereof and is effective in the therapy of the above described carcinoma with the ability to metastasize due to an endogenic irradiation, i.e. an irradiation with the radioactive compound within the body of the subject to be treated by the endoradiotherapy.
  • the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope may be administered to the subject in the need thereof via a parenteral, transdermal, intraluminal, intra-arterial, intrathecal or intravenous route or by direct injection into malignant tissue. Also within the scope of the invention is an administration wherein the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is bound to a matrix of a medical device.
  • the conjugate may be bound, for example, to a suitable matrix via an amino binding between the amino group of the phenylalanine and a matrix coated with polypeptides.
  • the medical device may be e.g. a wound plating such as a gauze, a metal plating or different, preferably biological, carrier material.
  • the effective compound is formulated in form of a pharmaceutical composition to the subject in the need thereof.
  • the term "pharmaceutical composition” relates to a composition for administration to a subject, preferably a human patient.
  • the pharmaceutical composition is preferably administered parenterally, transdermal ⁇ , intraluminally, intra-arterially, intrathecally or intravenously.
  • a direct injection of the pharmaceutical composition into malignant tissue is also preferred.
  • said pharmaceutical composition is administered to a patient via infusion or injection.
  • Administration of the suitable compositions may be effected by different ways, e.g., by intravenous, subcutaneous, intraperitoneal, intramuscular, topical or intradermal administration.
  • the pharmaceutical composition may further comprise a pharmaceutically acceptable carrier.
  • suitable pharmaceutical carriers include phosphate buffered saline solutions, water, emulsions, such as oil/water emulsions, various types of wetting agents, sterile solutions, etc.
  • Compositions comprising such carriers can be formulated by well known conventional methods. These pharmaceutical compositions can be administered to the subject at a suitable dose. The dosage regimen will be determined by the attending physician and clinical factors. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently.
  • compositions may be administered locally or systematically. Administration will generally be parenteral, e.g., intravenous. In an preferred embodiment, the pharmaceutical composition is administered subcutaneously and in an even more preferred embodiment intravenously. Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like.
  • the pharmaceutical composition might comprise proteinaceous carriers, like, e.g., serum albumine or immunoglobuline, preferably of human origin. It is envisaged that the pharmaceutical composition might comprise, in addition to L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope, further biologically active agents, depending on the intended use of the pharmaceutical composition in a treatment comprising the administration of additional agents for a concomitant therapy. Examples for such further biologically active agents are described herein below in the context of uses and methods comprising a concomitant therapy.
  • the L-phenyalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is to be administered in doses of 10 "5 to 10 ⁇ 18 g / kg body weight. More preferably, the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is administered in doses of 10 "7 to 10 ⁇ 15 g / kg body weight and more preferably, in doses of 10 "8 to 10 "10 g / kg body weight. It is preferred that such a dose is formulated contained in 1 to 10, preferably 2 to 5 ml of sterile solution, such as phosphate buffered saline solutions, water for injection, etc.
  • sterile solution such as phosphate buffered saline solutions, water for injection, etc.
  • the above described conjugates are capable to accumulate specifically and with marked retention in a limited group of hormone dependent carcinoma and hormone-refractory or metastasized carcinomas derived from hormone dependent carcinomas.
  • the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope specifically accumulates in prostate carcinoma cells, whereas other carcinoma with the ability to metastasize do not or do not specifically accumulate the conjugate.
  • carcinomas respectively for metastasized carcinomas which do not or do not specifically accumulate the conjugate are known in the art and include lung cancer, pancreatic cancer, colorectal cancer, metastases of malignant melanoma, myeloma, lymphoma and metastases of cancers with unknown primary localization.
  • this conjugate is useful in the treatment of such specific hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma by specifically targeting the cells of this carcinoma and to destroy it by the irradiation activity of the emitting isotope.
  • SPET single photon emission tomography
  • IPA-123 4- [ 123 l]iodo-L-phenylalanine
  • hormone dependent carcinomas are characterized by a progress to a hormone-independent and therapy resistant metastatic state, which has been associated with the activation of other signaling cascades by growth factors that control the strict balance between the growth rate and apoptosis [7, 22].
  • EGF and transforming growth factor alpha play a major role during carcinogenesis.
  • TGF- ⁇ insulin-like growth factor-l
  • IGF-I insulin-like growth factor-l 1 protein Kinase A cascades and members of the erbB family is understood to lead to recruitment of proteins that interact and activate androgen or estrogen receptors in the absence of hormones [7, 22, 23].
  • the expression levels of EGFR and its ligands EGF and TGF- ⁇ in prostate carcinoma cells is generally enhanced during the disease progression to more malignant hormone-independent and metastatic PC forms, where it regulates angiogenesis, tumor growth and progression, and metastasis [7, 22].
  • IPA-123 was previously known only as moderate in hormone dependent PC cells and in brain metastases of small cell lung cancers and adenocarcinomas [17]
  • the surprising feature of the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is the unexpected high accumulation in androgen-independent PC3 and DU 145 PC cells, and in metastatic breast tumor cells.
  • the invention teaches that the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope specifically accumulates in hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma as well as such metastasized carcinoma which migrated into the brain.
  • the conjugated L-phenylalanine is to be administered intravenously.
  • the intravenous administration may be effected by administering the conjugate in form of an above described pharmaceutical composition for parenteral administration.
  • the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L-phenylalanine is in the range of 0.1 to 1000 MBq/kg body weight. Generally, but not exclusively, this dose range overlaps with the above indicated dose range calculated in terms of g / kg body weight. More preferably, the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope is in the range of 10 to 400 MBq/kg body weight, more preferably the dose is in the range of 20 to 120 MBq/kg body weight.
  • the administered dose can be determined using an appropriate dose meter, calibrated to quantitatively measure alpha, beta or gamma radiation.
  • the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L-phenylalanine is to be administered as a single dose once or as fractionated doses in 2 to 60 fraction doses. More preferably, the conjugate is administered fractionized in 2 to 10 fraction doses.
  • Dose fractionation is an established procedure in radiation therapy. By fractionating a total administered dose, improved tolerability for healthy non-target tissue, as well as an increased cytotoxic effect to tumor tissue is achieved. Repeated fractionated irradiation allows to therapeutically impact a higher percentage of cells in radiation sensitive stages of the cell cycle, compared to a one time single high dose irradiation.
  • Therapeutic irradiation induces single and double strand breaks of DNA, which is counteracted by nuclear repair mechanisms upregulated following irradiation. It is believed that cells undergoing DNA repair, are more susceptible to a renewed irradiation than radiation- naive cells.
  • the conjugated L- phenylalanine is 4-[ 131 l]iodo-L-phenylalanine (IPA-131), 4-[ 124 l]iodo-L-phenylalanine (IPA-124) and 4-[ 211 At]astatine-L-phenylalanine (AtPA-211).
  • lodine-131 is widely available, has a favourable half life and can be handled by most institutions licensed to apply open radionuclides, lodine-131 allows for convenient extracorporal therapy monitoring using a gamma camera owing to a gamma ray component, emitted in a fixed ratio relative to the therapeutic beta particle emission, which is itself not detectable extracorporally.
  • lodine-124 has a positron emission component, allowing for PET imaging, in addition to the therapeutic beta- emission.
  • internal dosimetry measurements at an ongoing basis can be conducted for therapy planning and therapy monitoring for a period of up to 15 days following a single injection.
  • Astatine-211 is also preferred, as it emits high energy (6.8 MeV) alpha particles, with a short path length in tissue (65 ⁇ m), allowing to administer a highly cytotoxic radiation to targeted tissue, while minimising undesirable radiation effects to non-target tissue.
  • the pharmaceutical composition further comprises a chemo therapeutic agent, an immunotherapeutic agent, a gene therapeutic agent, a vaccine, an antisense nucleotide therapeutic agent, an siRNA therapeutic agent, a further endoradiotherapeutic agent and/or a further radiosensitising agent.
  • a chemo therapeutic agent, an immunotherapeutic agent, a gene therapeutic agent, a vaccine, an antisense nucleotide therapeutic agent, an siRNA therapeutic agent, a further endoradiotherapeutic agent and/or a further radiosensitising agent is understood as a concomitant therapy. Methods and means for concomitant therapies are well known in the art.
  • the conjugated L-phenylalanine and the additional therapeutic may be formulated as a single pharmaceutical composition for simultaneous administration of the effective compounds or in separate pharmaceutical composition for sequential administration. Accordingly, an administration of a composition comprising the conjugated L- phenylalanine prior to the administration of a composition comprising one or more therapeutics selected from the group of a chemo therapeutic, an immunotherapeutic, a gene therapeutic, a vaccine, an antisense nucleotide therapeutic, an siRNA therapeutic, a further endoradiotherapeutic agent and/or a further radiosensitising agent is envisaged as well as simultaneous or subsequent administration.
  • chemotherapeut agent comprises bioactive agents known to be effective in retarding or arresting the malignant growth or to be effective in the regression or elimination of malignant tissues or cells.
  • agents might be e.g. drugs acting as cytostatics.
  • a chemotherapy comprises in line with the medical standards in any systemic or local treatment the administration of cytostatic or cytotoxic agents.
  • Chemotherapeutic agents used in oncology include among others, nitroso urea compunds (ACNU [nimustin], BCNU [carmustin], CCNU [lomustin]), temozolomid, procarbacin, metothrexate, cytarabin, gemcitabine, fluorouracil, cyclophosphamide, mitoxantron, anthracyclins, estramustin, or taxanes.
  • the chemotherapeutic agents are intended to be administered in appropriate dosing regimens according to medical practise.
  • nitroso urea compounds, temozolomide, procarbacin, and methotrexate are preferred chemotherpeutic agents.
  • an immunotherapeutic agent comprise but are not limited to compounds such as antibodies, antibody fragments and/or derivatives thereof which specifically detect malignant tissue and/or cellular therapeutics, including those consisting of adoptively transferred autologous, heterologous, xenogenous or endogenous cells, which have the ability to eliminate malignant cells or tissues.
  • antibody fragment or derivative thereof relates to single chain antibodies, or fragments thereof, synthetic antibodies, antibody fragments, such as Fab, a F(ab2)', Fv or scFv fragments, single domain antibodies etc., or a chemically modified derivative of any of these.
  • Antibodies to be employed in accordance with the invention or their corresponding immunoglobulin chain(s) can be further modified outside the motifs using conventional techniques known in the art, for example, by using amino acid deletion(s), insertion(s), substitution(s), addition(s), and/or recombination(s) and/or any other modification(s) (e.g. posttranslational and chemical modifications, such as glycosylation and phosphorylation) known in the art either alone or in combination.
  • modification(s) e.g. posttranslational and chemical modifications, such as glycosylation and phosphorylation
  • a tumor-specific marker is a tumor-associated cell surface antigen which is either found exclusively on tumor cells or is overexpressed on tumor cells as compared to non- malignant cells.
  • Tumor-associated cell surface antigens can be expressed not only on tumor cells but also on cells/tissue which are/is not essential for survival or which can be replenished by stem cells not expressing tumor-associated cell surface antigen. Furthermore, a tumor-associated cell surface antigen can be expressed on malignant cells and non-malignant cells but is better accessible by a therapeutic agent of interest on malignant cells. Examples of over-expressed tumor-associated cell surface antigens are Her2/neu, EGF-Receptor, Her-3 and Her-4. An example of a tumor- associated cell surface antigen which is tumor specific is EGFRV-III. An example of a tumor-associated cell surface antigen which is presented on a cell which is non- essential for survival is PSMA.
  • tumor-associated cell surface antigens which are presented on cells which are replenished are CD19, CD20 and CD33.
  • immunotherapeutics may comprise agents such as T-cell co-stimulatory molecules or cytokines, agents activating B-cells, NK-cells or other cells of the immune system as well as drugs inhibiting immune reactions (e.g. corticosteroids).
  • gene therapeutic agent defines in the context of the invention means for a therapy comprising the administration of one or more nucleic acid constructs functionally encoding e.g. one or more antigens which are characteristic for malignant cells. Such antigens comprise tumor specific markers. The sequence encoding such antigen is operably linked to a nucleic acid sequence which is a regulatory sequence.
  • a gene therapy comprises the functional expression of a heterologous gene in a patient according to standard medical protocols using appropriate vector systems known in the art; see e.g. Haberkorn et al., Curr Med Chem. 2005; 12(7)779-94.
  • regulatory sequence refers to DNA sequences which are necessary to effect the expression of coding sequences to which they are ligated.
  • Control sequences in the context of the described gene therapy generally include promoters, terminators and, in some instances, enhancers, transactivators or transcription factors.
  • control sequence is intended to include, at a minimum, all components the presence of which are necessary for expression, and may also include additional advantageous components.
  • operably linked refers to a arrangement/configuration wherein the components so described are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • the administration of a vaccine aims in the context of the present invention at activating the innate or adaptive immune system of the patient to act against the tumor tissue or the malignant cells.
  • Such therapy comprises e.g. administering one or more antigen preparations containing tumor substances, or cells selected to react against tumor tissue or the malignant cells.
  • An antisense therapeutic agent is e.g. a nucleotide sequence being complementary to tumor-specific gene sequences, aiming at functionally neutralising tumor gene expression, and consequently inducing tumor cell death.
  • An siRNA therapeutic agent according to the invention is e.g. a small interfering RNA capable of sequence-specifically silencing the expression and activity of various tumor-specific target genes by triggering cleavage of specific unique sequences in the mRNA transcript of the target gene and disrupting translation of the target mRNA, consequently inducing tumor cell death.
  • a concomitant therapy which requires the administration of additional bioactive agents which are effective in the treatment of the carcinoma with the ability to metastasize may be accompanied by the administration of additional compounds which minimize potential side effects of said bioactive agents such as drugs acting on the gastro- intestinal system, drugs preventing hyperuricemia, and/or drugs acting on the circulatory system, e.g. on the blood pressure, known in the art.
  • additional bioactive agents may be formulated in the form of the same or a separate pharmaceutical composition.
  • radiosensitizing agents are 3-iodo-L-phenylalanine or 4-iodo- phenylalanine, which are characterized by a stable, non-radioactive iodine of the [ 127 I]- isotope.
  • radiation therapy includes external and internal radiation therapies.
  • the pharmaceutical composition is to be administered to a patient who is subsequently to be irradiated percutaneously (percutaneous radiotherapy).
  • percutaneous radiotherapy is understood in the context of the invention as a concomitant therapy.
  • Percutaneous radiotherapy is typically administered as an external beam radiation stemming from among others, radioactive cobalt-60 sources, linear accelerators, proton, neutron, or hadron beam sources.
  • the irradiation is started in a period of 0 to 7 days subsequent to the administration of the conjugated L- phenylalanine. More preferably, the irradiation is started in a period of 0.5 to 24 hours subsequent to the administration of the conjugated L-phenylalanine.
  • the concomitant radiotherapy may comprise a cumulative external irradiation of a patient in a dose of 1 to 100 Gy.
  • a preferred range of the irradiation dose is 1 to 60 Gy.
  • the external irradiation dose is administered in 1 to 60 fractional doses, more preferably in 5 to 30 fractional doses.
  • the fractionized doses are administered over a period of 1 to 26 weeks, more preferably over a period of 6 to 12 weeks.
  • the invention provides a method for the monitoring of the progress of a treatment of carcinoma with the ability to metastasize (in a human subject) selected from the group consisting of prostate carcinoma and mamma carcinoma (breast cancer), the method comprising the step of localizing and/or dosimetrically measuring a L-phenylalanine conjugated to an alpha-, beta- or Auger- electron emitting isotope selected from the group consisting of bromine-76, bromine- 77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 in the subject by using a ⁇ -camera subsequent to the administration of a pharmaceutical composition comprising said conjugate to a subject in the need thereof.
  • the detection of an interception of tumor progression, a decrease of the tumor size or an elimination of a tumor is indicative of a therapeutic success.
  • conjugated L-phenylalanine in the subject by using a ⁇ -camera is exemplified e.g. in [16, 17]. It is preferred that the conjugated L-phenylalanine is localized and/or dosimetrically measured at least 0 to 7 days, preferably 0.5 to 48 h subsequent to the administration of the pharmaceutical composition.
  • the invention provides a method for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma, wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma, the method comprising the steps of administering a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 to a subject in the need thereof. It is preferred that the effective compound, which is the conjugated L-phenylalanine, is formulated in form of a pharmaceutical composition.
  • composition has been defined herein above.
  • the route of administration of the effective compound depends inter alia from its formulation. Different routes for differentially formulated compositions have been described herein above. It is preferred for the method of the invention that the conjugated L-phenylalanine is administered intravenously.
  • the conjugated L-phenylalanine labeled with alpha-, beta- or Auger-electron emitting isotope is administered to the subject in a doses of 10 "5 to 10 " 18 g / kg body weight. More preferably, the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is administered in doses of 10 "7 to 10 "15 g / kg body weight of the subject and more preferably in doses of 10 "8 to 10 "10 g / kg body weight of the subject.
  • the radioactive dose of the conjugated L- phenylalanine labeled with alpha-, beta- or Auger-electron emitting isotope is in the range of 0.1 to 1000 MBq/kg body weight. Generally, but not exclusively, this dose range overlaps with the above indicated dose range calculated in terms of g / kg body weight. More preferably, the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope is in the range of 10 to 400 MBq/kg body weight and more preferably in the range of 20 to 120MBq/kg body weight. As described herein above, it is also preferred that such dose is formulated contained in 1 to 10, preferably 2 to 5 ml of sterile solution, such as phosphate buffered saline solutions, water for injection, etc.
  • sterile solution such as phosphate buffered saline solutions, water for injection, etc.
  • conjugated L-phenylalanine is 4 -[ 131 l]iodo-L- phenylalanine (IPA-131), 4-[ 124 l]iodo-L-phenylalanine (IPA-124) or 4-[ 211 At]astatine-L- phenylalanine (AtPA-211).
  • the method further comprises the step of a treatment of the subject by a concomitant therapy.
  • Said concomitant therapy may be selected from the group consisting of a surgical therapy, a chemotherapy, an endo- or exoradiotherapy, an immunotherapy, a gene therapy, a vaccine therapy, an antisense nucleotide therapy, an siRNA therapy, an intracavitary therapy, a radiosensitizer therapy, or a device-based treatment.
  • the step of the concomitant therapy may be effected prior, simultaneous or subsequent to the step of administering a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope.
  • a chemotherapy, an endo- or exoradiotherapy, an immunotherapy, a gene therapy, a vaccine therapy, an antisense nucleotide therapy, an siRNA therapy and radiosensitizer therapy have been provided herein above.
  • Methods and means for concomitant therapies are well known in the art.
  • An example for a surgical therapy comprises a resection of a solid tumour or of malignant tissue.
  • a further concomitant therapy in line with the invention comprises the surgical implantation of a radioactive device such as a radioactive seed. Such seed may be implanted locally to the tumor site.
  • a radioactive device such as a radioactive seed.
  • Such seed may be implanted locally to the tumor site.
  • the technique of implanting radioactive devices is known in the art and described herein above in the discussion of the state of the art.
  • the concomitant therapy is an exoradiotherapy (also called external beam radiation therapy) and wherein the irradiation is a percutaneous radiotherapy.
  • the irradiation is started in a period of 0 to 7 days subsequent to the administration of the conjugated L- phenylalanine. More preferably, the irradiation is started in a period of 0.5 to 24 hours subsequent to the administration of the conjugated L-phenylalanine.
  • the above described concomitant exoradiotherapy may comprise a cumulative external irradiation of a patient in a dose of 1 to 100 Gy.
  • a preferred range of the irradiation dose is 1 to 60 Gy.
  • the external irradiation dose is administered in 1 to 60 fractional doses, more preferably in 5 to 30 fractional doses.
  • the fractionized doses are administered over a period of 1 to 26 weeks, more preferably over a period of 6 to 12 weeks.
  • external field radiation therapy is typically administered as an external beam radiation stemming from among others, radioactive cobalt-60 sources, linear accelerators, proton, neutron, or hadron beam sources.
  • the term 'fractional dose' is to be understood to mean that the overall activity of the fractional dose adds up or essentially adds up to the cumulative external irradiation otherwise also achievable by administering one single dose.
  • the subject to be treated by all embodiments of the method of the invention is a human subject.
  • the invention relates to a method for the monitoring of the progress of a treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma (in a human subject), wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma, the method comprising the steps: (a) administering a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 to a subject in the need thereof; and (b) localizing and/or dosimethcally measuring the conjugated L-phenylalanine in the subject by using a ⁇ -camera.
  • a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-
  • the detection of an interception of tumor progression, a decrease of the tumor size or an elimination of a tumor is indicative of a therapeutic success.
  • a method for the administration of a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is described herein above.
  • an example for the localization and/or dosimetric measurement of conjugated L-phenylalanine in the subject by using a ⁇ -camera is exemplified e.g. in [16, 17].
  • the method for the monitoring of the progress of a treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma may also comprise the step of a treatment of the subject by an above described concomitant therapy.
  • the step of the concomitant therapy may be effected prior, simultaneous or subsequent to the step of administering a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope.
  • the conjugated L-phenylalanine is localized and/or dosimetrically measured at least 0 to 7 days, more preferably 0.5 to 48 h subsequent to its administration.
  • IPA-123 Cellular uptake kinetics of IPA (IPA-123) in hormone independent human prostate cancer cell lines in vitro
  • Example 1 4-Bromo-L-phenylalanine (4-BrPA), 3-bromo-L-phenylalanine (3-BrPA), 4-iodo-L- phenylalanine (4-IPA), 4-ter.butyltinn-L-phenylalanine (4-TBSnPA), 3-ter.butyltinn-L- phenylalanine (3-TBSnPA), 4-methylsilyl-L-phenylalanine (4-Me 3 SiPA) and 3- methylsilyl-L-phenylalanine (3-Me 3 SiPA) used as starting materials (precursor) for radiolabeling were either purchased commercially or prior synthesized in analogy to the literature.
  • HPLC purification was performed on a Hewlett Packard HPLC system consisting of a binary gradient pump (HP 1100), a Valco 6-port valve with 2500 ⁇ l loop, a variable wavelength detector (HP 1100) with a UV detection at 254 nm and a sodium iodide scintillation detector (Berthold, Wildbad, Germany), using reversed-phased column (250 x 4 mm, Nucleosil-100). The column was eluted at different flow rates in with water/ethanol/acetic acid (89:10:1 ; v/v) or PBS / ethanol (90:10; v/v).
  • the proposed radiolabeled phenylalanines were obtained either by non-isotopic halogen exchange (carrier-added/c.a.) or by radio-demetalation of the corresponding precursor as described in the general scheme 1 , resulting to no-carrier-added (n. c. a) products after HPLC separation.
  • X m- , p-Br or m-, p-l for 77/82Br n.c.a. (m, p)-IPA-124, -IPA-125, -IPA-131 or n.c.a. (m, p)-BrPA-77 and -BrPA-82
  • Scheme 1 scheme of the radiosyntheses of n.c.a. IPA-124, IPA-125, IPA-131 , BrPA- 77, BrPA-82 and AtPA-211
  • Example 2 phenylalanine (m,p-IPA-125) and 3,4-r 131 lliodo-L-phenylalanine (m,p-IPA-131) by non- isotopic radioiodo-debromination
  • a solution of carrier free sodium [ 124 l]iodide, sodium [ 125 l]iodide or sodium [ 131 l]iodide (up to 5 GBq) and 5 ⁇ l aqueous Na 2 S 2 O 5 (4.0 mg Na 2 S 2 O 5 ZmI) was evaporated to dryness by passing a stream of nitrogen through a reaction vessel at 100 0 C, followed by addition of 200 ⁇ l of the corresponding L-bromophenylalanine (0.25 - 0.5 mg/ ml 0.1 N H 3 PO 4 ), 20 ⁇ l aqueous L-ascorbic acid (10 mg/ml) and 20 ⁇ l aqueous Cu(II) sulphate (0.10 mol/l).
  • the reaction vessel was heated for 30 min at 170 0 C, cooled and the mixture diluted with up to 500 ⁇ l water.
  • the radioiodinated product was separated from unreacted starting materials and radioactive impurities by HPLC.
  • 3/4- IPA-124, 3/4-IPA-125 and m/p-IPA-131 were obtained in 88 ⁇ 10% radiochemical yield, with a specific activity > 500 GBq / ⁇ mol.
  • the fraction containing the radioiodinated products was collected into a sterile tube, buffered with 0.5 M phosphate buffered saline (pH 7.0; Braun, Melsungen, Germany), and sterile filtered through a 0.22 ⁇ m sterile membrane (Millex GS, Millipore, Molsheim, France) to an isotonic and injectable radiopharmaceutical for in vitro and in vivo investigations.
  • Example 4 Synthesis of 3.4-r 82 Br1bromo-L-phenylalanine (m. p-BrPA-82) and 3.4-f 77 Br1bromo-L- phenylalanine (m,p-BrPA-77)
  • (m, p)-BrPA-82 and (m, p)-BrPA-77 were prepared either by non-isotopic [ 77 ' 82 Br]bromo-deiodination at 160 0 C or by [ 77 ' 82 Br]bromo-demetalation, using (m,p)- iodo-L-phenylalanine or the corresponding (m,p)-tributyltin- or (m, p)-trialkylsilyl-L- phenylalanine, as starting materials in analogy to the procedure described above, (m, p)-BrPA-82 and (m, p)-BrPA-77 were isolated by means of HPLC as no carrier-added products with a specific activity > 500 GBq / ⁇ mol.
  • the fractions containing the radiobrominated products were buffered with phosphate buffered saline (PBS), and sterile filtered through a 0.22 ⁇ m sterile membrane to an isotonic and injectable radiopharmaceuticals for in vitro and in vivo investigations.
  • PBS phosphate buffered saline
  • the human prostate cancer cells PC3 and DU145 American Type Culture Collection, Rockville, MD
  • the human breast SK-BR-3 and BT-474 cancer cell lines as well as the melanoma cell lines SK-MEL25 and A101 D were purchased commercially or provided by the oncological research laboratory of the University Medical Center of Saarland (Homburg, Germany).
  • Cells were cultivated in RPMI-1640 medium or in Dulbecco's modified Eagle medium (sodium pyruvate-free, supplemented with L-glucose and pyridoxine), respectively, supplemented with 10 % (v/v) heat-inactivated foetal calf serum (FCS), penicillin (50 U/ml), streptomycin (50 ⁇ g/ml), and insulin (50 ⁇ g/ml; PromoCell, Heidelberg, Germany). All cells lines were maintained in appropriate flasks in a humidified incubator (5% CO 2 ) at 37°C.
  • FCS foetal calf serum
  • penicillin 50 U/ml
  • streptomycin 50 ⁇ g/ml
  • insulin 50 ⁇ g/ml
  • PromoCell Heidelberg, Germany
  • the tumor cells were preincubated for 5 min in 500 ⁇ L medium at 37°C in 1.5-ml Eppendorf centrifuge tubes. Aliquots of 30-50 ⁇ L (10 6 - 1.5 x 10 6 cpm) freshly prepared radiopharmaceutical were added and cells incubated at 37°C / 5% CO 2 for 1 , 2, 5, 15, 30, 60, 90 and 120 min while shaking. Uptake was stopped with 500 ⁇ L ice-cold PBS (pH 7.4) and an additional 3-min in an ice bath, the cells were centrifuged for 2 min at 300 x g, the supernatant removed and the pellet washed three time with ice-cold PBS.
  • cytostatic effect and radiosensitizing effect of IPA in EGFR positive human tumor cells is demonstrated in figures 1 and 2.
  • the cytostatic effect of IPA-131 on EGFR positiv tumor cells was more pronounced as compared to external irradiation up to 15 Gy (Fig. 1).
  • Combining IPA-131 with external irradiation led to a dramatic reduction of the cell survival rate (Fig. 2).
  • Flow-cytometric analyses of stained cells show dose dependent induction of primary necrosis and apoptosis, which was more significant than that caused by external irradiation, even with 15 Gy, and more pronounced with increasing IPA-concentration. This result attests the high radiosensitizing effect of IPA-131 on EGFR-positive cells.
  • the surviving cells contained only sparse cytoplasm, the nuclei were shrunken and contained condensed or clumped chromatin. Cytologically, the mode of cell death was apoptosis as the remaining tumour cells contained only sparsely cytoplasm and apoptotic bodies, in other cells the nuclei were shrunken and contained condensed chromatin. Identical results were obtained with BrPA-77 in prostate and breast cells in vitro. This results suggests the high therapeutic potential of 3/4-halogenated L-phenylalanine hormone-independent or metastatic human tumors, especially for hormone- independent prostate and metastatic breast cancer.
  • Reference Example Figure 3 shows an example of uptake kinetic of 4-iodo-L-phenylalanine in primary human androgen-independent tumor cells.
  • the radiolabeled derivative 4-[ 123 l]iodo-L- phenylalanine was used to facilitate quantification, using a gamma counter.
  • IPA exhibit high uptake in human tumor cells with a continuous increase over the investigation time. This result provides evidence of the high affinity of the proposed radiopharmaceuticals for human tumors, including the human malignant gliomas, pancreatic carcinomas, prostate and breast cancer.
  • Mishrat SK Mazumhar A, Vadlamudi RK, et al. MICoA, a novel metastasis- associated protein 1 (MTA1) interacting protein coactivator, regulates estrogen receptor- ⁇ transaction functions. J Biol Chem 2003; 278: 19209 - 19219.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physics & Mathematics (AREA)
  • Optics & Photonics (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides a use of a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 for the preparation of a pharmaceutical composition for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma. Furthermore, the invention provides an according method for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma and a method for the monitoring of the progress of such treatment.

Description

Therapy of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma
The present invention provides a use of a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine- 211 for the preparation of a pharmaceutical composition for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma. Furthermore, the invention provides a method for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma and a method for the monitoring of the progress of such treatment.
A variety of documents is cited throughout this specification. The disclosure content of said documents including manufacturer's manuals is herewith incorporated by reference in its entirety.
Breast and prostate cancer, which are hormone dependent carcinomas, are the second most common cause of cancer-related death, for women and men in the western world [1-4]. According to [5], the highest cancer incidence and death rates for each racial and ethnic population in the USA continued to be for cancers of the prostate, and lung, and colon among men and for cancers of the breast, lung and colon among women. Moreover, more than 30% of newly diagnosed cancers are breast and prostate cancer, making them a leading cause of death from cancer worldwide [2, 3].
The more current treatments used for prostate cancer (PC) are the androgen- deprivation therapies, radiotherapy and chemotherapy [6 - 8]. However, although these therapies are effective for patients with localized and androgen-responsive PCs, they have demonstrated limited curative effect against advanced metastatic and androgen-resistant states of PCs, and no demonstrable survival benefit has been demonstrated in randomized studies [8, 9]. Various new treatment modalities are currently being developed, including second-line hormonal therapy, immunotherapy, radical prostectomy, gene therapy, dendritic cell vaccination therapy, antisense therapy, growth factor inhibition, vitamin D, and biphosphonates. Unfortunately, in spite of all these efforts the prognosis of metastatic, hormone-refractory prostate cancer remains poor, and no significant improvement in median survival has been demonstrated [9].
For early-stage of breast cancer, two treatment regimens have been used: 1) breast- conserving surgery and radiation, and 2) adjuvant chemotherapy and hormonal therapy [5, 10, 11]. The first treatment regimen has shown to be an appropriate primary therapy for the majority of women with stage I and Il breast cancer, whereas the second one has been used in women with early-stage node-positive disease [10, 11]. Despite advances in screening for breast cancer, improved locoregional treatments, and adjuvant systemic therapy, the management of metastatic breast cancer remains in analogy to metastatic androgen-resistant prostate cancers a major clinical challenge. The main goals of treatment are palliation of symptoms, disease control, and, when possible, prolongation of life [12]. Cytotoxic chemotherapy using anthracyclines or melphalan as well as newer drugs such like paclitaxel or docetaxel, has been shown to be of clinical benefit [13]. However, pilot studies using these chemotherapy regimens to treat advanced stage cancers revealed high rates of congestive cardiac failure [18 % to 20 %], and toxic effects on bone marrow and other organs, limiting clinical application [14]. In summary, none of the treatments currently used has shown a significant survival benefit in patients with metastatic, hormone- refractory prostate cancer or metastatic breast cancer as yet. The median survival time for these patients remains in the range of 1 to 3 years [9, 12]. Accordingly, there is still a lack for a therapy approach which is capable to improve the outcome of patients with hormone-refractory and/or metastatic disease. A cytostatically active drug, with a favorable tolerability profile, overcoming cellular detoxification strategies, and ideally conveying additional anti-tumor activity, therefore, would be desirable as an alternative for maintenance or induction therapy in tumor, to improve tolerability and efficacy of existing chemo- and radiotherapy regimens.
Thus, the technical problem underlying the present invention is to provide means and methods for an improved treatment of hormone dependent carcinoma and hormone- refractory and/or metastatic disease. The solution to this technical problem is achieved by the embodiments characterized in the claims.
Accordingly, the present invention relates to the use of a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 for the preparation of a pharmaceutical composition for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma, wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma.
The term "hormone dependent carcinoma" describes in the context of the present invention any human cancer which is histologically derived from tissues of the primary or secondary male or female sexual organs, and the functional regulation of which is controlled primarily by sexual hormones of the steroid class, typically androgens (e.g. testosterone) or gestagens (e.g. progesterone) or estrogens (e.g .estradiol). Typically, the development of the primary tumor depends on hormone availability. Tumors can initially be controlled by anti-hormon agents (e.g. tamoxifen, an anti-estrogen). However, most tumors later progress to more malignant hormone-independent and/or metastatic forms. These forms may be resistant to many current therapies. The term "metastasized carcinoma derived from hormone dependent carcinoma" defines according to the invention such malignant hormone-independent and/or metastatic forms. The carcinoma to be treated by the use or method of the invention are selected from the group consisting of prostate carcinoma and mamma carcinoma. The term "prostate carcinoma" describes a type of hormone dependent cancer of the human prostate gland which is initially characterized by androgen-dependence, and which does typically progress to an androgen-resistant state. Such a carcinoma may demonstrate resistance to many chemotherapeutic agents that are diverse both in structure and mechanism of action.
The term "mamma carcinoma" is understood in the context of the invention as a synonym for "breast cancer". The term describes a further type of hormone dependent cancer of the human mammary glands which is also characterized by progress to therapy resistant metastatic state.
An indication to treat a subject according to the invention can be derived by the diagnosis of minimal residual disease, preferably early solid tumor, advanced solid tumor or metastatic solid tumor, which is characterized by the local and non-local recurrence of the tumor caused by the survival of single cells. According to the invention, it is envisaged that a L-phenylalanine is conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine- 211. Exemplary for the compounds of the invention and in particular as a proof of principle for IPA-131 , orphan status was granted by the EMEA on April 11 , 2006 under the EU designation number EU/3/06/363.
The term "alpha-, beta- or Auger-electron emitting isotope" defines in the context of the present invention radioactive istopes, characterised by the emission of different particles (rays) formed during radioactive decay or by nuclear transition processes. An alpha emitting isotope is defined as a radioactive nuclide emitting alpha particles, corresponding to a helium nucleus consisting of two protons and two neutrons. A beta emitting isotope is defined as a nuclide emitting fast nuclear electrons (negatrons) formed during radioactive decay. An Auger-electron emitting isotope is defined as a nuclide emitting low energy nuclear electrons, formed by nuclear electron capture or internal transition processes. The maximum path lengths of these particles are in a range from 10 nm to 12 mm.
L-phenylalanine derivatives in the forms applicable in accordance with the present invention are presented in by general formula I:
Figure imgf000005_0001
General formula I in which,
X is bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 or astatine-211 linked to L-phenylalanine at the 3- (meta-) or 4- (para-) position within the aromatic ring.
Ri is H, alkyl group, amino acid, peptide, protein or an other residue known to facilitate or improve tumor targeting. R2 is OH, amino acid, or an other residue known to facilitate or improve tumor targeting. Preferred conjugates according to the formula I are those in which X is a bromine-77, bromine-82, iodine-125, iodine-131 , or astatine-211 linked to L-phenylalanine at the para- position of the aryl group, while Ri is H and R2 is OH.
According to the physical half life of the radionuclide conjugated to the L- phenylalanine, also the conjugates have a corresponding half life of 16.2 h for bromine-76, 57.04 h for bromine-77, 35.3 h for bromine-82, 4.17 d for iodine-124, 59.41 d for iodine-125, 8.02 d for iodine-131 or 7.21 h astatine-211 labelled L- phenylalanine, respectively. As described in more detail in the appended examples, the halogen isotope may e.g. be conjugated following a protocol for a "non-carrier- added" (n.c.a.) conjugation as well as following a protocol for a "carrier-added" (c.a.) conjugation; see e.g. appended example 1.
Due to the presence of a radionuclide, which is an alpha-, beta- or Auger-electron emitting isotope, in the described conjugate the effective compound is an endoradiotherapeutic agent. The term "endoradiotherapeutic agent" defines in the context of the present invention an agent which comprises at least one type of radioactive isotopes. Such agent is to be administered to a subject in the need thereof and is effective in the therapy of the above described carcinoma with the ability to metastasize due to an endogenic irradiation, i.e. an irradiation with the radioactive compound within the body of the subject to be treated by the endoradiotherapy. The L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope may be administered to the subject in the need thereof via a parenteral, transdermal, intraluminal, intra-arterial, intrathecal or intravenous route or by direct injection into malignant tissue. Also within the scope of the invention is an administration wherein the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is bound to a matrix of a medical device. The conjugate may be bound, for example, to a suitable matrix via an amino binding between the amino group of the phenylalanine and a matrix coated with polypeptides. The medical device may be e.g. a wound plating such as a gauze, a metal plating or different, preferably biological, carrier material.
It is preferred that the effective compound is formulated in form of a pharmaceutical composition to the subject in the need thereof. In accordance with this invention, the term "pharmaceutical composition" relates to a composition for administration to a subject, preferably a human patient. As generally noted above the pharmaceutical composition is preferably administered parenterally, transdermal^, intraluminally, intra-arterially, intrathecally or intravenously. Also preferred is a direct injection of the pharmaceutical composition into malignant tissue. It is in particular envisaged that said pharmaceutical composition is administered to a patient via infusion or injection. Administration of the suitable compositions may be effected by different ways, e.g., by intravenous, subcutaneous, intraperitoneal, intramuscular, topical or intradermal administration. The pharmaceutical composition may further comprise a pharmaceutically acceptable carrier. Examples of suitable pharmaceutical carriers are well known in the art and include phosphate buffered saline solutions, water, emulsions, such as oil/water emulsions, various types of wetting agents, sterile solutions, etc. Compositions comprising such carriers can be formulated by well known conventional methods. These pharmaceutical compositions can be administered to the subject at a suitable dose. The dosage regimen will be determined by the attending physician and clinical factors. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. Preferred dosages for the administration of the L- phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope are described herein below. The compositions may be administered locally or systematically. Administration will generally be parenteral, e.g., intravenous. In an preferred embodiment, the pharmaceutical composition is administered subcutaneously and in an even more preferred embodiment intravenously. Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like. In addition, the pharmaceutical composition might comprise proteinaceous carriers, like, e.g., serum albumine or immunoglobuline, preferably of human origin. It is envisaged that the pharmaceutical composition might comprise, in addition to L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope, further biologically active agents, depending on the intended use of the pharmaceutical composition in a treatment comprising the administration of additional agents for a concomitant therapy. Examples for such further biologically active agents are described herein below in the context of uses and methods comprising a concomitant therapy.
Generally, the L-phenyalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is to be administered in doses of 10"5 to 10~18 g / kg body weight. More preferably, the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is administered in doses of 10"7 to 10~15 g / kg body weight and more preferably, in doses of 10"8 to 10"10 g / kg body weight. It is preferred that such a dose is formulated contained in 1 to 10, preferably 2 to 5 ml of sterile solution, such as phosphate buffered saline solutions, water for injection, etc.
It has been surprisingly found that the above described conjugates are capable to accumulate specifically and with marked retention in a limited group of hormone dependent carcinoma and hormone-refractory or metastasized carcinomas derived from hormone dependent carcinomas. As demonstrated in the appended examples, the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope specifically accumulates in prostate carcinoma cells, whereas other carcinoma with the ability to metastasize do not or do not specifically accumulate the conjugate. Examples for carcinomas, respectively for metastasized carcinomas which do not or do not specifically accumulate the conjugate are known in the art and include lung cancer, pancreatic cancer, colorectal cancer, metastases of malignant melanoma, myeloma, lymphoma and metastases of cancers with unknown primary localization.
Due to the specific accumulation of the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope this conjugate is useful in the treatment of such specific hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma by specifically targeting the cells of this carcinoma and to destroy it by the irradiation activity of the emitting isotope.
The effectiveness and safety of single photon emission tomography (SPET) with 4- [123l]iodo-L-phenylalanine (IPA-123) for brain tumor imaging not only in the experimental C6 glioma model but also in patients with malignant gliomas has been previously demonstrated [15 - 17]. This data demonstrates that this group of compounds is also capable to cross the blood-brain barrier after the intravenous administration. Moreover, and advantageous for the safety of the administration according to the present invention, it was demonstrated that the accumulation in the normal tissue and also in cerebral parenchyma is moderate and decreases rapidly with time. However, due to the complete difference of the origin and the stages of development of hormone dependent carcinoma and malignant gliomas it could not be expected that an L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope might be effective in the treatment of hormone dependent carcinoma. On the one hand development and upgrading of gliomas appear to be etiologically hormone independent [18, 19]. In addition, malignant gliomas lack the ability to form metastases. It is understood that the high accumulation of IPA-123 in gliomas is primarily associated with the increased amino acid transport into glioma cells. This has been shown to be specific for many tumors [20, 21]. On the other hand hormone dependent carcinomas are characterized by a progress to a hormone-independent and therapy resistant metastatic state, which has been associated with the activation of other signaling cascades by growth factors that control the strict balance between the growth rate and apoptosis [7, 22]. Among them, the EGF and transforming growth factor alpha (TGF-α) play a major role during carcinogenesis. In particular, the stimulation of EGFR, insulin-like growth factor-l (IGF-I)1 protein Kinase A cascades and members of the erbB family is understood to lead to recruitment of proteins that interact and activate androgen or estrogen receptors in the absence of hormones [7, 22, 23]. It is further understood that the expression levels of EGFR and its ligands EGF and TGF-α in prostate carcinoma cells is generally enhanced during the disease progression to more malignant hormone-independent and metastatic PC forms, where it regulates angiogenesis, tumor growth and progression, and metastasis [7, 22]. Because the uptake of IPA-123 was previously known only as moderate in hormone dependent PC cells and in brain metastases of small cell lung cancers and adenocarcinomas [17], the surprising feature of the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is the unexpected high accumulation in androgen-independent PC3 and DU 145 PC cells, and in metastatic breast tumor cells. This provides the rationale for an attractive novel tool with a clinical impact on targeting of relapsed carcinomas derived from hormone dependent tissues. The surprisingly high accumulation of the para-iodinated L-phenylalanine in androgen- independent PC3 and DU145 PC cells, and in hormon-independent or metastatic breast tumor cells is understood as associated with the modified transcriptional activity or inactivation of the hormonal receptor pathway in disease, which leads to enhanced erbB transduction and, therefore, to enhanced protein demand in malignant cells or metastases [24, 25]. There is no known correlation between cellular overexpression of EGFR and enhanced amino acid transport in the tumor cell, although overexpression of EGFR, and increased erbB oncogene has been demonstrated in high grade gliomas [19, 26].
Accordingly, the invention teaches that the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope specifically accumulates in hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma as well as such metastasized carcinoma which migrated into the brain. As described herein above it is preferred that the conjugated L-phenylalanine is to be administered intravenously. The intravenous administration may be effected by administering the conjugate in form of an above described pharmaceutical composition for parenteral administration.
It is also preferred that the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L-phenylalanine is in the range of 0.1 to 1000 MBq/kg body weight. Generally, but not exclusively, this dose range overlaps with the above indicated dose range calculated in terms of g / kg body weight. More preferably, the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope is in the range of 10 to 400 MBq/kg body weight, more preferably the dose is in the range of 20 to 120 MBq/kg body weight. The administered dose can be determined using an appropriate dose meter, calibrated to quantitatively measure alpha, beta or gamma radiation.
It is also preferred that the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L-phenylalanine is to be administered as a single dose once or as fractionated doses in 2 to 60 fraction doses. More preferably, the conjugate is administered fractionized in 2 to 10 fraction doses. Dose fractionation is an established procedure in radiation therapy. By fractionating a total administered dose, improved tolerability for healthy non-target tissue, as well as an increased cytotoxic effect to tumor tissue is achieved. Repeated fractionated irradiation allows to therapeutically impact a higher percentage of cells in radiation sensitive stages of the cell cycle, compared to a one time single high dose irradiation. Therapeutic irradiation induces single and double strand breaks of DNA, which is counteracted by nuclear repair mechanisms upregulated following irradiation. It is believed that cells undergoing DNA repair, are more susceptible to a renewed irradiation than radiation- naive cells.
In a further preferred embodiment of the use of the invention the conjugated L- phenylalanine is 4-[131l]iodo-L-phenylalanine (IPA-131), 4-[124l]iodo-L-phenylalanine (IPA-124) and 4-[211At]astatine-L-phenylalanine (AtPA-211). lodine-131 is widely available, has a favourable half life and can be handled by most institutions licensed to apply open radionuclides, lodine-131 allows for convenient extracorporal therapy monitoring using a gamma camera owing to a gamma ray component, emitted in a fixed ratio relative to the therapeutic beta particle emission, which is itself not detectable extracorporally. Another preferred embodiment of the method of the invention makes use of 4-[124l]iodo-L-phenylalanine. lodine-124 has a positron emission component, allowing for PET imaging, in addition to the therapeutic beta- emission. Using quantitative PET imaging, internal dosimetry measurements at an ongoing basis can be conducted for therapy planning and therapy monitoring for a period of up to 15 days following a single injection. Astatine-211 is also preferred, as it emits high energy (6.8 MeV) alpha particles, with a short path length in tissue (65μm), allowing to administer a highly cytotoxic radiation to targeted tissue, while minimising undesirable radiation effects to non-target tissue.
Moreover, it is preferred that the pharmaceutical composition further comprises a chemo therapeutic agent, an immunotherapeutic agent, a gene therapeutic agent, a vaccine, an antisense nucleotide therapeutic agent, an siRNA therapeutic agent, a further endoradiotherapeutic agent and/or a further radiosensitising agent. The administration of a chemo therapeutic agent, an immunotherapeutic agent, a gene therapeutic agent, a vaccine, an antisense nucleotide therapeutic agent, an siRNA therapeutic agent, a further endoradiotherapeutic agent and/or a further radiosensitising agent is understood as a concomitant therapy. Methods and means for concomitant therapies are well known in the art.
The conjugated L-phenylalanine and the additional therapeutic may be formulated as a single pharmaceutical composition for simultaneous administration of the effective compounds or in separate pharmaceutical composition for sequential administration. Accordingly, an administration of a composition comprising the conjugated L- phenylalanine prior to the administration of a composition comprising one or more therapeutics selected from the group of a chemo therapeutic, an immunotherapeutic, a gene therapeutic, a vaccine, an antisense nucleotide therapeutic, an siRNA therapeutic, a further endoradiotherapeutic agent and/or a further radiosensitising agent is envisaged as well as simultaneous or subsequent administration. An example for a chemo therapeutic agent comprises bioactive agents known to be effective in retarding or arresting the malignant growth or to be effective in the regression or elimination of malignant tissues or cells. Such agents might be e.g. drugs acting as cytostatics. Accordingly, a chemotherapy comprises in line with the medical standards in any systemic or local treatment the administration of cytostatic or cytotoxic agents. Chemotherapeutic agents used in oncology include among others, nitroso urea compunds (ACNU [nimustin], BCNU [carmustin], CCNU [lomustin]), temozolomid, procarbacin, metothrexate, cytarabin, gemcitabine, fluorouracil, cyclophosphamide, mitoxantron, anthracyclins, estramustin, or taxanes. The chemotherapeutic agents are intended to be administered in appropriate dosing regimens according to medical practise. In line with the invention nitroso urea compounds, temozolomide, procarbacin, and methotrexate are preferred chemotherpeutic agents.
Examples for an immunotherapeutic agent comprise but are not limited to compounds such as antibodies, antibody fragments and/or derivatives thereof which specifically detect malignant tissue and/or cellular therapeutics, including those consisting of adoptively transferred autologous, heterologous, xenogenous or endogenous cells, which have the ability to eliminate malignant cells or tissues. The term "antibody fragment or derivative thereof relates to single chain antibodies, or fragments thereof, synthetic antibodies, antibody fragments, such as Fab, a F(ab2)', Fv or scFv fragments, single domain antibodies etc., or a chemically modified derivative of any of these. Antibodies to be employed in accordance with the invention or their corresponding immunoglobulin chain(s) can be further modified outside the motifs using conventional techniques known in the art, for example, by using amino acid deletion(s), insertion(s), substitution(s), addition(s), and/or recombination(s) and/or any other modification(s) (e.g. posttranslational and chemical modifications, such as glycosylation and phosphorylation) known in the art either alone or in combination. Methods for introducing such modifications in the DNA sequence underlying the amino acid sequence of an immunoglobulin chain are well known to the person skilled in the art; see, e.g., Sambrook et al.; Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory Press, 2nd edition 1989 and 3rd edition 2001. The specific detection of malignant tissue or cells may be effected via the detection of tumor specific markers by the antibodies, antibody fragments and/or derivatives thereof. A tumor-specific marker is a tumor-associated cell surface antigen which is either found exclusively on tumor cells or is overexpressed on tumor cells as compared to non- malignant cells. Tumor-associated cell surface antigens can be expressed not only on tumor cells but also on cells/tissue which are/is not essential for survival or which can be replenished by stem cells not expressing tumor-associated cell surface antigen. Furthermore, a tumor-associated cell surface antigen can be expressed on malignant cells and non-malignant cells but is better accessible by a therapeutic agent of interest on malignant cells. Examples of over-expressed tumor-associated cell surface antigens are Her2/neu, EGF-Receptor, Her-3 and Her-4. An example of a tumor- associated cell surface antigen which is tumor specific is EGFRV-III. An example of a tumor-associated cell surface antigen which is presented on a cell which is non- essential for survival is PSMA. Examples of tumor-associated cell surface antigens which are presented on cells which are replenished are CD19, CD20 and CD33. An example of a tumor-associated cell surface antigen which is better accessible in a malignant state than in a non-malignant state is EpCAM. Moreover, the definition of "immunotherapeutics" may comprise agents such as T-cell co-stimulatory molecules or cytokines, agents activating B-cells, NK-cells or other cells of the immune system as well as drugs inhibiting immune reactions (e.g. corticosteroids).
The term "gene therapeutic agent" defines in the context of the invention means for a therapy comprising the administration of one or more nucleic acid constructs functionally encoding e.g. one or more antigens which are characteristic for malignant cells. Such antigens comprise tumor specific markers. The sequence encoding such antigen is operably linked to a nucleic acid sequence which is a regulatory sequence. Thus, a gene therapy comprises the functional expression of a heterologous gene in a patient according to standard medical protocols using appropriate vector systems known in the art; see e.g. Haberkorn et al., Curr Med Chem. 2005; 12(7)779-94. The term "regulatory sequence" refers to DNA sequences which are necessary to effect the expression of coding sequences to which they are ligated. Control sequences in the context of the described gene therapy generally include promoters, terminators and, in some instances, enhancers, transactivators or transcription factors. The term "control sequence" is intended to include, at a minimum, all components the presence of which are necessary for expression, and may also include additional advantageous components. The term "operably linked" refers to a arrangement/configuration wherein the components so described are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
The administration of a vaccine aims in the context of the present invention at activating the innate or adaptive immune system of the patient to act against the tumor tissue or the malignant cells. Such therapy comprises e.g. administering one or more antigen preparations containing tumor substances, or cells selected to react against tumor tissue or the malignant cells.
An antisense therapeutic agent is e.g. a nucleotide sequence being complementary to tumor-specific gene sequences, aiming at functionally neutralising tumor gene expression, and consequently inducing tumor cell death.
An siRNA therapeutic agent according to the invention is e.g. a small interfering RNA capable of sequence-specifically silencing the expression and activity of various tumor-specific target genes by triggering cleavage of specific unique sequences in the mRNA transcript of the target gene and disrupting translation of the target mRNA, consequently inducing tumor cell death.
A concomitant therapy which requires the administration of additional bioactive agents which are effective in the treatment of the carcinoma with the ability to metastasize may be accompanied by the administration of additional compounds which minimize potential side effects of said bioactive agents such as drugs acting on the gastro- intestinal system, drugs preventing hyperuricemia, and/or drugs acting on the circulatory system, e.g. on the blood pressure, known in the art. Such additional bioactive agents may be formulated in the form of the same or a separate pharmaceutical composition.
The term "endoradiotherapeutic agent" has been described herein above.
Examples for radiosensitizing agents are 3-iodo-L-phenylalanine or 4-iodo- phenylalanine, which are characterized by a stable, non-radioactive iodine of the [127I]- isotope. The term "radiosensitizing effect" describes in the context of the present invention the capacity of a compound to enhance the therapeutic response to concomitantly administered radiation therapy. Such radiation therapy includes external and internal radiation therapies. This is understood as the ability to induce an increased response to a given radiation dose administered in the presence of the radiosensitizing compound, compared to the response induced by the same radiation dose in the absence of the radiosensitizing compound, or, alternatively, to induce selectively the sensitivity of neoplastic cells for a radiotherapy, not present in the absence of the compound.
It is preferred for the use of the invention that the pharmaceutical composition is to be administered to a patient who is subsequently to be irradiated percutaneously (percutaneous radiotherapy). Such percutaneous exoradiotherapy is understood in the context of the invention as a concomitant therapy. Percutaneous radiotherapy is typically administered as an external beam radiation stemming from among others, radioactive cobalt-60 sources, linear accelerators, proton, neutron, or hadron beam sources. Preferably, the irradiation is started in a period of 0 to 7 days subsequent to the administration of the conjugated L- phenylalanine. More preferably, the irradiation is started in a period of 0.5 to 24 hours subsequent to the administration of the conjugated L-phenylalanine.
The concomitant radiotherapy may comprise a cumulative external irradiation of a patient in a dose of 1 to 100 Gy. A preferred range of the irradiation dose is 1 to 60 Gy. It is preferred that the external irradiation dose is administered in 1 to 60 fractional doses, more preferably in 5 to 30 fractional doses. Preferably, the fractionized doses are administered over a period of 1 to 26 weeks, more preferably over a period of 6 to 12 weeks.
In an alternative embodiment, the invention provides a method for the monitoring of the progress of a treatment of carcinoma with the ability to metastasize (in a human subject) selected from the group consisting of prostate carcinoma and mamma carcinoma (breast cancer), the method comprising the step of localizing and/or dosimetrically measuring a L-phenylalanine conjugated to an alpha-, beta- or Auger- electron emitting isotope selected from the group consisting of bromine-76, bromine- 77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 in the subject by using a γ-camera subsequent to the administration of a pharmaceutical composition comprising said conjugate to a subject in the need thereof. The detection of an interception of tumor progression, a decrease of the tumor size or an elimination of a tumor is indicative of a therapeutic success.
An example for the localization and/or dosimetric measurement of conjugated L- phenylalanine in the subject by using a γ-camera is exemplified e.g. in [16, 17]. It is preferred that the conjugated L-phenylalanine is localized and/or dosimetrically measured at least 0 to 7 days, preferably 0.5 to 48 h subsequent to the administration of the pharmaceutical composition.
In a further alternative embodiment, the invention provides a method for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma, wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma, the method comprising the steps of administering a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 to a subject in the need thereof. It is preferred that the effective compound, which is the conjugated L-phenylalanine, is formulated in form of a pharmaceutical composition. The term "pharmaceutical composition" has been defined herein above. The route of administration of the effective compound depends inter alia from its formulation. Different routes for differentially formulated compositions have been described herein above. It is preferred for the method of the invention that the conjugated L-phenylalanine is administered intravenously.
It is further preferred that the conjugated L-phenylalanine labeled with alpha-, beta- or Auger-electron emitting isotope is administered to the subject in a doses of 10"5 to 10" 18 g / kg body weight. More preferably, the L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is administered in doses of 10"7 to 10"15 g / kg body weight of the subject and more preferably in doses of 10"8 to 10"10 g / kg body weight of the subject. It is preferred that the radioactive dose of the conjugated L- phenylalanine labeled with alpha-, beta- or Auger-electron emitting isotope is in the range of 0.1 to 1000 MBq/kg body weight. Generally, but not exclusively, this dose range overlaps with the above indicated dose range calculated in terms of g / kg body weight. More preferably, the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope is in the range of 10 to 400 MBq/kg body weight and more preferably in the range of 20 to 120MBq/kg body weight. As described herein above, it is also preferred that such dose is formulated contained in 1 to 10, preferably 2 to 5 ml of sterile solution, such as phosphate buffered saline solutions, water for injection, etc.
Also preferred for the method for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma of the invention is the administration of the conjugated L-phenylalanine to a subject in the need thereof, wherein the conjugated L-phenylalanine is 4 -[131l]iodo-L- phenylalanine (IPA-131), 4-[124l]iodo-L-phenylalanine (IPA-124) or 4-[211At]astatine-L- phenylalanine (AtPA-211).
In an also preferred embodiment of the invention it is envisaged that the method further comprises the step of a treatment of the subject by a concomitant therapy. Said concomitant therapy may be selected from the group consisting of a surgical therapy, a chemotherapy, an endo- or exoradiotherapy, an immunotherapy, a gene therapy, a vaccine therapy, an antisense nucleotide therapy, an siRNA therapy, an intracavitary therapy, a radiosensitizer therapy, or a device-based treatment. The step of the concomitant therapy may be effected prior, simultaneous or subsequent to the step of administering a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope.
Definitions for a chemotherapy, an endo- or exoradiotherapy, an immunotherapy, a gene therapy, a vaccine therapy, an antisense nucleotide therapy, an siRNA therapy and radiosensitizer therapy have been provided herein above. Methods and means for concomitant therapies are well known in the art. An example for a surgical therapy comprises a resection of a solid tumour or of malignant tissue. A further concomitant therapy in line with the invention comprises the surgical implantation of a radioactive device such as a radioactive seed. Such seed may be implanted locally to the tumor site. The technique of implanting radioactive devices is known in the art and described herein above in the discussion of the state of the art.
In a preferred embodiment of the method of the invention the concomitant therapy is an exoradiotherapy (also called external beam radiation therapy) and wherein the irradiation is a percutaneous radiotherapy. Preferably, the irradiation is started in a period of 0 to 7 days subsequent to the administration of the conjugated L- phenylalanine. More preferably, the irradiation is started in a period of 0.5 to 24 hours subsequent to the administration of the conjugated L-phenylalanine.
The above described concomitant exoradiotherapy may comprise a cumulative external irradiation of a patient in a dose of 1 to 100 Gy. A preferred range of the irradiation dose is 1 to 60 Gy. It is preferred that the external irradiation dose is administered in 1 to 60 fractional doses, more preferably in 5 to 30 fractional doses. Preferably, the fractionized doses are administered over a period of 1 to 26 weeks, more preferably over a period of 6 to 12 weeks. As described herein above, external field radiation therapy is typically administered as an external beam radiation stemming from among others, radioactive cobalt-60 sources, linear accelerators, proton, neutron, or hadron beam sources. In accordance with the present invention, the term 'fractional dose' is to be understood to mean that the overall activity of the fractional dose adds up or essentially adds up to the cumulative external irradiation otherwise also achievable by administering one single dose.
It is also preferred that the subject to be treated by all embodiments of the method of the invention is a human subject.
In an alternative embodiment the invention relates to a method for the monitoring of the progress of a treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma (in a human subject), wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma, the method comprising the steps: (a) administering a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 to a subject in the need thereof; and (b) localizing and/or dosimethcally measuring the conjugated L-phenylalanine in the subject by using a γ-camera.
The detection of an interception of tumor progression, a decrease of the tumor size or an elimination of a tumor is indicative of a therapeutic success.
A method for the administration of a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope is described herein above. As also described herein above, an example for the localization and/or dosimetric measurement of conjugated L-phenylalanine in the subject by using a γ-camera is exemplified e.g. in [16, 17]. The method for the monitoring of the progress of a treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma may also comprise the step of a treatment of the subject by an above described concomitant therapy. As described herein above, the step of the concomitant therapy may be effected prior, simultaneous or subsequent to the step of administering a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope.
Preferably, the conjugated L-phenylalanine is localized and/or dosimetrically measured at least 0 to 7 days, more preferably 0.5 to 48 h subsequent to its administration.
The figures show:
Figure 1 :
Induction of primary necrosis and apoptosis by IPA-131 vs. External irradiation (15 Gy) in a EGFR-positive human model cell line Figure 2:
Effect of IPA-131 and external irradiation on survival of EGFR-positive cells (human model cell line in vitro) Figure 3:
Cellular uptake kinetics of IPA (IPA-123) in hormone independent human prostate cancer cell lines in vitro
The invention will now be described by reference to the following examples which are merely illustrative and are not to be construed as a limitation of scope of the present invention.
Example 1 : 4-Bromo-L-phenylalanine (4-BrPA), 3-bromo-L-phenylalanine (3-BrPA), 4-iodo-L- phenylalanine (4-IPA), 4-ter.butyltinn-L-phenylalanine (4-TBSnPA), 3-ter.butyltinn-L- phenylalanine (3-TBSnPA), 4-methylsilyl-L-phenylalanine (4-Me3SiPA) and 3- methylsilyl-L-phenylalanine (3-Me3SiPA) used as starting materials (precursor) for radiolabeling were either purchased commercially or prior synthesized in analogy to the literature. Unless stated otherwise, all other chemicals and solvents were of analytical grade and obtained commercially or via our local hospital pharmacy. Sodium [124l]iodide, sodium [125l]iodide, sodium [131l]iodide, sodium [77Br]bromide, sodium [82Br]bromide, and sodium [211At]astate for radiolabeling was obtained in the highest obtainable radiochemical purity, generally in 0.01 N NaOH or in phosphate buffered saline (PBS) from different suppliers. HPLC purification was performed on a Hewlett Packard HPLC system consisting of a binary gradient pump (HP 1100), a Valco 6-port valve with 2500 μl loop, a variable wavelength detector (HP 1100) with a UV detection at 254 nm and a sodium iodide scintillation detector (Berthold, Wildbad, Germany), using reversed-phased column (250 x 4 mm, Nucleosil-100). The column was eluted at different flow rates in with water/ethanol/acetic acid (89:10:1 ; v/v) or PBS / ethanol (90:10; v/v).
The proposed radiolabeled phenylalanines were obtained either by non-isotopic halogen exchange (carrier-added/c.a.) or by radio-demetalation of the corresponding precursor as described in the general scheme 1 , resulting to no-carrier-added (n. c. a) products after HPLC separation.
Figure imgf000022_0001
R = 124/125/131| or 77/82gr
X = m- , p-Br or m-, p-l for 77/82Br n.c.a. (m, p)-IPA-124, -IPA-125, -IPA-131 or n.c.a. (m, p)-BrPA-77 and -BrPA-82
Figure imgf000022_0002
X = t-Bu3Sn or R = (124/125/131)^ 77/82Br
(CH3)3Si Or 21 1At n.c.a. (m, p)-BrPA-77, -BrPA-82, -IPA-124, -IPA-125, -IPA-131 and n.c.a. (m, p)-AtPA-211
Scheme 1 : scheme of the radiosyntheses of n.c.a. IPA-124, IPA-125, IPA-131 , BrPA- 77, BrPA-82 and AtPA-211
Example 2:
Figure imgf000022_0003
phenylalanine (m,p-IPA-125) and 3,4-r131lliodo-L-phenylalanine (m,p-IPA-131) by non- isotopic radioiodo-debromination A solution of carrier free sodium [124l]iodide, sodium [125l]iodide or sodium [131l]iodide (up to 5 GBq) and 5 μl aqueous Na2S2O5 (4.0 mg Na2S2O5ZmI) was evaporated to dryness by passing a stream of nitrogen through a reaction vessel at 1000C, followed by addition of 200 μl of the corresponding L-bromophenylalanine (0.25 - 0.5 mg/ ml 0.1 N H3PO4), 20 μl aqueous L-ascorbic acid (10 mg/ml) and 20 μl aqueous Cu(II) sulphate (0.10 mol/l). The reaction vessel was heated for 30 min at 1700C, cooled and the mixture diluted with up to 500 μl water. The radioiodinated product was separated from unreacted starting materials and radioactive impurities by HPLC. Generally, 3/4- IPA-124, 3/4-IPA-125 and m/p-IPA-131 were obtained in 88 ± 10% radiochemical yield, with a specific activity > 500 GBq / μmol. The fraction containing the radioiodinated products was collected into a sterile tube, buffered with 0.5 M phosphate buffered saline (pH 7.0; Braun, Melsungen, Germany), and sterile filtered through a 0.22 μm sterile membrane (Millex GS, Millipore, Molsheim, France) to an isotonic and injectable radiopharmaceutical for in vitro and in vivo investigations.
Example 3:
Synthesis of 3,4-r124niodo-L-phenylalanine (m, p-IPA-124), 3,4-r125πiodo-L- phenylalanine (m,p-IPA-125). 3.4-r131πiodo-L-phenylalanine (m.p-IPA-13P). and 3.4- f211llastatine-L-phenylalanine (m, p-AtPA-211) by iodo-demetalation Alternatively (m, p)-IPA-124, (m, p)-IPA-125, (m, p)-IPA-131 , and (m, p)-AtPA-211 were prepared by iodo-demetalation in acidic condition in the presence of chloramine- T for in situ oxidation of the corresponding radioiodide and astatine-211 , using the protected tributyltin- or trialkylsilyl-L-phenylalanine as starting material. In details: Sodium [124/125/131l]iodide (up to 2 GBq in 50 μl PBS) was added to a mixture consisting of protected (m,p)-tert. tributyltin- or trialkylsilyl-L-phenylalanine (50 - 80 μg) in 50 μl of methanol and 10 μl of 1 N HCI in a 1 ml-vial, followed by 5 μl aqueous chloramine-T (CAT) from a solution of 1 mg CAT / ml water, while shaking. After a 2 min reaction time at room temperature, 20 μl aqueous Na2S2O5 (4.0 mg Na2S2O5ZmI) was added to the reaction vessel to bind the unreacted free radioisotop, followed by deprotection of the amino and carbonyl groups under acidic condition at 100 0C. (m, p)-IPA-124, (m, p)-IPA-125, (m, p)-IPA-131 , and (m, p)-AtPA-211 were isolated by HPLC and formulated as described above for in vitro and in vivo studies. The radiochemical yields were > 90 %, with a specific activity > 1000 GBq / μmol.
Example 4: Synthesis of 3.4-r82Br1bromo-L-phenylalanine (m. p-BrPA-82) and 3.4-f77Br1bromo-L- phenylalanine (m,p-BrPA-77)
(m, p)-BrPA-82 and (m, p)-BrPA-77 were prepared either by non-isotopic [77 ' 82Br]bromo-deiodination at 160 0C or by [77 ' 82Br]bromo-demetalation, using (m,p)- iodo-L-phenylalanine or the corresponding (m,p)-tributyltin- or (m, p)-trialkylsilyl-L- phenylalanine, as starting materials in analogy to the procedure described above, (m, p)-BrPA-82 and (m, p)-BrPA-77 were isolated by means of HPLC as no carrier-added products with a specific activity > 500 GBq / μmol. The fractions containing the radiobrominated products were buffered with phosphate buffered saline (PBS), and sterile filtered through a 0.22 μm sterile membrane to an isotonic and injectable radiopharmaceuticals for in vitro and in vivo investigations.
Example 5: Cell lines and cell cultures
Two human prostate cancer cell lines, two human breast cancer cell lines, and two melanoma cell line were investigated. The human prostate cancer cells PC3 and DU145 (American Type Culture Collection, Rockville, MD), the human breast SK-BR-3 and BT-474 cancer cell lines, as well as the melanoma cell lines SK-MEL25 and A101 D were purchased commercially or provided by the oncological research laboratory of the University Medical Center of Saarland (Homburg, Germany). Cells were cultivated in RPMI-1640 medium or in Dulbecco's modified Eagle medium (sodium pyruvate-free, supplemented with L-glucose and pyridoxine), respectively, supplemented with 10 % (v/v) heat-inactivated foetal calf serum (FCS), penicillin (50 U/ml), streptomycin (50 μg/ml), and insulin (50 μg/ml; PromoCell, Heidelberg, Germany). All cells lines were maintained in appropriate flasks in a humidified incubator (5% CO2) at 37°C. Before the experiment, subconfluent cell cultures were trypsinized with a solution of 0.05% trypsin containing 0.02% EDTA but without Ca2+ and Mg2+, and resuspended in fresh medium to various cell concentrations after counting by vital staining on a hemocytometer, depending upon the study. Cells were free of mycoplasms. Viability of the cells was > 95 %.
Example 6:
Example of internalisation experiments Uptake experiments were undertaken to evaluate the affinity of the proposed L- phenylalanine derivatives for the proposed human tumors, and to assess their therapeutic activity in vitro.
All experiments were performed fourfold, simultaneously with 250000, 500000 and 106 freshly prepared human tumor cells, including human malignant glioma cells, pancreatic prostatic and breast cancer cells. Before experiments, subconfluent cells were trypsinized as described above. The suspension was mixed thoroughly, transferred to a 50-ml centrifuge tube (Falcon®, Becton Dickinson, USA). Cells were centrifuged for 5 min at 200 x g; the resulting supernatant was removed and the pellet resuspended in serum-free Dulbecco's Modified Eagle medium and then transferred to Eppendorf tubes at concentrations of 106 cells/ml for the uptake investigations. Before incubation with the corresponding radiolabeled phenylalanine, the tumor cells were preincubated for 5 min in 500 μL medium at 37°C in 1.5-ml Eppendorf centrifuge tubes. Aliquots of 30-50 μL (106 - 1.5 x 106 cpm) freshly prepared radiopharmaceutical were added and cells incubated at 37°C / 5% CO2 for 1 , 2, 5, 15, 30, 60, 90 and 120 min while shaking. Uptake was stopped with 500 μL ice-cold PBS (pH 7.4) and an additional 3-min in an ice bath, the cells were centrifuged for 2 min at 300 x g, the supernatant removed and the pellet washed three time with ice-cold PBS. Cell pellets were counted for radioactivity together with 3 aliquots of standards on a Berthold LB951 counter. The percentage of binding of the radiopharmaceutical was calculated by the formula: (cpm cell pellet/mean cpm radioactive standards) x 100. The results were expressed either as percent of the applied dose per 106 cells or as cpm/1000 cells for better comparison.
Example 7:
Evaluation of the cell survival rate after treatment with 3/4-bromo-L-phenylalanine and
3/4-iodo-L-phenylalanine
After development of a confluent lawn of cells, the cultures were exposed to 0.1 to 5 μmol/ml of the corresponding pharmaceutical for up to 48 hours at 37°C/5% CO2. In a parallel experiment, cells were irradiated using a 6-MeV linear accelerator with doses from 2 to 15 Gy or treated with IPA-131 or BrPA-79/81 for comparison of cell survival rate. In order to be able to observe the morphology of the glioma cells, the cells were grown on standard glass slides or in standard culture dishes. Then the medium was removed and the cells were fixed either in 70% ethanol for at least 30 min on ice for flow-cytometric analyses after staining or in 4 % neutral buffered formalin for immunohistopathological analysis. Example 8:
Results and discussion
In vitro studies
The cytostatic effect and radiosensitizing effect of IPA in EGFR positive human tumor cells is demonstrated in figures 1 and 2. As shown, the cytostatic effect of IPA-131 on EGFR positiv tumor cells was more pronounced as compared to external irradiation up to 15 Gy (Fig. 1). Combining IPA-131 with external irradiation led to a dramatic reduction of the cell survival rate (Fig. 2). Flow-cytometric analyses of stained cells show dose dependent induction of primary necrosis and apoptosis, which was more significant than that caused by external irradiation, even with 15 Gy, and more pronounced with increasing IPA-concentration. This result attests the high radiosensitizing effect of IPA-131 on EGFR-positive cells. The surviving cells contained only sparse cytoplasm, the nuclei were shrunken and contained condensed or clumped chromatin. Cytologically, the mode of cell death was apoptosis as the remaining tumour cells contained only sparsely cytoplasm and apoptotic bodies, in other cells the nuclei were shrunken and contained condensed chromatin. Identical results were obtained with BrPA-77 in prostate and breast cells in vitro. This results suggests the high therapeutic potential of 3/4-halogenated L-phenylalanine hormone-independent or metastatic human tumors, especially for hormone- independent prostate and metastatic breast cancer.
The results demonstrate that meta- and para-halogenated-L-phenylalanins represent a new class of therapeutic agents for tumour therapy in these tumor entities.
Reference Example Figure 3 shows an example of uptake kinetic of 4-iodo-L-phenylalanine in primary human androgen-independent tumor cells. The radiolabeled derivative 4-[123l]iodo-L- phenylalanine was used to facilitate quantification, using a gamma counter. As shown, IPA exhibit high uptake in human tumor cells with a continuous increase over the investigation time. This result provides evidence of the high affinity of the proposed radiopharmaceuticals for human tumors, including the human malignant gliomas, pancreatic carcinomas, prostate and breast cancer. References:
1. Meng S, Tripathy D1 Frenkel EP, et al. Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res 2004; 10: 8152 - 8162.
2. Crawford ED. Epidemiology of prostate cancer. Urology 2003; 62: 3 - 12. 3. Parkin DM, Bray Fl, Devesa SS. Cancer burden in the year 2000. The global picture. Eur J Cancer 2001 ; 37: S4 - 66.
4. American Cancer Society: Cancer Facts & Figures 2003; Atlanta, GA: American Cancer Society, 2003.
5. Edwards BK, Brown ML, Wingo PA, et al. Annual report to the nation on the status of cancer, 1975-2002, featuring population-based trends in cancer treatment. J
Natl Cancer Inst 2005; 97: 1407-27.
6. Aquilina JW, Lipsky JJ, Bostwick DG. Androgen deprivation as a strategy for prostate cancer chemoprevention. J Natl Cancer Inst 1997; 89: 689-696.
7. Mimeault M, Pommery N, Henichart JP. New advances on prostate carcinogenesis and therapies: Involvement of EGF-EGFR transduction system. Growth Factors
2003; 21: 1 - 14.
8. Sternberg CN. Systemic treatment and new developments in advanced prostate cancer. Eur J Cancer 2001 ; 37: S147-S157.
9. Sternberg CN. What's new in the treatment of advanced prostate cancer. Eur J Cancer 2003; 39: 136-146. l O. Dorval M, Maunsell E, Deschenes L, et al. Long-term quality of life after breast cancer: Comparison of 8-year survivors with population controls. J Clin Oncol 1998; 16: 487-494.
H . Ganz PA, Desmond KA, Leedham B, et al. Quality of life in long-term, disease-free survivors of breast cancer: A follow-up study. J Natl Cancer Inst 2002; 94: 39 - 49.
12. Langley RE, Carmichael J, Jones AL, et al. Phase III trial of Epirubicin plus Paclitaxel compared with Epirubicin plus Cyclophosphamide as first-line chemotherapy for metastatic breast cancer: United Kingdom National Cancer Research Institute Trial AB01. J Clin Oncol 2005; 33: 8322- 8330. 13. Schindlbeck C, Janni W, Shabani N, et al. Comparative analysis between the HER2 status in primary breast cancer tissue and the detection of isolated tumor cells in the bone marrow. Breast Cancer Res Treat 2004; 87: 65-74.
14. Naumov GN, Chambers AF. Ineffectiveness of doxorubicin treatment on solitary dormant mammary carcinoma cells or late-developing metastases. Breast Cancer Res Treat 2003; 82: 199-206.
15. Samnick S, Richter S, Romeike BF, Heimann A, Feiden W, Kempski O, Kirsch CM: Investigation of iodine-123-labelled amino acid derivatives for imaging cerebral gliomas: uptake in human glioma cells and evaluation in stereotactically implanted C6 glioma rats. Eur J Nucl Med 27: 1543-1551 , 2000. 16. S. Samnick, D. Hellwig, B.F. Romeike, J.-R. Moringlane, W. Feiden and C-M. Kirsch. Initial Evaluation on the feasibility of single photon emission tomography with L-p-[123l]lodophenylalanine for routinely brain tumor imaging. Nucl Med Commun 2002; 23: 121-130.
17. Hellwig D, Ketter R, Romeike BF, Sell N, Schaefer A, Moringlane JR, Kirsch CM, Samnick S: Validation of brain tumour imaging with p-[123l]iodo-L-phenylalanine and SPECT. Eur J Nucl Med MoI Imaging 32: 1041-1049, 2005.
18. Bigner SH, Mark J, Burger PC, et al. Specific chromosome abnormalities in malignant human gliomas. Cancer Res 1988; 48: 405 - 411.
19. Henn W, Blin N, Zang KD. Polysomy of chromosome # 7 is correlated with overexpression of the erbB oncogene in human glioblastoma cell lines. Hum Genet 1986; 74: 104 - 106.
2O.Jager PL, Vaalburg W, Pruim J, de Vries EG, Langen KJ, Piers DA. Radiolabeled amino acids: basic aspects and clinical applications in oncology. J Nucl Med 2001 ; 42: 432-445.
21. l_averman P, Boerman OC, Corstens FH, Oyen WJ. Fluorinated amino acids for tumour imaging with positron emission tomography. Eur J Nucl Med MoI Imaging
2002; 29: 681-690.
22. Dil_orenzo G, Tortora G, D' Armiento FP, et al. Expression of epidermal growth factor receptor correlates with disease relapse and progression to androgen- independence in human prostate cancer. Clin Cancer Res 2002; 8: 3438 - 3444. 23. Tan M, Yao J, Yu D, et al. Overexpression of the c-erbB-2 gene enhanced intrinsic metastasis potential in human breast cancer cells without increasing their transformation abilities. Cancer Res 1997; 57: 1199- 1205.
24. Mishrat SK, Mazumhar A, Vadlamudi RK, et al. MICoA, a novel metastasis- associated protein 1 (MTA1) interacting protein coactivator, regulates estrogen receptor-α transaction functions. J Biol Chem 2003; 278: 19209 - 19219.
25. Herynk MH, Fuqua SA. Estrogen receptor mutation in human disease. Endocr Rev 2004; 25: 869-898.
26. Emrich JG, Brady LW, Quang TS, et al. Radioiodinated (1-125) monoclonal antibody 425 in the treatment of high grade glioma patients: ten-year synopsis of a novel treatment. Am J Clin Oncol. 2002; 25(6):541-546.
27.Zalutsky MR, Bigner DD: Radioimmunotherapy with alpha-particle emitting radioimmunoconjugates. Acta Oncol 35: 373-379, 1996.
28. Hofer KG, Keough G, Smith JM. Biological toxicity of Auger emitters: molecular fragmentation versus electron irradiation. Curr Top Radiat Res Quar 12: 335-354, 1977

Claims

1. Use of a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 for the preparation of a pharmaceutical composition for the treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma, wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma.
2. The use according to claim 1 , wherein the conjugated L-phenylalanine is to be administered intravenously.
3. The use according to claim 1 or 2, wherein the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L-phenylalanine is in the range of 0.1 to 1000 MBq/kg body weight.
4. The use according to anyone of claims 1 to 3, wherein the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L- phenylalanine is to be administered as a single dose once or as a fractionated dose in 2 to 60 fraction doses.
5 The use according to anyone of claims 1 to 4, wherein the conjugated L- phenylalanine is 4-[131l]iodo-L-phenylalanine (IPA-131), 4-[124l]iodo-L- phenylalanine (IPA-124) or 4-[211At]astatine-L-phenylalanine (AtPA-211).
6. The use according to anyone of claims 1 to 5, wherein the pharmaceutical composition further comprises a chemo therapeutic, an immunotherapeutic, a gene therapeutic, a vaccine, an antisense nucleotide therapeutic, an siRNA therapeutic agent, a further endoradiotherapeutic agent and/or a further radiosensitising agent.
7. The use according to anyone of claims 1 to 6, wherein the pharmaceutical composition is to be administered to a patient who is subsequently to be irradiated percutaneously.
8. A method for the monitoring of the progress of a treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma, wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma, the method comprising the step of localizing and/or dosimetrically measuring a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine- 76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine- 211 in the subject by using a γ-camera subsequent to the administration of a pharmaceutical composition comprising said conjugate.
9. The method according to claim 8, wherein the conjugated L-phenylalanine is localized and/or dosimetrically measured at least 0 to 7 days, preferably 0.5 to 48 h subsequent to the administration of the pharmaceutical composition.
10. A method for the treatment of hormone dependent carcinoma and hormone- refractory or metastasized carcinoma derived from hormone dependent carcinoma, wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma, the method comprising the step of administering a L-phenylalanine conjugated to an alpha-, beta- or Auger-electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine-211 to a subject in the need thereof.
11. The method according to claim 10, wherein the conjugated L-phenylalanine is administered intravenously.
12. The method according to claim 10, wherein the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L-phenylalanine is in the range of 0.1 to 1000 MBq/kg body weight.
13. The method according to claim 10, wherein the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L-phenylalanine is administered as a single dose once or as a fractionated dose in 2 to 60 fraction doses.
12. The method according to claim 10, wherein the conjugated L-phenylalanine is 4-[131l]iodo-L-phenylalanine (IPA-131), 4-[124l]iodo-L-phenylalanine (IPA-124) or 4-[211At]astatine-L-phenylalanine (AtPA-211).
13. The method according to claim 10, further comprising the step of a treatment of the subject by a concomitant therapy selected from the group consisting of a surgical therapy, a chemotherapy, an endo- or exoradiotherapy, an immunotherapy, a gene therapy, a vaccine therapy, an antisense nucleotide therapy, an siRNA therapy, an intracavitary therapy, a radiosensitizer therapy or a device-based treatment.
14. The method according to claim 13, wherein the concomitant therapy is an exoradiotherapy wherein the irradiation is started in a period of 0 to 7 days subsequent to the administration of the conjugated L-phenylalanine.
15. The method according to claim 13, wherein the concomitant therapy is an endoradiotherapy comprising the implantation of a radioactive device.
16. The method according to claim 10, wherein the subject is a human subject.
17. A method for the monitoring of the progress of a treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma, wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma, the method comprising the steps:
(a) administering a L-phenylalanine conjugated to an alpha-, beta- or Auger- electron emitting isotope selected from the group consisting of bromine-76, bromine-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine- 211 to a subject in the need thereof; and
(b) localizing and/or dosimetrically measuring the conjugated L-phenylalanine in the subject by using a γ-camera.
18. The method according to claim 17, wherein the conjugated L-phenylalanine is localized and/or dosimetrically measured at least 0 to 7 days, preferably 0.5 to 48 h subsequent to its administration
12. The method according to claim 10, wherein the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L-phenylalanine is in the range of 0.1 to 1000 MBq/kg body weight.
13. The method according to claim 10, wherein the irradiation dose of the alpha-, beta- or Auger-electron emitting isotope conjugated to the L-phenylalanine is administered as a single dose once or as a fractionated dose in 2 to 60 fraction doses.
14. The method according to claim 10, wherein the conjugated L-phenylalanine is 4-[131l]iodo-L-phenylalanine (IPA-131), 4-[124l]iodo-L-phenylalanine (IPA-124) or 4-[211At]astatine-L-phenylalanine (AtPA-211).
15. The method according to claim 10, further comprising the step of a treatment of the subject by a concomitant therapy selected from the group consisting of a surgical therapy, a chemotherapy, an endo- or exoradiotherapy, an immunotherapy, a gene therapy, a vaccine therapy, an antisense nucleotide therapy, an siRNA therapy, an intracavitary therapy, a radiosensitizer therapy or a device-based treatment.
16. The method according to claim 15, wherein the concomitant therapy is an exoradiotherapy wherein the irradiation is started in a period of 0 to 7 days subsequent to the administration of the conjugated L-phenylalanine.
17. The method according to claim 15, wherein the concomitant therapy is an endoradiotherapy comprising the implantation of a radioactive device.
18. The method according to claim 10, wherein the subject is a human subject.
19. A method for the monitoring of the progress of a treatment of hormone dependent carcinoma and hormone-refractory or metastasized carcinoma derived from hormone dependent carcinoma, wherein the hormone dependent carcinoma is selected from the group consisting of prostate carcinoma and mamma carcinoma, the method comprising the steps:
(a) administering a L-phenylalanine conjugated to an alpha-, beta- or Auger- electron emitting isotope selected from the group consisting of bromine-76, bromiπe-77, bromine-82, iodine-124, iodine-125, iodine-131 and astatine- 211 to a subject in the need thereof; and
(b) localizing and/or dosimetrically measuring the conjugated L-phenylalanine in the subject by using a γ-camera.
20. The method according to claim 19, wherein the conjugated L-phenylalanine is localized and/or dosimetrically measured at least 0 to 7 days, preferably 0.5 to 48 h subsequent to its administration
RECTIFIED SHEET (RULE 91) ISA/EP
PCT/EP2006/011368 2005-11-25 2006-11-27 Use of l-phenylalanine conjugated to an emitting isotope for therapy of hormone dependent carcinoma Ceased WO2007060012A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP05025775 2005-11-25
EP05025775.7 2005-11-25

Publications (2)

Publication Number Publication Date
WO2007060012A2 true WO2007060012A2 (en) 2007-05-31
WO2007060012A3 WO2007060012A3 (en) 2007-08-09

Family

ID=37806108

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2006/011368 Ceased WO2007060012A2 (en) 2005-11-25 2006-11-27 Use of l-phenylalanine conjugated to an emitting isotope for therapy of hormone dependent carcinoma

Country Status (1)

Country Link
WO (1) WO2007060012A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011061154A1 (en) 2009-11-17 2011-05-26 Bayer Schering Pharma Aktiengesellschaft Iodine-labeled homoglutamic acid and glutamic acid derivatives
EP3663307A4 (en) * 2017-08-04 2021-03-24 Osaka University PROCESS FOR THE PRODUCTION OF A RADIO-LABEL ARYL COMPOUND

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; August 2003 (2003-08), SAMNICK S ET AL: "Establishment of an in vivo model of human pancreatic tumor for preclinical studies and evaluation of radioiodinated phenylalanine-analogues as radiopharmaceuticals to image pancreatic carcinomas." XP009084578 Database accession no. PREV200300483386 & JOURNAL OF LABELLED COMPOUNDS AND RADIOPHARMACEUTICALS, vol. 46, no. Supplement 1, August 2003 (2003-08), page S78, 15TH INTERNATIONAL SYMPOSIUM ON RADIOPHARMACEUTICAL CHEMISTRY; SYDNEY, AUSTRALIA; AUGUST 10-14, 2003 ISSN: 0362-4803 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; November 2004 (2004-11), ROMEIKE BERND F M ET AL: "Action and efficacy of p-[I-131]iodo-L-phenylalanine on primary human glioma cell cultures and rats with C6-gliomas" XP009084572 Database accession no. PREV200510311779 & ANTICANCER RESEARCH, vol. 24, no. 6, November 2004 (2004-11), pages 3971-3976, ISSN: 0250-7005 *
DATABASE EMBASE [Online] ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL; 2006, KERSEMANS V ET AL: "Optimization by experimental design of precursor synthesis and radiolabeling of 2-iodo-L-phenylalanine, a novel amino acid for tumor imaging" XP002436016 Database accession no. EMB-2006355082 & CANCER BIOTHERAPY AND RADIOPHARMACEUTICALS 2006 UNITED STATES, vol. 21, no. 3, 2006, pages 235-242, ISSN: 1084-9785 *
DATABASE MEDLINE [Online] US NATIONAL LIBRARY OF MEDICINE (NLM), BETHESDA, MD, US; August 2006 (2006-08), KERSEMANS VEERLE ET AL: "123/125I-labelled 2-iodo-L: -phenylalanine and 2-iodo-D: phenylalanine: comparative uptake in various tumour types and biodistribution in mice." XP019422319 Database accession no. NLM16572305 & EUROPEAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING AUG 2006, vol. 33, no. 8, August 2006 (2006-08), pages 919-927, ISSN: 1619-7070 *
FUKUSHIMA K ET AL: "ANTI TUMOR ACTIVITY OF AMINO-ACID DERIVATIVES IN THE PRIMARY SCREENING" GANN, JAPANESE CANCER ASSOCIATION, TOKYO, JP, vol. 66, no. 1, 1975, pages 29-36, XP009079453 ISSN: 0016-450X *
HELLWIG D ET AL: "Radioiodinated phenylalanine derivatives to image pancreatic cancer: a comparative study with [<18>F]fluoro-2-deoxy-d-glucose in human pancreatic carcinoma xenografts and in inflammation models" NUCLEAR MEDICINE AND BIOLOGY, ELSEVIER, NY, US, vol. 32, no. 2, February 2005 (2005-02), pages 137-145, XP004756711 ISSN: 0969-8051 *
MERTENS J ET AL: "Synthesis, radiosynthesis, and in vitro characterization of [<125>I]-2- iodo-L-phenylalanine in a R1M rhabdomyosarcoma cell model as a new potential tumor tracer for SPECT" NUCLEAR MEDICINE AND BIOLOGY, ELSEVIER, NY, US, vol. 31, no. 6, August 2004 (2004-08), pages 739-746, XP004674814 ISSN: 0969-8051 *
SAMNICK ET AL: "Preparation and tumor affinity testing of the radioiodinated tetrahydroisoquinoline derivative [<123>I]TIC(OH) for targeting prostate cancer" APPLIED RADIATION AND ISOTOPES, ELSEVIER, OXFORD, GB, vol. 64, no. 5, May 2006 (2006-05), pages 563-569, XP005331227 ISSN: 0969-8043 *
SAMUEL SAMNICK ET AL: "P-[123ÜIODO-L-PHENYLALANINE FOR DETECTION OF PANCREATIC CANCER:BASIC INVESTIGATIONS OF THE UPTAKE CHARACTERISTICS IN PRIMARY HUMAN PANCREATIC TUMOUR CELLS AND EVALUATION IN IN VIVO MODELS OF HUMAN PANCREATIC ADENOCARCINOMA" EUROPEAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING, SPRINGER VERLAG, HEIDELBERG, DE, vol. 31, no. 4, 14 January 2004 (2004-01-14), pages 532-541, XP008078228 ISSN: 1619-7070 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011061154A1 (en) 2009-11-17 2011-05-26 Bayer Schering Pharma Aktiengesellschaft Iodine-labeled homoglutamic acid and glutamic acid derivatives
JP2013510894A (en) * 2009-11-17 2013-03-28 バイエル・ファルマ・アクチェンゲゼルシャフト Iodine-labeled homoglutamate and glutamate
EP3663307A4 (en) * 2017-08-04 2021-03-24 Osaka University PROCESS FOR THE PRODUCTION OF A RADIO-LABEL ARYL COMPOUND
US11731917B2 (en) 2017-08-04 2023-08-22 Osaka University Production method for radiolabeled aryl compound
US12421179B2 (en) 2017-08-04 2025-09-23 Osaka University Production method for radiolabeled aryl compound

Also Published As

Publication number Publication date
WO2007060012A3 (en) 2007-08-09

Similar Documents

Publication Publication Date Title
KR102033819B1 (en) Method for upregulating antigen expression
Ma et al. In vitro and in vivo evaluation of 211At-labeled fibroblast activation protein inhibitor for glioma treatment
Robinson et al. Effective treatment of established human breast tumor xenografts in immunodeficient mice with a single dose of the α-emitting radioisotope astatine-211 conjugated to anti-HER2/neu diabodies
HK1256497A1 (en) Combination therapy comprising anti-cd20 antibody and the radiolabeled hh1 monoclonal antibody
RU2741794C2 (en) Radiopharmaceutical solutions with preferred properties
Mohajershojai et al. In Vitro characterization of 177Lu-DOTA-M5A anti-carcinoembryonic Antigen Humanized antibody and HSP90 inhibition for Potentiated Radioimmunotherapy of Colorectal Cancer
Fathi et al. Demonstration of dose dependent cytotoxic activity in SW480 colon cancer cells by 177Lu-labeled siRNA targeting IGF-1R
WO2007060012A2 (en) Use of l-phenylalanine conjugated to an emitting isotope for therapy of hormone dependent carcinoma
Fathi et al. Synthesis of a stabilized 177Lu–siRNA complex and evaluation of its stability and RNAi activity
WO2007079953A2 (en) In vitro method to predict the tumor sensitivity to pharmacotherapy and endoradiotherapy
US20070128108A1 (en) Therapy of malignant neoplasias
EP1962822B1 (en) Therapy of malignant neoplasias
Berckmans et al. Targeted Alpha Therapy of Ovarian Cancer with ²¹²Pb-Labeled Anti-PTK7 Antibody: On-site ²¹²Pb Production and Preclinical Insights
Deshayes et al. Anti-MISRII radiolabeled antibodies: new tools for a theranostic approach in ovarian cancer
EP1656160B1 (en) Radioimmunoconjugates for targeted alpha therapy
Lee et al. Preparation and Bioevaluation of 177Lu-labelled Anti-CD44 for Radioimmunotherapy of Colon Cancer
HK1217438B (en) Combination therapy comprising rituximab and the radiolabeled hh1 monoclonal antibody

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 06818856

Country of ref document: EP

Kind code of ref document: A2