US20150265594A1 - Combination of opioids and anticancer drugs for cancer treatment - Google Patents

Combination of opioids and anticancer drugs for cancer treatment Download PDF

Info

Publication number
US20150265594A1
US20150265594A1 US14/434,138 US201314434138A US2015265594A1 US 20150265594 A1 US20150265594 A1 US 20150265594A1 US 201314434138 A US201314434138 A US 201314434138A US 2015265594 A1 US2015265594 A1 US 2015265594A1
Authority
US
United States
Prior art keywords
methadone
cells
doxorubicin
cancer
opioid receptor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/434,138
Other languages
English (en)
Inventor
Claudia Friesen
Erich Miltner
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.)
Universitaet Ulm
Original Assignee
Universitaet Ulm
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 Universitaet Ulm filed Critical Universitaet Ulm
Assigned to UNIVERSITÄT ULM reassignment UNIVERSITÄT ULM ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: Friesen, Claudia, Dr., MILTNER, ERICH, DR.
Publication of US20150265594A1 publication Critical patent/US20150265594A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/485Morphinan derivatives, e.g. morphine, codeine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4468Non condensed piperidines, e.g. piperocaine having a nitrogen directly attached in position 4, e.g. clebopride, fentanyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5014Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing toxicity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the invention relates to novel strategies for the treatment of cancer patients based on a combination of an opioid receptor agonist and an anticancer compound.
  • Cancer can be defined as an abnormal growth of tissue characterized by a loss of cellular differentiation. This term encompasses a large group of diseases in which there is an invasive spread of undifferentiated cells from a primary site to other parts of the body where further undifferentiated cellular replication occurs, which eventually interferes the normal functioning of tissues and organs.
  • Cancers are primarily an environmental disease with 90-95% of cases attributed to environmental factors and 5-10% due to genetics.
  • Environmental as used by cancer researchers, means any cause that is not inherited genetically, not merely pollution.
  • Common environmental factors that contribute to cancer death include tobacco (25-30%), diet and obesity (30-35%), infections (15-20%), radiation (both ionizing and non-ionizing, up to 10%), stress, lack of physical activity, and environmental pollutants.
  • cancer With more than 3 million new cases and 1.7 million deaths each year, cancer represents the second most important cause of death and morbidity in Europe. On a global scale, cancer accounted for 7.4 million deaths (around 13% of the total) in 2004.
  • cancer is a leading cause of death, causing 20% of the total in the European Region. Noticeably, Europe comprising only one eighth of the total world population but has around one quarter of the global total of cancer cases: some 3.2 million new patients per year.
  • the most common forms of cancer were prostate, colorectal, breast, leukemia and lung cancer.
  • the risk of getting cancer before the age of 75 years is 26.5%, or around one in four.
  • the rate of new cases of cancer is also expected to increase.
  • Each cancer is characterized by the site, nature, and clinical cause of undifferentiated cellular proliferation, whereby the underlying mechanism for the initiation of cancer is not completely understood.
  • Cancer is usually treated with chemotherapy, radiation therapy and surgery.
  • Chemotherapy in addition to surgery has proven useful in a number of different cancer types including: breast cancer, colorectal cancer, pancreatic cancer, osteogenic sarcoma, testicular cancer, ovarian cancer, and certain lung cancers.
  • Radiation therapy involves the use of ionizing radiation in an attempt to either cure or improve the symptoms of cancer. It is used in about half of all cases and the radiation can be from either internal sources in the form of brachytherapy or external sources. Radiation is typically used in addition to surgery and or chemotherapy but for certain types of cancer such as early head and neck cancer may be used alone. For painful bone metastasis it has been found to be effective in about 70% of people.
  • anticancer therapies are frequently ineffective due to resistance of the tumour cells to radio- and/or chemotherapy.
  • This objective is solved by using a combination of opioid receptor agonists and anticancer drugs for the treatment of cancer wherein this combination is given in a specific administration scheme according to claim 1 of the invention.
  • the invention relates to a combination of an opioid receptor agonist and at least one anticancer agent for use in the treatment of cancer, wherein
  • This combination therapy is based on the unexpected finding that opioid receptor agonists together with anticancer agents kill cancer cells more effectively. Furthermore, the inventors could show that the interaction between opioid receptor agonists and anticancer agents represents a self-reinforcing feedback loop as illustrated by FIG. 26 . In the first path of this loop opioid receptor agonists enhance the cellular uptake and inhibit the efflux of anticancer drugs. In the second path of said loop the accumulating anticancer drugs lead to an increased expression of opioid receptors on the surface of the cancer cell. Hence, both the opioid receptor agonist and the anticancer agent can exert their cytotoxic potential to a higher extent.
  • the invention is based on the unexpected finding that the amount of opioid receptor expressed on the cell surface of cancer cells is varying among the different cancer types and also exhibiting inter-individual differences and that this surface-associated opioid receptor expression can be increased by anticancer agents.
  • anticancer agents for example doxorubicin, idarubicin, epirubicin, daunorubicin, carboplatin, oxaliplatin, cisplatin, etoposide, methotrexate, cytarabine, teniposide, rituximab fludarabine, are able to induce an increase of the number of opioid receptors which are expressed on the cell surface of cancer cells.
  • the combination therapy of opioid receptor agonist and anticancer drug according to the invention can improve cancer therapy by one or more of the following ways:
  • opioid receptor agonist is defined as a chemical heterogeneous group of natural, synthetic or semi-synthetic substances, working agonistic at the same type of receptor, the so called opioid receptor.
  • opioid receptor agonist a chemical heterogeneous group of natural, synthetic or semi-synthetic substances, working agonistic at the same type of receptor, the so called opioid receptor.
  • opioid receptor agonist as used herein comprises full agonists as well as mixed agonists/antagonists or partial agonists such as buprenorphine.
  • the group of opioids includes natural opiates such as alkaloids like morphine or dihydrocodeine, as well as semi-synthetic opiates, derived from the natural opiates (e.g. hydromorphone or oxycodone), or fully synthetic opioids, such as fentanyl or buprenorphine. It also includes endogenous opioid peptides, which may be produced naturally in the body as endorphins, dynorphins or enkephalins but which can also be synthesized.
  • anticancer drug encompasses all chemical or physical interventions that are used for the treatment of cancer. It therefore includes chemotherapeutical agents such as cytotoxic agents or immunotoxic agents but also radioactively labelled antibodies, peptides and chemical substances, which might emit alpha, beta and gamma rays as well as electrons.
  • the radiotherapy further includes photons of sufficiently high energy, charged particles such as electrons, positrons, muons, protons, alpha particles, and heavy atomic nuclei from accelerators, but also neutrons and gamma rays.
  • therapeutically effective plasma level is defined as a plasma level that is between the plasma level of the drug that causes a lethal effect and the minimum plasma level that causes a therapeutic effect.
  • therapeutic effect of the opioid receptor agonist is given by the increase in cellular uptake and/or the inhibition of the cellular efflux of the co-administered anticancer drug and/or the induction of cell death by e.g. apoptosis, necrosis, mitotic catastrophe and autophagy.
  • the therapeutic effect of the anticancer drug is given by its ability to kill cancer cells and/or to induce the opioid receptor expression on the cancer cells.
  • the words “treat,” “treating” or “treatment” refer to using the combination of the present invention or any composition comprising them to either prophylactically prevent a cancer, or to mitigate, ameliorate or stop cancer. They encompass either curing or healing as well as mitigation, remission or prevention, unless otherwise explicitly mentioned.
  • the word “patient” refers to a mammal, including a human.
  • the treatment specifically refers to the inhibition of cancer cell proliferation and/or growth.
  • This activity can include e.g. cytostatic or cytotoxic activity as well arresting growth of cells and/or tumours.
  • Cancer cell proliferation is the result of the inhibition of cell division.
  • opioid receptor agonists induce cell death in tumours.
  • Cell death in the context of the invention includes all types of cells death. This can include necrotic as well as apoptotic cell death or autophagy.
  • the cell death is induced by the activation of the caspases-dependent or caspases-independent pathway.
  • opioid receptor agonists can induce cell death via various pathways.
  • opioid receptor agonists induce apoptosis in cancer cells.
  • cancer which is synonymously used to the term “neoplasm” refers to diseases in which abnormal cells divide without control and are able to invade other tissues. Cancer cells can spread to other parts of the body through the blood and lymph systems.
  • the term “conventional therapy time” in the context of the present invention is defined as the time in a conventional therapy where an anticancer agent is applied to a patient without an opioid receptor agonist according the invention.
  • the therapy time starts with the first application of the anticancer agent, and may include iteration-periods which are specific for cancer and anticancer agent (for example application of a dose two times a day for a week than a pause of three weeks and then again application of a dose two times a day for a week followed by a pause of three weeks), up to time point, at which the anticancer agent is below the therapeutic plasma level of the patient.
  • Cancer and its different types in the context of the present invention can be classified by the ICD-O Standard which is a specialised classification of the ICD-10 Standard Classes C00-C97 and D00-D36.
  • the classification of Boecker et al. 2008 in chapter 6 can be used.
  • said opioid receptor agonist is capable of inhibiting cell proliferation.
  • said anticancer agent and said opioid receptor agonist are administered simultaneously or successively.
  • the period of the therapeutically effective plasma levels of the anticancer agent is completely within the respective period of the opioid receptor agonist.
  • the opioid receptor agonist is given in a way that the patient develops a habituation against said opioid receptor agonist. It is thus preferable to wait with the anticancer treatment until the habituation period has begun or even reaches a plateau.
  • the habituation can be a result of a decreased drug efficacy and/or a decrease in side effects such as respiratory depression.
  • Side effects of opioid receptor agonist are hypotension, respiratory depression, vomiting, constipation, dizziness, sedation, euphoria and cardiac effects. This side effects have to be taken in account for determine the therapy scheme with the opioid receptor agonist and the cancer agent. This means that the opioid receptor agonist is given at a starting dose on a very low level i.e.
  • the administration regimen and thus the period within a therapeutically effective plasma level of the anticancer agent is defined by the conventional therapy regimen.
  • the patient treated with the combination according to the invention has received a pre-treatment comprising an anticancer agent.
  • the pre-treatment with the anticancer agent has been discontinued or even terminated.
  • the pre-treatment has been terminated due to resistance against the anticancer treatment.
  • the period with a therapeutically effective plasma level of the anticancer agent lasts for at least 1 day, preferably 3 days, and more preferably for at least 5 days.
  • the period with a therapeutically effective plasma level for the opioid receptor agonist is at least two weeks, more preferably four weeks and even more preferably represents a chronic treatment.
  • chronic treatment is defined as a opioid receptor agonist treatment with an administration period above four weeks, which preferably lasts over several months.
  • this chronic treatment differs from the conventional therapy regimen as prescribed or known to the person skilled in art. or is published in therapeutic guidelines of associations or federations like Deutsche Krebs Vietnamese or Academic Krebsippo; NCCN, NCI or similar health or cancer organizations or the guidelines of producer or distributer of drugs which are used for treatment of cancer
  • the use of at least one anticancer agent refers to the use of one or more anticancer agents to be given in combination with the opioid receptor agonist according the invention.
  • the combination includes the use of one, two, three, four, five or even more anticancer agents.
  • the “death receptor pathway” (or extrinsic pathway) includes the TNF-receptor-induced (tumour necrosis factor) model and the Fas-receptor-induced model (the Fas-receptor is also known as Apo-1 or CD95). Bindings to these receptors result in the formation of death-inducing signalling pathways in the cell, including the activation of caspase-8.
  • the “mitochondrial pathway” (or intrinsic pathway) involves the release of cytochrome c from mitochondria, binding of Apaf-1 and activation of procaspase-9.
  • Several regulators are known to activate or deactivate the apoptosis pathways, such as the pro-apoptotic proteins Bax and Bak or the anti-apoptotic proteins Bcl-2, Bcl- XL or XIAP.
  • the opioid receptor agonists induce apoptosis by cleavage of caspase-3 and poly(ADP-ribose) polymerase (PARP) in the tumour cell, and/or cleavage of caspase-9 and down regulation of X-linked inhibitor of apoptosis protein (XIAP), and/or down regulation of the B-cell lymphoma-extra large protein (Bcl- XL ).
  • PARP poly(ADP-ribose) polymerase
  • XIAP X-linked inhibitor of apoptosis protein
  • Bcl- XL B-cell lymphoma-extra large protein
  • the opioid receptor agonist is a member of the methadone group, comprising DA-methadone, levomethadone, levacetylmethadol and piritramide.
  • metalhadone group relates to opioids which are derivatives of 3,3-diphenylpropylamine. These compounds possess the 3,3-diphenylamine core structure as shown by the following formula (I):
  • R 1 is an aliphatic ketone, a 3-acetoxypropyl residue, a cyano group, or a 1-pyrrolidino-methylketone, —(C ⁇ O)C 2 H 5 , R 2 and R 3 are CH 3 or together forming a heterocyle, preferably a morpholino group, and R 4 is H or an alkyl residue, being preferably CH 3 .
  • a non-limited list of examples for compounds of the methadone group includes methadone, normethadone, dextromoramide, isomethadone, acetylmethadol, alphacetylmethadol, levoacetylmethadol, premethadone, racemoramid, phenadoxone, dextropropoxyphene, dipipanone, benzitramide, piritramide, loperamide, themalon (which represents a 3,3-dithiophenylpropylamine) and levomoramid.
  • the racemic form of DA-methadone is preferably provided in form of a hydrochloride.
  • the opioid methadone induces apoptosis in cancer cells via the mitochondrial pathway.
  • the opioid receptor agonist is selected from the list consisting of compounds of the methadone group such as D/L-methadone, D-methadone, L-methadone, normethadone, fentanyl derivatives such as fentanyl, sufentanyl, lofenantil, alfentanil, remifentanil, ohmefentanyl and carfentanyl; morphinane compounds such as morphine, codeine, heroine, dextrallorphane, dextromethorphan, dextrophanol, dimemorfan, levalorphanol, butorphanol, levofurethylnormorphanol, levomethorphane, levophenacylmorphane, levorphanol, methorphane, morphanol, oxilorphan, phenomorphan, and xorphanol, benzomorphane derivatives such as 5,9-DEHB, alazocine,
  • the opioid receptor agonists of the invention are capable of inhibiting cell proliferation.
  • the opioid receptor agonist is combined with at least one additional opioid receptor agonist.
  • the combination of the invention consists of two, three, four or more opioid receptor agonists.
  • Preferably a combination of two different opioid receptor agonists is used. It could be demonstrated that the combined use of different opioids leads to a synergistic pro-apoptotic effect on cancer cells (see Example 38 and FIG. 56 ).
  • said combination of opioid receptor agonists comprises morphine and fentanyl.
  • said combination consists of morphine and fentanyl.
  • the synergistic effect of morphine and fentanyl was e.g. shown for the leukemia cell line HL60 (see Example 38 and FIG. 56 ).
  • the methadone preferably the D,L-methadone and most preferably the hydrochloride form of D,L-methadone is given to the patient in particular to yield a plasma level which is between 0.05 ⁇ g/mL and 3 ⁇ g/mL.
  • methadone preferably the D,L-methadone and most preferably the hydrochloride form of D,L-methadone is given to the patient in particular to yield a plasma level which is between 0.01 ⁇ g/mL and 3 ⁇ g/mL.
  • the anticancer agent is selected from the list consisting of intercalating substances such as anthracycline doxorubicin, idarubicin, epirubicin, and daunorubicin; topoisomerase inhibitors such as irinotecan, topotecan, camptothecin, lamellarin D, etoposide, teniposide, mitoxantrone, amsacrine, ellipticines and aurintricarboxylic acid; nitrosourea compounds such as carmustine (BCNU), lomustine (CCNU), streptozocin; nitrogen mustards such as cyclophosphamide, mechlorethamine, uramustine, bendamustine, melphalan, chlorambucil, mafosfamide, trofosfamid and ifosfamide; alkyl sulfonates such as busulfan and treosulfan; alkylating agents such as busulfan and
  • anticancer agents comprise also modifications such as PEGylation and formulations such as the use of liposomes (i.e. PEGylated liposomal doxorubicin).
  • the anticancer agent can be a radioactively labeled chemical compound, peptide, protein or monoclonal antibody, wherein the radioactive label could emit alpha, beta or gamma rays and also ionizing particles.
  • the anticancer agent is methotrexate, cytarabine, gemcitabine, paclitaxel, docetaxel, carboplatin, oxaliplatin, etoposide, vincristine, fludarabine especially cisplatin, doxorubicin, anthracycline, idarubicin, daunorubicin, epirubicin, or alpha-, beta-, or gamma irradiation.
  • the patient When treating a cancer entity for which an induction of the opioid receptor is desired the patient is preferably treated with an anticancer agent selected from the group consisting doxorubicin, idarubicin, epirubicin, daunorubicin, carboplatin, oxaliplatin, cisplatin, etoposide, methotrexate, cytarabine, teniposide, gemcitabine, paclitaxel, rituximab and trastuzumab.
  • an anticancer agent selected from the group consisting doxorubicin, idarubicin, epirubicin, daunorubicin, carboplatin, oxaliplatin, cisplatin, etoposide, methotrexate, cytarabine, teniposide, gemcitabine, paclitaxel, rituximab and trastuzumab.
  • the patient who is treated with the combination of the invention suffers from a neoplasm as classified according the International statistical classification of Diseases and related health problems 10 th Revision (ICD-10), wherein the neoplasm is from the group consisting of malignant neoplasms of classes COO to C97, in situ neoplasms of classes DOO to D09, benign neoplasms of classes D10 to D36, and neoplasms of uncertain or unknown behaviour of classes D37 to D48.
  • ICD-10 International statistical classification of Diseases and related health problems 10 th Revision
  • the classes are defined as follows: (C00) Malignant neoplasm of lip, (C01) Malignant neoplasm of base of tongue, (C02) Malignant neoplasm of other and unspecified parts of tongue, (C03) Malignant neoplasm of gum, (C04) Malignant neoplasm of floor of mouth, (C05) Malignant neoplasm of palate, (C06) Malignant neoplasm of other and unspecified parts of mouth, (C07) Malignant neoplasm of parotid gland, (C08) Malignant neoplasm of other and unspecified major salivary glands, (C09) Malignant neoplasm of tonsil, (C10) Malignant neoplasm of oropharynx, (C11) Malignant neoplasm of nasopharynx, (C12) Malignant neoplasm of piriform sinus, (C13) Malignant neoplasm of hypopharynx,
  • the patient who is treated with the combination of the invention suffers from metastases.
  • the patient who is treated with the combination of the invention suffers from a neoplasm selected from list of classes consisting of C25, C50, C56, C71, C91, and C92.
  • the patient suffers from a neoplasm selected from the list comprising of acute lymphoblastic leukemia (C91.0), B-cell chronic lymphatic leukemia (C91.2), acute promyelocytic leukemia (C92.4), acute myeloid leukemia (C92.0) chronic myeloid leukemia (C92.1), all forms of glioblastoma (C71), all forms of pancreatic cancer (C25), all forms of ovarian cancer (C56), classes of breast cancer (C50) and tumour stem cells such as glioblastoma initiating stem cells.
  • C91.0 acute lymphoblastic leukemia
  • C91.2 B-cell chronic lymphatic leukemia
  • acute promyelocytic leukemia C92.4
  • acute myeloid leukemia C92.0 chronic myeloid leukemia
  • C92.1 all forms of glioblastoma
  • C71 all forms of pancreatic cancer
  • C25 all forms of ovarian cancer
  • C56 classes
  • the patient suffers from a breast cancer resistant to HER2-targeted therapies, like e.g. a Trastuzumab resistant breast cancer.
  • the patient suffering from acute lymphoblastic leukemia is treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxe
  • the patient suffering from acute lymphoblastic leukemia is treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from acute lymphoblastic leukemia (C91.0) is treated with the combination comprising D,L-methadone and etoposide or D,L-methadone and doxorubicin.
  • the patient suffering from B-cell chronic lymphatic leukemia is treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docet
  • the patient suffering from B-cell chronic lymphatic leukemia is treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from B-cell chronic lymphatic leukemia (C 91.2) is treated with the combination comprising D,L-methadone and fludarabine, or buprenorphine and fludarabine or fentanyl and fludarabine or morphine and fludarabine.
  • the patient suffering from acute promyelocytic leukemia is treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline, doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline, doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel
  • the patient suffering from acute promyelocytic leukemia is treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from acute promyelocytic leukemia is treated with the combination comprising D,L-methadone and doxorubicin or buprenorphine and doxorubicin or fentanyl and doxorubicin or morphine and doxorubicin.
  • the patient suffering from acute myeloid leukemia is treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline, doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide gemcitabine, paclitaxel, docetaxel or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline, doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide gemcitabine, paclitaxel, docetaxel
  • the patient suffering from acute myeloid leukemia is treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from acute myeloid leukemia is treated with the combination comprising D,L-methadone and doxorubicin or buprenorphine and doxorubicin or fentanyl and doxorubicin or morphine and doxorubicin.
  • the patient suffering from chronic myeloid leukemia is treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel
  • the patient suffering from chronic myeloid leukemia is treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from chronic myeloid leukemia is treated with the combination comprising D,L-methadone and imatinib or buprenorphine and imatinib or fentanyl and imatinib or morphine and imatinib.
  • the patient suffering from chronic myeloid leukemia is treated with the combination comprising D,L-methadone and fludarabine or buprenorphine and fludarabine or fentanyl and fludarabine or morphine and fludarabine.
  • the patient suffering from leukemia is treated with at least one further opioid receptor agonist in addition to the combination of the invention.
  • the patient is treated with at least two opioid receptor agonists.
  • This strategy is based on the finding that the combination of different opioids shows a synergistic pro-apoptotic effect on cancer cell lines (see FIG. 56 ).
  • the combination of the invention comprises morphine, fentanyl and at least one anticancer agent and in a further embodiment the combination consists of morphine, fentanyl and a further anticancer agent.
  • the synergistic effect of morphine and fentanyl was e.g. shown for the leukemia cell line HL60 (see Example 38).
  • the patient suffering from glioblastoma is treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, temozolomide, anthracycline, doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel, or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, temozolomide, anthracycline, doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide
  • the patient suffering from glioblastoma is treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from glioblastoma is treated with the combination comprising D,L-methadone and doxorubicin.
  • said doxorubicin is given in a formulation that enhances the transfer of the doxorubicin across the blood brain barrier.
  • the doxorubicin could be packed into liposomes or bound to transferrin.
  • the patient suffering from glioblastoma is treated with the combination comprising D,L-methadone and daunorubicin, with a combination comprising D,L-methadone and idarubicin, or with a combination comprising D,L-methadone and temozolomide.
  • the patient suffering from glioblastoma initiating stem cells are treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline, doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline, doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxe
  • the patient suffering from glioblastoma initiating stem cells are treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from glioblastoma initiating stem cells are treated with the combination comprising D,L-methadone and doxorubicin.
  • the patient suffering from pancreatic cancer (C25) is treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, oxaliplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, oxaliplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide
  • the patient suffering from pancreatic cancer (C25) is treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from pancreatic cancer (C25) is treated with the combination comprising D,L-methadone and cisplatin.
  • the patient suffering from pancreatic cancer (C25) is treated with the combination comprising D,L-methadone and oxaliplatin.
  • the patient suffering from cancer is treated with a combination comprising D,L-methadone and temozolomide.
  • the patient suffering from ovarian cancer (C56) is treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, carboplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, carboplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine
  • the patient suffering from ovarian cancer (C56) is treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from ovarian cancer (C56) is treated with the combination comprising D,L-methadone and cisplatin.
  • the patient who is treated with the combination of D,L-methadone and cisplatin suffers from a cisplatin resistant ovarian cancer.
  • the patient suffering from breast cancer (C50) is treated with the combination according the invention including an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel or alpha, beta or gamma irradiation.
  • an anticancer agent selected from the list consisting of methotrexate, cytarabine, carboplatin, oxaliplatin, vincristine, fludarabine, being preferably cisplatin, anthracycline doxorubicin, idarubicin, daunorubicin, epirubicin, etoposide, gemcitabine, paclitaxel, docetaxel or
  • the patient suffering from breast cancer (C50) is treated with the combination according the invention including an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • an opioid receptor agonist selected from the list consisting of D,L-methadone, buprenorphine, fentanyl, and morphine, being preferably D,L-methadone.
  • the patient suffering from breast cancer (C50) is treated with the combination comprising D,L-methadone and cisplatin.
  • breast cancer which preferably is a breast cancer resistant to HER2 targeted therapies, such as
  • trasstuzumab resistant breast cancer is treated with the combination comprising D,L-methadone and doxorubicin.
  • the patient suffering from prostate cancer is treated with the combination comprising D,L-methadone and cisplatin.
  • the patient suffering from leukemia is treated with a combination of D,L-methadone and one of the following anticancer agents: etoposide, cytarabine, methotrexate, cyclophosphamide, thioguanine, gemcitabine, paclitaxel, docetaxel or vincristine.
  • D,L-methadone one of the following anticancer agents: etoposide, cytarabine, methotrexate, cyclophosphamide, thioguanine, gemcitabine, paclitaxel, docetaxel or vincristine.
  • the cancer to be treated is a neoplasm according the International classification of Diseases for Oncology ICD-O in the actual version ICD-O-3 from 2000.
  • the cancer to be treated is a cancer as included in the TNM Classification of Malignant Tumours (TNM), which represents a cancer staging system that describes the extent of cancer in a patient's body.
  • TNM Malignant Tumours
  • the cancer to be treated is disclosed by Boecker et al., 2008 in chapter 6 (Pathologie, Elsevier, Urban & Fischer, p. 167-218), which is incorporated by reference in its entirety.
  • the patient that is treated with said combination suffers from non-solid tumours from the group consisting of leukemia, breast cancer, skin cancer, bone cancer, prostate cancer, liver cancer, lung cancer, brain cancer, cancer of the larynx, gallbladder, pancreas, rectum, parathyroid, thyroid, adrenal, neural tissue, head and neck, colon, stomach, bronchi, kidneys, basal cell carcinoma, squamous cell carcinoma of both ulcerating and papillary type, metastatic skin carcinoma, osteosarcoma, Ewing's sarcoma, veticulum cell sarcoma, myeloma, giant cell tumour, small-cell lung tumour, islet cell tumour, primary brain tumour, acute and chronic lymphocytic and granulocytic tumours, hairy-cell tumour, adenoma, hyperplasia, medullary carcinoma, pheochromocytoma, mucosal neuromas, intestinal ganglioneuromas, hyperplastic corneal nerve tumour,
  • non-solid tumours
  • the patient to be treated suffers from a neoplasm selected from the group consisting of pancreatic carcinoma, hepatoblastoma, colon carcinoma, (small cell lung cancer, melanoma, mamma carcinoma, ovarian carcinoma, prostate carcinoma, glioblastoma, acute lymphoblastic leukemia, acute myeloid leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, pro-forms of leukemia, hairy cell leukemia, Hodgkin's disease, Non-Hodgkin lymphoma, lymphoma, tumour stem cells, glioblastoma-initiating stem cells and multiple myeloma.
  • a neoplasm selected from the group consisting of pancreatic carcinoma, hepatoblastoma, colon carcinoma, (small cell lung cancer, melanoma, mamma carcinoma, ovarian carcinoma, prostate carcinoma, glioblastoma, acute lymphoblastic leukemia, acute myeloid le
  • the patient exhibits either an intrinsic or an acquired resistance.
  • a “resistance” can either be total or partly; in other words, the patients considered treatable according to the invention can exhibit a reduced sensitivity or even a full lack of sensitivity to conventional anticancer treatments. These patients can also be determined as “non-responders” or “poor-responders”.
  • a further synonym for a “resistant” cancer or tumour is a “refractory” type of cancer, which can also be either completely or partly refractory. Intrinsic resistance can thus also be determined as a “primary refractory cancer”.
  • a particular form of refractory or resistant cancer cells are the so called “kinetically refractory cells”; a phenomenon known e.g. from leukemia cells, when the cells are at first killed, but reproduce fast that an effective treatment is hardly possible.
  • the term “conventional” treatment or therapy refers to the currently accepted and widely used therapeutic treatment of a certain type of cancer, based on the results of past researches and/or regulatory approval.
  • Conventional anticancer drugs include cytotoxic and cytostatic agents, which kill the cancer cells or reduce and/or stop their growth or proliferation.
  • the modes of action of these anticancer drugs can vary; examples are antimetabolites (e.g. cytarabine, methotrexate, mercaptopurine or clofarabine), DNA cross-linking agents (e.g. cisplatin and its derivatives), DNA intercalating substances (e.g. doxorubicin), Topoisomerase poisons (e.g. etoposide), kinase inhibitors (e.g. cetuximab), steroids (e.g. dexamethasone) or mitotic inhibitors (e.g. vincristine).
  • antimetabolites e.g. cytarabine, methotrexate, mercaptopurine or clofarabine
  • DNA cross-linking agents e.g. cisplatin and its derivatives
  • DNA intercalating substances e.g.
  • the conventional radiotherapy can also include radiation therapy, which means the use of high-energy radiation from x-rays, alpha, beta and gamma rays, Auger electrons, Ultraviolet rays, neutrons, protons, and other sources to kill cancer cells and shrink tumours.
  • Radiation may originate from an outside the body device (external-beam radiation therapy), or it may originate from radioactive sources placed in the body in the vicinity of the cancer cells (internal radiation therapy).
  • Systemic radiation therapy uses a radioactive substance, such as a radiolabeled monoclonal antibody, that travels in the blood stream to the target tissue. Radio resistant cancer cells do not or only partly respond to these treatments.
  • the opioid receptor agonists are applied for overcoming or “breaking” the intrinsic or acquired resistance of cancer cells to conventional anticancer treatments and/or radiation treatment or apoptosis resistance.
  • cancer cells considered treatable according to the invention express an opioid receptor, in particular the p opioid receptor.
  • the terms “resistance”, “radioresistance” or “chemoresistance” are defined as a reduced sensitivity of a cancer cell to at least one conventional cancer therapy, i.e. either an anticancer drug or radiotherapy.
  • a patient suffering from such a cancer is determined as a “resistant” cancer patient. Since the resistance can be intrinsic or acquired the observed reduction in sensitivity is either compared to fully sensitive “normal” cancer cells, which are responsive to the therapeutically effective dosage of the applied anticancer drug and/or radiation compared to the original sensitivity upon therapy onset. In the later case the resistance manifests either in a diminished amount of tumour regression for the same dose (either of the radiation or the anticancer drug) or an increased dose which is necessary for an equal amount of tumour regression.
  • the patient exhibits one or more of the subsequent resistances: apoptosis resistance, multi-drug resistance, anticancer drug resistance, cytotoxic drug resistance, resistance to reactive oxygen species, resistance to DNA damaging agents, resistance to toxic antibodies, doxorubicin resistance, single or cross resistance, irradiation resistance (e.g. alpha, beta, gamma or Auger electrons).
  • the patient is resistant to one or more of the following drug substances: methotrexate, cytarabine, thioguanine cisplatin, oxaliplatin, etoposide, vincristine, paclitaxel, carboplatin, teniposide, dexamethasone, prednisolone, cyclophosphamide, diphosphamide, doxorubicin, epirubicin, daunorubicin, idarubicin, mercaptopurine, fludarabine, gemcitabine, temozolomide, anti-HER2, and anti-CD20.
  • methotrexate methotrexate
  • cytarabine thioguanine cisplatin
  • oxaliplatin etoposide
  • vincristine paclitaxel
  • carboplatin teniposide
  • dexamethasone prednisolone
  • cyclophosphamide diphosp
  • the anticancer agent that is administered together with the opioid receptor agonist is given at a dose, which is equal than or lower than the recommended dose for the respective cancer.
  • the recommended dose is given by a conventional cancer therapy without the administration of an opioid receptor agonist.
  • the respective dose of the anticancer agent from the perspective of the skilled person represents a suboptimal or sub therapeutic dose, which have the advantage for the patient to have less side effects.
  • the main effect is that the uptake of the dose of the anticancer drug is increased in the cancer cells, while the plasma concentration is on the level of the conventional therapy. This has the effect that non responder to conventional therapy could be treated.
  • the anticancer agent that is administered together with the opioid receptor agonist is given at a dose, which is 2 times lower, preferably 3, 5, 10, or 30 times lower and even more preferably 100 times lower than the recommended dose for the treatment of cancer using the anticancer agent only.
  • the anticancer agent that is administered together with the opioid receptor agonist is given at a dose, which is equal than or lower than the recommended dose for the respective cancer, wherein the period of effective plasma levels of the anticancer agent is completely within the period of effective plasma levels of the opioid receptor agonist.
  • the recommended dose is given by a conventional cancer therapy without the administration of an opioid receptor agonist.
  • the opioid receptor agonist is D/L-methadone and the anticancer agents are methotrexate and dexamethasone.
  • the opioids or opioid receptor agonist can be used as a composite with at least one anticancer drug.
  • anti-Her2 denotes to any ligand that binds to and interacts with the gene product of the Her-2/Neu gene. This encompasses antibodies such as Trastuzumab (herceptin) or any organic compounds.
  • a “composite” within the context of the present invention relates to a pharmaceutical preparation comprising a therapeutically effective amount of any of the opioid receptor agonist (component A) as defined according to the invention and at least one further anticancer substance (component B).
  • This “composite” can constitute a single composition or at least two compositions, which can be administered to the patients either concomitantly or subsequently.
  • the above mentioned substances are preferably combined with methadone, more preferably with the hydrochloride form of D/L-methadone.
  • the composite of the invention can be advantageous for the effective treatment of cancer cells, since it can exhibit synergistic effects compared to the single compositions.
  • methadone as component A and one of the agents as component B as follows is preferred: methotrexate, cytarabine, cisplatin, carboplatin, oxaliplatin, etoposide, vincristine, doxorubicin, idarubicin, epirubicin, daunorubicin, fludarabine.
  • gemcitabine paclitaxel, docetaxel, temozolomide, anti-CD20, anti-HER2.
  • combinatorial treatment also comprising irradiation treatments is possible.
  • a further preferred composite consists of methadone as component A and temozolomide as component B.
  • opioids are used to treat either resistant or sensitive non-solid cancers, i.e. all haematological malignancies affecting blood, bone marrow and lymph nodes, including acute lymphoblastic leukemia, acute myeloid leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia and all pro-forms of leukemia, hairy cell leukemia, Hodgkin's disease, Non-Hodgkin lymphoma, lymphoma and multiple myeloma.
  • resistant or sensitive non-solid cancers i.e. all haematological malignancies affecting blood, bone marrow and lymph nodes, including acute lymphoblastic leukemia, acute myeloid leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia and all pro-forms of leukemia, hairy cell leukemia, Hodgkin's disease, Non-Hodgkin lymphoma, lymphoma and multiple myeloma.
  • the invention provides a method for the selection of a combination of an opioid receptor agonist and one or more anticancer drugs. This method comprises the following steps:
  • the in vitro cultured cancer cells can be an immortalized cell line, xenografted cells, a secondary or a primary cancer cell line or primary cells.
  • the cell line and/or cells is derived from a cancer biopsy, in more preferred embodiment the biopsy or blood sampling or cerebrospinal fluid sampling or pleural fluid sampling or amniotic fluid sampling or peritoneal fluid sampling is taken from the patient to be treated with the combination according the invention.
  • the cancer cell line can represent a homogenous cell line based only on one cancer cell type or a heterogeneous cancer cell line comprising of different cell types.
  • the analysis of the opioid receptor expression in step (b) can be performed by techniques which are known to the person skilled in art.
  • a non-limiting list of examples include immunofluorescence using an antibody or antibody fragment directed against said opioid receptor, the immunoprecipitation of the opioid receptors, or the use of labelled opioid receptor ligands such as naloxone-fluorescein.
  • cytolysis or membrane leakage assays such as the lactate dehydrogenase assay, the propidium iodide assay, the Trypan blue assay, the 7-Aminoactinomycin D assay,
  • mitochondrial activity or caspase assays such as the Resazurin and Formazan (MTT/XTT) can assay for various stages in the apoptosis process that foreshadow cell death
  • functional assays which in the case of red blood cells measure the cell deformability, osmotic fragility, haemolysis, ATP level, and haemoglobin content
  • genomic and proteomic assays which include the analysis of the activation of stress pathways using DNA microarrays and protein chips.
  • the cell viability is measured by the propidium iodide assay and the apoptosis is measured by determination of hypodiploid DNA (subG1) and FSC/SSC analyses by flow cytometry.
  • the cultured cells are preferably treated in parallel experiments comprising the use of the opioid alone, the anticancer agent alone and a combination of the two substances.
  • the potency of the effect is analysed by studying the dose dependency of the respective effect.
  • several anticancer agents can be combined to increase the anti-apoptotic effect or opioid receptor expression or to reduce the side effect profile.
  • the initial selection of the test compounds will depend on the characteristics of the tumour.
  • the patient characteristics can be taken in consideration including the age, the sex, the body weight, co-morbidities, individual metabolic capabilities, allergies and incompatibilities, genetic predisposition, the course of the disease and the family history.
  • the opioid receptor agonists as described above can be used for testing.
  • D,L-methadone, L-methadone, fentanyl, buprenorphine, morphine, codeine, oxycodone, tramadol and tapentadol are used.
  • an anti cancer agent is chosen which is well known to have an effect on the respective cancer cell type, cell line or cells.
  • the cultured cells are analysed for opioid receptor expression prior anticancer treatment and after the anticancer treatment under conditions which allow a comparison of the opioid receptor expression levels. Said comparison allows to identify anticancer agents which increase the opioid receptor expression on the respective cancer cell.
  • step (e) prioritizes the drug combination and/or the respective doses in order to maximise the efficacy while retaining a side effect profile which is acceptable for the patient.
  • the selection in step (f) prioritizes an anticancer agent with regard to its ability to increase the opioid receptor expression on the cancer cell.
  • the anti-apoptotic effect of the opioid agonist, as well as the anti-apoptotic affect of the anticancer agent is maximised.
  • step (c) the cell culture was treated with a combination of opioid receptor agonist and anticancer agent the prioritization of the combination which is used, is done under the aspect which combination of doses has the better lethal effect on the cells in culture.
  • FIG. 2 c for example shows that with doxorubicin in conventional therapeutical dose as described in the instruction leaflet a D,L-methadone dose of 0,1 ⁇ g/mL would be preferable.
  • the invention provides a method for selection of an opioid receptor agonist for the treatment of cancer comprising the following steps:
  • steps (a) to (d) can be performed by methods and strategies as described above.
  • the analysis of the opioid receptor expression allows a selection of cancer type which might be treated with an opioid receptor agonist. Due to the in vitro treatment with an opioid receptor agonist, the individual dose for the cancer in vivo treatment can be determined.
  • D,L-methadone hydrochloride D,L-methadone
  • doxorubicin D,L-methadone hydrochloride
  • naloxone from Fagron GmbH&Co. KG (Barsbüttel, Germany)
  • PTX pertussis toxin
  • D,L-methadone Metalhaddict, Hexal, Germany
  • the tablets were pulverized and solubilized freshly before use in 10% Tween 80 in saline.
  • Doxorubicin (Hexal) was purchased as injection solution (5 mg/ml) and diluted freshly with saline to the appropriate concentrations.
  • BCP-ALL human B-cell leukemia
  • FCS 10% heat inactivated FCS (Lonza, Verviers, Belgium), 1 mmol/L glutamine (Invitrogen), 1% penicillin/streptomycin (Invitrogen), 25 mmol/L HEPES (Biochrom) at 37° C., 95% air/5% CO 2 .
  • the leukemia cells were seeded in a density of 10,000 cells/mL.
  • Serum concentrations of methadone Determination of methadone in serum samples was carried out after liquid/liquid extraction using a mass spectrometer equipped with a gas chromatograph (GC/MS). As internal standard d 9 -methadone was added. The mass selective detector was operated in electron impact mode. Data were acquired in the selected-ion monitoring mode. The analytes were identified with the following masses m/z 294, 223, 72 (target ion) for methadone and m/z 303, 226, and 78 for d 9 -methadone with a limit of detection of 0,8 ng/ml and a limit of quantification of 1,2 ng/ml.
  • ALL-SCID6 model was chosen. Fragments from in vivo passaged tumours were transplanted at day zero subcutaneously to 32 male NOD/SCID/IL2ry null (NSG) mice. After randomization oral treatment (by gavage) with D,L-methadone was initiated one day later and performed daily until the end of the experiment with increasing doses: 1 st week 20 mg/kg/d, 2 nd week 30 mg/kg/d, 3 rd week 40 mg/kg/d, 4 th week 60 mg/kg/d, 5 th -10 th week 2 ⁇ 60 mg/kg/inj.. The dose adaptation was necessary to avoid toxic deaths because of an overdosage of D,L-methadone.
  • the maximum tolerated dose of D,L-methadone in the employed mouse strain is 2 ⁇ 60 mg/kg/inj.
  • 53, 60 and 76 doxorubicin 3 mg/kg was administered i.v..
  • Tumour size was measured twice weekly at two dimensions and tumour volumes were calculated according to the formula (length ⁇ width 2 )/2.
  • Mean tumour volumes and standard deviations were calculated per group.
  • Treated to control values (T/C) in percent were calculated by relating mean tumour volumes of each group at each measurement day to the controls.
  • Individual body weight was determined twice per week as parameter for tolerability and body weight changes in percent were calculated by relating the mean values of each group to the first measurement day.
  • Serum from D,L-methadone treated mice was taken 0.5, 1, 4 and 24 hours after last D,L-methadone treatment at day 76, respectively, and stored at ⁇ 20° C. until the determination of methadone concentration. Mice were sacrificed at day 77 for ethical reasons.
  • Flow cytometric assay for determination of cell surface opioid-receptors Cells were washed in PBS supplemented with 1% FCS, centrifuged and resuspended in PBS/1% FCS containing naloxone-fluoresceine (0.05 mM, Invitrogen) (Hedin et al., 1997). After 30 min of incubation at RT, the cells were washed twice with PBS/1/0 FCS, centrifuged and resuspended in icecold PBS/1% FCS. Flowcytometry analysis was performed using FACSCalibur (BD, Heidelberg, Germany).
  • ALL cells were treated with D,L-methadone ( ⁇ 3 ⁇ g/mL therapeutic plasma concentration) alone or in addition to doxorubicin in 175 cm 2 flasks or 96-well plates. Further experiments were performed simultaneously after addition of 60 ⁇ g/mL naloxone, 200 ⁇ M IBMX or 200 ng/mL PTX. After different points in time, apoptosis rates were measured by flowcytometry (Carbonari et al., 1994; Nicoletti et al., 1991).
  • apoptosis To determine apoptosis, cells were lysed with Nicoletti-buffer containing sodium citrate (0.1%), Triton X-100 (0.1%) and propidium iodide (50 ⁇ g/mL) as described by Nicoletti (Nicoletti et al., 1991). Apoptotic cells were determined by hypodiploid DNA (subG1) or forward scatter/side scatter analysis (Carbonari et al., 1994). The percentage of specific apoptosis was calculated as follows: 100 ⁇ [experimental dead cells (%) ⁇ spontaneous dead cells in medium (%)]/[100% -spontaneous dead cells in medium (%)]. The spontaneous dead cells were in the rage of 5 to 10% using cell lines. The viability of the untreated patient cells (spontaneous dead cells) was less than 35% at 24 h and 48h.
  • leukemia cells were treated with the pancaspase inhibitor of caspases, zVAD.fmk (benzoylcarbonyl-Val-Ala-Asp-fluoromethylketone; Enzyme-Systems-Products, Dubli, USA) as described (Friesen et al., 2007). 50 ⁇ M zVAD.fmk was added to the cells 1h before stimulation with D,L-methadone and doxorubicin. After different time points, the percentage of apoptotic cells was determined by FSC/SSC analysis via flowcytometry(Carbonari et al., 1994).
  • peroxidase-conjugated-goat-anti-mouse IgG or peroxidase-conjugated-goat-anti-rabbit IgG (1:5000, Santa-Cruz) were used for the enhanced chemoluminescence system (ECL, Amersham-Pharmacia, Freiburg, Germany). Equal protein loading was controlled by ⁇ -actin detection.
  • the BCP-leukemia cell line Tanoue was seeded in a density of 100,000 cells/mL in 175 cm 2 flasks and was either left untreated or incubated with 0.3 ⁇ g/mL doxorubicin or a combination of 0.3 ⁇ g/mL doxorubicin and 3 ⁇ g/mL D,L-methadone at 37° C./5%/CO 2 . After 24h, cells were washed twice with ice-cold PBS/1% FCS. Relative doxorubicin uptake in cells was analyzed using flowcytometry.
  • doxorubicin efflux For analysis of doxorubicin efflux, cells were washed to remove doxorubicin from medium after incubation for 24h. Next, cells were incubated with fresh medium without doxorubicin or fresh medium containing 3 ⁇ g/mL D,L-methadone at 37° C./5%/CO 2 without doxorubicin to measure doxorubicin efflux. After different time points cells were harvested, washed and relative doxorubicin content in leukemia cells was analyzed using flowcytometry.
  • D,L-Methadone Induces Cell Death in Xenograft-Derived all-Cells Depending on Opioid Receptor Expression
  • D,L-methadone in treatment of leukemia and the role of opioid receptor triggering in cell death induction, the anti-cancer effect of D,L-methadone was analyzed in different xenograft-derived ALL-cells.
  • the xenografts were originally established from patients with T-cell (ALL-SCID6, ALL-SCID3), B-cell (ALL-SCID7) (Borgmann et al., 2000) and B-cell precursor (BCP, pre-B-ALL-SCID) acute leukemia.
  • ALL-SCID6 ALL-SCID6
  • ALL-SCID7 Borgmann et al., 2000
  • BCP pre-B-ALL-SCID
  • ALL-SCID6, ALL-SCID3 and the ALL-SCID7 leukemia cells displayed opioid-receptors in high amounts ( FIG. 1A ), whereas the pre-B-ALL-SCID expressed only moderate levels of opioid-receptors ( FIG. 1A ).
  • ALL-SCID6, ALL-SCID3, ALL-SCID7 and pre-B-ALL-SCID were treated with different concentrations of D,L-methadone ( FIG. 1 B).
  • D,L-methadone ⁇ 3 ⁇ g/mL
  • a higher concentration of 10 ⁇ g/mL D,L-methadone was used, because levels of D,L-methadone in lymphatic tissue and marrow may be higher, but have not been measured (Singh et al., 2011). It was found that therapeutic plasma concentrations of D,L-methadone ( ⁇ 3 ⁇ g/mL) induced a strong cell death in xenograft-derived ALL-cells expressing high amounts of opioid-receptors on their cell surface (FIG. 1 A,B). In comparison to these observations, the pre-B-ALL-SCID having a moderate opioid-receptor level ( FIG. 1A ) could only be slightly killed with therapeutic concentrations of D,L-methadone ( FIG. 1B ). This clearly reveals that apoptosis induction by D,L-methadone is depend on the level of opioid-receptor expression.
  • BCP-ALL-cell lines could only be killed slightly by D,L-methadone ( FIG. 2 b ) as observed for the pre-B-ALL-SCID ( FIG. 1B ).
  • the cell lines Tanoue, Reh, Nalm6 and pre-B-ALL-SCID were treated with different concentrations of D,L-methadone and doxorubicin alone or in combination with each other ( FIG. 2B , 2 C). It was observed that the combination treatment strongly induced cell kill in BCP-ALL-cell lines as well as in xenograft-derived-BCP-ALL-patient-cells (pre-B-ALL-SCID) (FIG. 2 B,C).
  • PARP poly-(ADP-ribose)-polymerase
  • BCP-ALL-cells were pre-incubated with or without 50 pM of zVAD.fmk and treated with D,L-methadone in addition to doxorubicin.
  • zVAD.fmk strongly decreased cell death after combination treatment with D,L-methadone and doxorubicin in BCP-ALL-cells ( FIG. 3B ) underlining the dependence on caspases activation.
  • the apoptotic machinery is tightly controlled by anti-apoptotic factors like XIAP and Bcl-x L (Fulda, 2009a; Fulda, 2009b) which we found to be strongly downregulated in BCP-ALL-cells treated with D,L-methadone in addition to doxorubicin ( FIG. 3C ).
  • D,L-methadone and doxorubicin sensitizes BCP-ALL-cells for apoptosis via the activation of caspases and downregulation of XIAP and Bc1-x L .
  • doxorubicin might influence the opioid-receptor expression
  • the BCP-ALL-cell line Tanoue was treated with doxorubicin for 96h. Afterwards, the relative amount of opioid-receptors compared to untreated cells was measured by flowcytometry. It was found that doxorubicin strongly increased opioid-receptor expression ( FIG. 4A ) suggesting that D,L-methadone can bind in higher amounts to cells co-treated with doxorubicin. This effect could presumably result in the higher cytotoxic potential of the combination treatment with D,L-methadone and doxorubicin.
  • Opioids Like D,L-Methadone Enhances the Uptake of Doxorubicin and Inhibits its Efflux
  • Opioids are substrates of the in multi-drug resistances-involved efflux pump P-glycoprotein (P-gp).
  • P-gp multi-drug resistances-involved efflux pump
  • D,L-methadone might influence the uptake and/or efflux of doxorubicin in leukemia cells
  • the BCP-ALL-cell line Tanoue was incubated for different intervals with doxorubicin alone or with a combination of doxorubicin and D,L-methadone. After 24h (Oh), an enhanced doxorubicin concentration in the cells co-incubated with doxorubicin and D,L-methadone ( FIG. 4B ) was observed.
  • the BCP-ALL-cell line Tanoue was treated with D,L-methadone, doxorubicin or with the opioid-receptor antagonist naloxone alone or in different combinations with each other (FIG. 5 A,B).
  • Opioid receptor stimulation activates inhibitory G i -proteins which in turn block adenylyl cyclase activity reducing cAMP ( FIG. 7 ).
  • cAMP is an inhibitor of DNA-damage—as well as doxorubicin-induced apoptosis in leukemia cells (Naderi et al., 2009; Safa et al., 2010a).
  • Pertussis toxin (PTX) inactivates G i -proteins and blocks downregulation of cAMP (Law et al., 1985) ( FIG. 7 ).
  • IBMX however increases cAMP levels as a result of phosphodiesterase inhibition ( FIG. 7 ).
  • ALL-SCID6 patient-derived-ALL-xenograft-model
  • NSG NOD/SCID/IL2ry null mice
  • D,L-methadone was orally administered starting at day one after ALL-inoculation with increasing doses.
  • doxorubicin treatment was initiated.
  • D,L-methadone and doxorubicin treatment led to a significant inhibition of tumour growth at comparable levels ( FIG. 6 ).
  • Combination treatment with D,L-methadone and doxorubicin had a similar anti-tumour efficacy as D,L-methadone or doxorubicin alone until day 70. At later time points, the tumour inhibition was longer lasting during the combined treatment of D,L-methadone and doxorubicin.
  • the therapy was well-tolerated with body weight changes of ⁇ 10% for the combination and ⁇ 8% or ⁇ 4% for the D,L-methadone or doxorubicin treatment, respectively.
  • To analyze D,L-methadone serum concentrations in mice 0.5, 1, 4 and 24 hours after the last D,L-methadone application, serum was taken and D,L-methadone quantified by mass spectrometry.
  • the serum concentrations of methadone were found between 28 ng/mL and 138 ng/mL in the time course of 0.5 until 4 hours after D,L-methadone application indicating that levels comparable with the in vitro concentrations could be reached.
  • the serum concentrations of doxorubicin were found between 156 ng/mL and 198 ng/mL.
  • the glioblastoma cell lines A172 and U118MG express opioid receptors.
  • primary glioblastoma cells s. FIG. 11A
  • glioblastoma-initiating stem cells s. FIG. 12A
  • opioid receptors express opioid receptors.
  • the combination treatment of D,L-methadone and doxorubicin dose-dependently induces apoptosis (see FIGS. 9 , 10 , 11 B and 12 B).
  • glioblastoma cell line A172 As exemplified for the glioblastoma cell line A172 it could be shown that cell death induction of glioma cells using D,L-methadone and doxorubicin cotreatment depends on caspase activation (s. FIG. 13 ). Furthermore, it could be shown that D,L-methadone reversed deficient activation of apoptosis pathways by doxorubicin in glioblastoma-initiating stem cells (s. FIG. 14 ).
  • doxorubicin leads to a 6-fold increase in opioid receptor expression (s. FIG. 16 ). It could be shown that this mechanism holds also true for other cancer types and anticancer drugs since in the promyelocytic leukemia cell line HL60, the cisplatin-treatment leads to a 2.1-fold increase in opioid receptor expression (s. FIG. 19 ).
  • D,L-methadone sensitizes leukemia cancer cells (Nalm-6), pancreatic cancer cells (Nalm6) and ovarian cancer cells (A2780) for etoposide or cisplatin treatment (s. FIG. 17 ).
  • CLL chronic lymphocytic leukemia cells
  • the Her2/Neu-resistent mamma carcinoma cell line JIMT-1 expresses the p-opioid receptor (s. FIG. 20 ).
  • the combination treatment of D,L-methadone and doxorubicin dose-dependently induces apoptosis in JIMT-1 cells (see FIG. 21 ). It could be shown that cell death induction of JIMT-1 cells using D,L-methadone and doxorubicin cotreatment depends on caspase activation (s. FIGS. 22 and 23 ).
  • fentanyl As exemplified for the T-cell derived leukemia cell line CEM it could be shown that also the opioid fentanyl was able to sensitize the CEM cells for treatment using doxorubicin (s. FIG. 24 ). In a further in vitro experiment, the opioid buprenorphine sensitized leukemia cells (HL-60) for apoptosis due to doxorubicin (s. FIG. 25 ).
  • the cell death potential of D,L-methadone on different leukemia-cell lines was shown on human T cell leukemia, human acute myeloid leukemia, human B cell precursor leukemia, human B cell leukemia. All tested cell lines expressing opioid-receptors in a moderate level on their cell surface ( FIG. 27 ).
  • the molecular pathways of cell killing was shown in more detail and it was shown how the combination treatment with an opioid receptor agonist i.e. D,L-methadone and an anticancer agent i.e. cisplatin induce apoptosis.
  • an opioid receptor agonist i.e. D,L-methadone
  • an anticancer agent i.e. cisplatin induce apoptosis.
  • Combination treatment of D,L methadone in combination with Cisplatin (+CDDP) was compared to cells treated with D,L-methadone ( ⁇ CDDP) or cisplatin alone.
  • zVAD.fmk Different leukemia cells (human T cell leukemia, human acute myeloid leukemia, human B cell precursor leukemia) were pre-incubated with 50 ⁇ M of zVAD.fmk (+zVAD.fmk, white bars) or without zVAD.fmk ( ⁇ zVAD.fmk, black bars) and treated with D,L-methadone in addition to cisplatin.
  • zVAD.fmk strongly decreased cell death after combination treatment with D,L-methadone and cisplatin ( FIG. 30 ) underlining the dependence on caspases activation.
  • apoptotic machinery is tightly controlled by anti-apoptotic factors like XIAP and Bcl-xL and pro-apoptotic factors like Bax (Fulda, 2009a; Fulda, 2009b).
  • XIAP was strongly downregulated (p57) and or cleaved (p30) in different leukemia cells treated with D,L-methadone in addition to cisplatin (+CDDP) ( FIG. 29 ) depending on different concentrations of cisplatin or/and different concentration of D,L-methadone.
  • a strong upregulation of Bax (p21) is induced in human T cell leukemia induced after treatment with D,L-methadone in addition to cisplatin (+CDDP).
  • the combination of D,L-methadone and cisplatin sensitizes different leukemia cells for apoptosis via the activation of caspases and by downregulation and inhibition of anti-apoptotic factors such as XIAP and upregulation of pro-apoptotic factors such as Bax.
  • the opioid receptors are receptors which induce cell death and activate apoptosis pathways involving caspase activation, downregulation and or cleavage of PARP, and or downregulation of anti-apoptotic factors, and or upregulation of pro-apoptotic factors, and or downregulation and inhibition of inhibitory apoptotic proteins (IAP). Therefore the opioid receptors are a new unknown way with an new mechanism of inducing cell death, beside the common known cell death receptors/death inducing ligands systems and mechanisms like the CD95/CD95L-System.
  • Cisplatin Strongly Induces Opioid-Receptor Expression in Leukemia Cells
  • D,L-methadone The efficiency of cell death induction and activation of effector molecules in apoptosis pathways after treating leukemia cells with opioid-receptor-agonists i.e. D,L-methadone depend on the amount of opioid-receptors displayed on the cell's surface. Combination treatment with D,L-methadone and cisplatin profoundly kills leukemia cells with moderate opioid receptor expression, which could only be killed slightly by D,L-methadone or cisplatin alone. Chemotherapeutics enhance the expression of the receptor CD95 (FAS,APO-1) in leukemia cells which is a special known death receptor (Posovszky et al., 1999).
  • cisplatin has an influence to the opioid-receptor expression
  • the different leukemia cells human T cell leukemia, human acute myeloid leukemia, human B cell precursor leukemia
  • the relative amount of opioid-receptors compared to untreated cells was measured by flowcytometry. It was shown that cisplatin strongly increased opioid-receptor expression ( FIG. 31 ). Therefore opioid receptor-agonists like D,L-methadone can bind in higher amounts to cells, co-treated with cisplatin or other anticancer agents which are able to induce a higher level of expressed opioid receptors. This effect results in the higher cell death potential of the combination treatment of an opioid receptor agonist i.e D,L-methadone and an anticancer agent i.e. cisplatin.
  • the opioid receptor agonist which has a longer minimal duration of effectiveness like D,L methadone has a better result than one, that has a shorter minimal duration of effectiveness like morphine compared to D,L methadone ( FIG. 33 and FIG. 34 .
  • glioblastoma cells were treated with D,L-methadone, doxorubicin or with the opioid-receptor antagonist naloxone alone or in different combinations with each other ( FIG. 32 ). After 120h and 144h it was shown that blocking opioid-receptors by naloxone strongly reduced the apoptosis rates of the combination treatment with D,L-methadone and doxorubicin ( FIG. 32 ).
  • the efficiency of cell death induction after treating glioblastoma cells or leukemia cells with opioids depends on the duration of effectiveness of the opioids.
  • the minimal duration of effectiveness of methadone is 5-7 hours and the minimal duration of effectiveness of morphine is 2-4 hours.
  • Combination treatment with D,L-methadone and doxorubicin strongly induced high cell death rates in glioblastoma cells ( FIG. 33 A) and leukemia cells ( FIG. 34 A).
  • combination treatment with morphine and doxorubicin induced lower cell death rates in glioblastoma cells ( FIG. 33 B) and leukemia cells ( FIG. 34 B). This indicates that the rates of induction of cell death after combination treatment of opioids with anticancer drugs depend also on the duration of effectiveness of opioids. The effect is found also at other anticancer agents.
  • G1 gap 1
  • S synthesis
  • M mitosis
  • CDKs protein kinases
  • Opioid receptor agonist for example Methadone in combination with doxorubicin inhibits proliferation of cancer cells such as glioblastoma cells and induces S/G2-M cell cycle arrest in glioblastoma cells.
  • cAMP-related signaling can control apoptosis induction and cell growth.
  • glioblastoma cells A172 were treated with the opioid receptor agonist D,L-methadone, the anticancer agent doxorubicin or with the opioid receptor antagonist naloxone alone or in different combinations ( FIGS. 36A , 36 B and 36 C).
  • U87MG glioblastoma cells were subcutaneously inoculated per nude-mouse. After randomization of 16 mice, D,L-methadone was daily orally administered in 8 mice starting at day 1 until the end of experiment. D,L-Methadone dosage was increased weekly from 60 to 120 to 240 mg/kg/d bid. At day 33, 24h after the last treatment with D,L-methadone the mice were sacrificed. For analyzing serum concentrations of D,L-methadone in mice 0.5, 1 and 4h after last D,L-methadone-application, serum was taken and D,L-methadone quantified by mass spectrometry.
  • the D,L-methadone treated mice had a significantly reduced tumour size at day days 19 to 33 with an optimum T/C value of 49% (see FIG. 37 ).
  • the D,L-methadone treatment was well tolerated in the dose used and induced only a minor body weight loss of 9%. Serum concentrations were found between 136 ng/ml and 1608 ng/mL of methadone in the time course of 0.5 to 4h after D,L-methadone application.
  • Opioids Such as D,L-Methadone Increase Cisplatin-Induced Cell Death in Ovarian Cancer Cell after Short Term Treatment
  • A2780 ovarian cancer cells were treated with cisplatin (5, 3 ⁇ g/mL) or D,L-methadone (3, 1 ⁇ g/mL) alone or in combination.
  • cisplatin 5, 3 ⁇ g/mL
  • D,L-methadone 3, 1 ⁇ g/mL
  • opioids such as D,L-methadone strongly potentiates cisplatin-induced apoptosis in ovarian cancer cells.
  • Opioids Such as D,L-Methadone Increase Cisplatin-Induced Cell Death in Ovarian Cancer Cell after Long Term Treatment
  • A2780 ovarian cancer cells were treated with cisplatin (2, 1, 0.5, 0.3 ⁇ g/mL) or D,L-methadone (10, 3, 1 ⁇ g/mL) alone or in combination.
  • cisplatin 2, 1, 0.5, 0.3 ⁇ g/mL
  • D,L-methadone 10, 3, 1 ⁇ g/mL
  • opioids such as D,L-methadone strongly potentiates cisplatin-induced apoptosis in ovarian cancer cells and breaks chemoresistance.
  • Opioids Such as D,L-Methadone Increase Cisplatin-Induced Cell Death in Cisplatin-Resistant Ovarian Cancer Cell
  • A2780cis ovarian cancer cells were treated with cisplatin (3, 2, 1 ⁇ g/mL) or D,L-methadone (10, 3, 1 ⁇ g/mL) alone or in combination.
  • cisplatin 3, 2, 1 ⁇ g/mL
  • D,L-methadone 10, 3, 1 ⁇ g/mL
  • opioids such as D,L-methadone strongly potentiates cisplatin-induced apoptosis in cisplatin-resistant ovarian cancer cells.
  • A2780 ovarian cancer cells were treated with cisplatin (2 ⁇ g/ml) alone. As shown in FIG. 41 , cisplatin in a concentration of 2 ⁇ g/mL induced cell death of 90% after 144h. However, treatment with 0.5 ⁇ g/mL cisplatin in addition to D,L-methadone (10, 3, 1 ⁇ g/mL) induced a cell death of 95%. Furthermore, treatment with 0.2 ⁇ g/mL cisplatin in addition to D,L-methadone (10, 3, 1 ⁇ g/mL) induced a cell death of between 70 and 85% depending on concentrations of D,L-methadone.
  • D,L-methadone strongly potentiates cisplatin-induced apoptosis/cell death in ovarian cancer cells.
  • D,L-methadone increases the effectiveness of cisplatin in treatment of ovarian cancer, suggesting that a strong reduction of anti-cancer drugs concentrations can be used by cotreatment with D,L-methadone to get comparable cell death rates and therefore less side effects of the anti-cancer drugs will be observed.
  • the anticancer agent can be given at a dose which it at least 2 or 3 times lower and up to 100 times lower than the recommended dose for the treatment of the respective cancer.
  • A2780 ovarian cancer cells were treated with D,L-methadone (3, 1 ⁇ g/mL) or cisplatin (5, 3 ⁇ g/mL) alone or in combination. As shown in FIG.
  • the combination treatment using cisplatin and D,L-methadone leads to a strong caspase activation in ovarian cancer cells by activating caspase-3, caspase-9, and caspase-8 and cleavage of Poly-(ADP-ribose)-polymerase (PARP).
  • PARP Poly-(ADP-ribose)-polymerase
  • the Trastuzumab resistant breast cancer cells JIMT-1 were treated with the opioid receptor agonist D,L-methadone, doxorubicin or with the opioid receptor antagonist naloxone alone or in different combinations ( FIG. 43 ). Blocking opioid receptors by naloxone strongly reduced apoptosis induced by combination treatment with D,L-methadone and doxorubicin. This indicates that opioid receptor activation plays a critical role in apoptosis induction.
  • Opioids Such as D,L-Methadone Increase Cisplatin-Induced Cell Death in Prostate Cancer Cells
  • Prostate cancer cells PC-3 were treated with cisplatin (5, 3 ⁇ g/mL) or D,L-methadone (10, 3, 1 ⁇ g/mL) alone or in combination.
  • cisplatin 5, 3 ⁇ g/mL
  • D,L-methadone 10, 3, 1 ⁇ g/mL
  • opioids such as D,L-methadone strongly potentiates cisplatin-induced apoptosis in prostate cancer cells.
  • Opioids Such as D,L-Methadone Increase Cell Death Induction of Different Anti-Cancer Drugs from Different Anti-Cancer Drug Classes in Leukemia Cells
  • Leukemia cells Nalm6 were treated with different anti-cancer drugs alone (white columns) or in combination with D,L-methadone (black columns). As shown in FIG. 45 , a strong induction of cell death was observed by co-treatment of D,L-methadone and anti-cancer drugs (black columns). This suggests that opioids such as D,L-methadone strongly potentiates apoptosis induction of different anticancer drugs from different anticancer classes in leukemia cells.
  • Opioids Such as D,L-Methadone Increase Anti-Cancer Drug-Induced Cell in Glioblastoma Cells
  • Glioblastoma cells A172 were treated with different anti-cancer drugs from the same anti-cancer drug class such as anthracyclines (Doxorubicin, Idarubicin, and Daunorubicin). Glioblastoma cells were treated with anthracyclines alone (white columns) or in combination with D,L-methadone (black columns). As shown in FIG. 46 , a strong induction of cell death was observed by co-treatment of D,L-methadone and different anthracyclines. This suggests that opioids such as D,L-methadone strongly potentiates apoptosis induction of different anti-cancer drugs from the same class in glioblastoma cells.
  • anthracyclines Doxorubicin, Idarubicin, and Daunorubicin
  • Glioblastoma cells were treated with anthracyclines alone (white columns) or in combination with D,L-methadone (black columns).
  • Pancreatic cancer cells Colo 357 were stained with naloxone fluorescein measuring opioid receptor expression by flow cytometry. As a result a strong expression of opioid receptors on the surface of pancreatic cancer was found (see FIG. 47 ).
  • Opioids Such as D,L-Methadone Increase Anti-Cancer Drug-Induced Cell in Pancreatic Cancer Cells
  • Pancreatic cancer cells Colo 357 were treated with different anti-cancer drugs from the same anti-cancer drug class such as cisplatin metal complexes (oxaliplatin, cisplatin). Pancreatic cancer cells Colo 357 were treated with different concentration of cisplatin metal complexes, oxaliplatin or cisplatin alone or in combination with D,L-methadone. As shown in FIG. 48 , a strong induction of cell death was observed by co-treatment of D,L-methadone and the platin metal complexes (A) oxaliplatin or (B) cisplatin. This suggests that opioids such as D,L-methadone strongly potentiates apoptosis induction of different anticancer drugs from the same anti-cancer drug class in pancreatic cancer cells.
  • opioids such as D,L-methadone strongly potentiates apoptosis induction of different anticancer drugs from the same anti-cancer drug class in pancreatic cancer cells
  • pancreatic cancer cells Colo 357 were treated with D,L-methadone (3, 1 ⁇ g/mL) or oxaliplatin (3, 2 ⁇ g/mL; see FIG. 49A ) or cisplatin (0.5, 0.7 ⁇ g/mL; see FIG.
  • FIG. 49 B alone or in combination with methadone and oxaliplatin (see FIG. 49A ) or cisplatin (see FIG. 49B ).
  • the combination treatment leads to strong caspase activation in pancreatic cancer cells by activation of caspase-3, caspase-9, and cleavage of Poly-(ADP-ribose)-polymerase (PARP).
  • PARP Poly-(ADP-ribose)-polymerase
  • pancreatic cancer cells Colo 357 were incubated with the broad spectrum inhibitor of caspases zVAD.fmk. Incubation with zVAD.fmk almost completely inhibited apoptosis in pancreatic cancer cells induced by D,L-methadone in addition to oxaliplatin (see FIG. 50 A,B) or by D,L-methadone in addition to cisplatin (see FIG. 50 C,D), suggesting that caspases are central for opioid receptor activation-mediated sensitization of pancreatic cancer cells for oxaliplatin and cisplatin treatment.
  • opioid receptor activation such as D,L-methadone reverses deficient activation of caspases by oxaliplatin or cisplatin in pancreatic cancer cells.
  • Opioids Such as D,L-Methadone Increase Temozolomide (Temodal)—Induced Cell Death in Glioblastoma Cells
  • Glioblastoma cells A172 were treated with temozolomide or D,L-methadone (3, 1 ⁇ g/mL) alone or in combination. As shown in FIG. 51 , a strong induction of cell death was observed by co-treatment of D,L-methadone and temozolomide. This suggests that opioids such as D,L-methadone strongly potentiates temozolomide-induced apoptosis in glioblastoma cells.
  • Pancreatic cancer cells Colo 357 were treated with different concentrations of oxaliplatin (A) or cisplatin (B) alone or in combination with D,L-methadone (hatched columns, white columns). As shown in FIG. 52 , a strong induction of cell death was observed by co-treatment of D,L-methadone and different cisplatin metal complexes oxaliplatin (A) or cisplatin (B).
  • Pancreatic cancer cells Colo 357 were treated with (A) oxaliplatin (10 ⁇ g/ml) alone. 10 ⁇ g/mL cisplatin induced cell death of 60% after 120h. However, treatment with 3 ⁇ g/mL oxaliplatin in addition to D,L-methadone (10, 3, 1 ⁇ g/mL) induced a cell death of 65%. In addition, treatment with 2 ⁇ g/mL oxaliplatin in addition to D,L-methadone (10, 3, 1 ⁇ g/mL) induced a cell death of 45%.
  • Pancreatic cancer cells Colo 357 were treated with (B) cisplatin (10 ⁇ g/ml) alone. 10 ⁇ g/mL cisplatin induced cell death of 70% after 144h. However, treatment with 0.7 ⁇ g/mL cisplatin in addition to D,L-methadone (10, 3, 1 ⁇ g/mL) induced a cell death of 85%. In addition, treatment with 0.5 ⁇ g/mL cisplatin in addition to D,L-methadone (10, 3, 1 ⁇ g/mL) induced a cell death of 60%.
  • D,L-methadone increases the effectiveness of cisplatin or oxaliplatin in treatment of pancreatic cancer, suggesting that a strong reduction of anti-cancer drugs concentrations can be used by cotreatment with D,L-methadone to get comparable cell death rates and therefore less side effects of the anti-cancer drugs will be observed.
  • opioids such as D,L-methadone breaks chemoresistance because conventional therapies using anti-cancer drugs are limited by the toxicity of anti-cancer drugs concentrations used for patients treatment.
  • BCP-ALL cell lines (Nalm6, Reh and Tanoue) were treated with doxorubicin alone or in combination with D,L-methadone and doxorubicin (hatched columns).
  • doxorubicin alone or in combination with D,L-methadone and doxorubicin (hatched columns).
  • FIG. 53 a strong induction of cell death was observed by co-treatment using D,L-methadone. This suggests that D,L-methadone increases the effectiveness of doxorubicin in treatment of leukemia cells, suggesting that a strong reduction of anti-cancer drugs concentrations can be used by cotreatment with D,L-methadone to get comparable cell death rates and therefore less side effects of the anti-cancer drugs will be observed.
  • Trastuzumab resistant breast cancer cells (JIMT-1) were treated with doxorubicin alone or in combination with D,L-methadone and doxorubicin (hatched columns).
  • doxorubicin alone or in combination with D,L-methadone and doxorubicin (hatched columns).
  • FIG. 54 a strong induction of cell death was observed by co-treatment of D,L-methadone.
  • 0.1 ⁇ g/mL doxorubicin induced cell death of 70% after 120 h.
  • treatment with 0.015 ⁇ g/mL doxorubicin in addition to D,L-methadone (10, 3, 1, 0.1 ⁇ g/mL) induced a cell death between 70 and 55% depending on D,L-methadone concentration.
  • D,L-methadone increases the effectiveness of doxorubicin in treatment of breast cancer cells, suggesting that a strong reduction of anti-cancer drugs concentrations can be used by cotreatment with D,L-methadone to get comparable cell death rates and therefore less side effects of the anti-cancer drugs will be observed.
  • Glioblastoma cells A172 were treated with different concentrations of doxorubicin alone or in combination with D,L-methadone (hatched columns, white columns). As shown in FIG. 55 , a strong induction of cell death was observed by co-treatment of D,L-methadone and doxorubicin.
  • Glioblastoma cells A172 were treated with doxorubicin (1 ⁇ g/ml) alone. 1 ⁇ g/mL doxorubicin induced cell death of 80% after 144 h. However, treatment with 0.1 ⁇ g/mL doxorubicin in addition to D,L-methadone (10, 3, 1 ⁇ g/mL) induced a cell death of 85%-50% depending on concentrations of D,L-methadone.
  • D,L-methadone increases the effectiveness of doxorubicin in treatment of glioblastoma, suggesting that a strong reduction of anti-cancer drugs concentrations can be used by cotreatment with D,L-methadone to get comparable cell death rates and therefore less side effects of the anti-cancer drugs will be observed.
  • opioids such as D,L-methadone breaks chemoresistance because conventional therapies using anti-cancer drugs are limited by the toxicity of anti-cancer drugs concentrations used for patients treatment.
  • Leukemia cells HL60 were treated with fentanyl (3, 1 ⁇ g/mL) alone (A) or morphine (3, 1 ⁇ g/mL) alone (A) or in combination of fentanyl and morphine (B) at concentrations as indicated.
  • fentanyl 3, 1 ⁇ g/mL
  • morphine 3, 1 ⁇ g/mL
  • B morphine
  • FIG. 56 a strong synergistically increased induction of cell death was observed by co-treatment of morphine and fentanyl (B). This suggests that the combination of different opioids enhances the pro-apoptotic effect and argues for a combined use of opioids also in the combination with a further anticancer agent.
  • the examples provide evidence that D,L-methadone induces apoptosis, activates caspases and increases doxorubicin-induced cell death in leukemia cells depending on opioid-receptor activation inducing the downregulation of cAMP.
  • D,L-methadone can strongly reduce tumour growth of ALL in a xenograft-model in vivo. Noticeably, this tumour-killing effect could be enhanced by the combination of D,L-methadone with the anticancer drug doxorubicin.
  • Methadone is a p-opioid receptor agonist binding to p-opioid receptors if presented on cells. It was found that D,L-methadone kills strongly xenograft-derived ALL-cells expressing high levels of opioid receptors. In contrast, D,L-methadone induces cell death only slightly in xenograft-derived ALL-cells and -cell lines expressing moderate opioid receptor amounts indicating that D,L-methadone-induced apoptosis seems to depend on critical levels of opioid receptor expression in leukemia cells.
  • Combination treatment may prove to be advantageous in malignancies that still partially respond to either treatment alone as different therapeutics are known to interact with each other amplifying weaker death signals.
  • Combination treatment with D,L-methadone and doxorubicin enhances the anti-tumour efficacy of both agents synergistically in BCP-ALL-cells expressing moderate levels of opioid-receptors and increases strongly caspase activation playing a critical role in apoptosis induction in sensitive and resistant cancer cells (Fulda, 2009c).
  • the downregulation of the anti-apoptotic proteins XIAP and Bc1-x L involved in the occurrence of resistances in many malignancies like ALL or NHL is markedly enhanced. This suggests that combination treatment of D,L-methadone and doxorubicin strongly increases apoptosis induction and could improve their anti-tumour efficacy synergistically.
  • Opioid receptors signal by catalysing ligand-dependent nucleotide exchange on G i , thereby inhibiting adenylyl cyclase and modulating N-type calcium channels as well as G protein—gated inwardly rectifying potassium (GIRK)-type potassium channels leading to changes in cell signalling ( FIG. 7 ).
  • GIRK gated inwardly rectifying potassium
  • cancers Broad spectrum of cancers. Several diverse cancer types can be treated with the combination of opioid receptor agonists such as e.g. breast cancer, pancreatic cancer, prostate cancer, ovarian cancer, glioblastoma or leukemia.
  • opioid receptor agonists such as e.g. breast cancer, pancreatic cancer, prostate cancer, ovarian cancer, glioblastoma or leukemia.
  • anticancer drugs For several structurally and pharmacologically distinct anticancer drugs it could be shown that they increase opioid receptor expression and show increased influx/decreased efflux due to the co-applied opioid agonist.
  • opioids In the first path of this loop opioids enhance the cellular uptake and inhibit the efflux of anticancer drugs. On the second path of said loop the accumulating anticancer drugs lead to an increased expression of opioid receptors. Hence, both agents can exert their cytotoxic potential to a higher extent.
  • the present examples could verify the clinical relevance with patient-derived ALL-cells, patient-derived glioblastoma cells and glioblastoma initiating stem cells ex vivo and could show for the first time that D,L-methadone as monotherapy or in combination with doxorubicin leads to a strong tumour growth inhibition in a patient-derived leukemia model and in a glioblastoma xenograft model.
  • the anti-leukemic efficacy, the tumour growth inhibition of glioblastoma and the side effects of D,L-methadone alone or in combination with doxorubicin were comparable with those of doxorubicin alone. However, only the combination treatment was able to achieve a longer lasting growth inhibition.
  • the serum concentrations of methadone in mice correlated with the concentrations showing in vitro cytotoxicity.
  • FIG. 1 D,L-methadone kills ALL cells ex vivo depending on critical levels of opioid receptor expression
  • FIG. 2 Combination treatment with D,L-methadone and doxorubicin induces apoptosis in ALL-cells expressing moderate amounts of opioid receptors
  • FIG. 3 D,L-methadone in combination with doxorubicin restores deficient activation of apoptotic pathways in BCP-ALL-cells expressing moderate amounts of opioid receptors in vitro
  • FIG. 4 Doxorubicin enhances opioid receptor expression whereas D,L-methadone enhances doxorubicin uptake and inhibits its efflux
  • FIG. 5 Combination treatment with D,L-methadone and doxorubicin induced apoptosis depends on opioid-receptor triggering via downregulation of cAMP
  • D,L-methadone was used in weekly increasing doses from 20 up to 120 mg/kg/day and doxorubicin in a dose of 3 mg/kg.
  • FIG. 7 Opioid receptor signaling. Stimulation of opioid receptors (OR) by agonists like D,L-methadone leads to an activation of the inhibitory G i -protein.
  • the ⁇ i -subunit inactivates adenylyl cyclase (AC) resulting in a reduction of cAMP levels within the cell which in turn leads to apoptosis which might be mediated by several different modulators.
  • the ⁇ -subunits of the G i -protein modulate the activity of different effectors like the inhibition of Ca 2+ - and the activation of K + -channels.
  • FIG. 8 Opioid receptor expression on glioblastoma cells.
  • the glioblastoma cell lines U118MG and A172 were stained with naloxone fluorescein measuring opioid receptor expression (OR, thick black curve) and analysed by flow cytometry. Controls (Co) are exhibited as thin black curves.
  • FIG. 9 D,L-methadone sensitizes glioblastoma cells for doxorubicin treatment.
  • Glioblastoma cells A172 were incubated with 3 ⁇ g/mL D,L-methadone alone, with 0.1 ⁇ g/mL doxorubicin (0.1 ⁇ g/mL Doxo) alone or with 3 ⁇ g/mL D,L-methadone in combination with 0.1 ⁇ g/mL doxorubicin (Doxo+D,L-Methadone).
  • Control represents untreated glioblastoma cells.
  • After 144h light microscopy pictures were taken.
  • Cotreatment of A172 with 3 ⁇ g/mL D,L-methadone and 0.1 ⁇ g/mL doxorubicin led to detachment of the cells from the ground, membrane-blebbing and cell-shrinkage.
  • FIG. 10 Combination treatment with D,L-methadone and doxorubicin induces apoptosis in glioblastoma cells.
  • A172 and U118MG glioblastoma cells were treated with different concentrations of D,L-methadone (10, 3, 1 ⁇ g/mL) alone (Medium, white columns), with doxorubicin (0.1 ⁇ g/mL Doxo, black columns) alone or with different concentrations of D,L-methadone (10, 3, 1 ⁇ g/mL) in addition to doxorubicin (0.1 ⁇ g/mL Doxo, black columns).
  • the percentages of apoptotic cells were measured by hypodiploid DNA analysis.
  • the percentage of specific apoptosis was calculated as follows: 100 ⁇ [experimental dead cells (%) ⁇ spontaneous dead cells in medium (%)]/[100% ⁇ spontaneous dead cells in medium (%)]. Columns, mean of triplicates; bars, SD ⁇ 10%. Similar results were obtained in three independent experiments.
  • FIG. 11 D,L-methadone sensitizes primary human glioblastoma cells for doxorubicin treatment.
  • A Primary human glioblastoma cells were stained with naloxone fluorescein measuring opioid receptor expression (OR, thick black curve) and analyzed by flow cytometry. Control (Co) is exhibited as thin black curve.
  • FIG. 12 D,L-methadone sensitizes glioblastoma-initiating stem cells for doxorubicin treatment.
  • A Glioblastoma-initiating stem cells were stained with naloxone fluorescein measuring opioid receptor expression (OR, thick black curve) and analyzed by flow cytometry. Control (Co) is exhibited as thin black curve.
  • FIG. 13 Cell death induction of glioblastoma cells using D,L-methadone and doxorubicin cotreatment depends on caspases activation.
  • D,L-methadone restored deficient caspases activation by doxorubicin in glioblastoma cells.
  • A172 were treated with different concentrations of D,L-methadone (3, 1 ⁇ g/mL) alone, with 0.1 ⁇ g/mL doxorubicin (0.1 ⁇ g/mL Doxo) alone or with different concentrations of D,L-methadone (3, 1 ⁇ g/mL) in addition to doxorubicin (0.1 ⁇ g/mL Doxo).
  • Glioblastoma cells A172 were treated with different concentrations of D,L-methadone (10, 3, 1 ⁇ g/mL) in combination with 0.1 ⁇ g/mL doxorubicin (+0.1 ⁇ g/mL Doxo) in the absence (Medium, black columns) or presence of 50 ⁇ mol/L of zVAD.fmk (white columns, 50 ⁇ mol/L zVAD.fmk). After 120h and 144h, the percentages of apoptotic cells were measured by hypodiploid DNA analysis. The percentage of specific apoptosis was calculated as described in FIG. 1 c . Columns, mean of triplicates; bars, SD ⁇ 10%. Similar results were obtained in three independent experiments.
  • FIG. 14 D,L-methadone reversed deficient activation of apoptosis pathways by doxorubicin in glioblastoma-initiating-stem cells.
  • A Glioblastoma-initiating-stem cells were treated with D,L-methadone (3 ⁇ g/mL) alone, with 0.1 ⁇ g/mL doxorubicin (0.1 ⁇ g/mL Doxo) alone or with D,L-methadone (3 ⁇ g/mL) in addition to doxorubicin (0.1 ⁇ g/mL Doxo).
  • D,L-methadone 3 ⁇ g/mL
  • doxorubicin 0.1 ⁇ g/mL Doxo
  • procaspase-10 Downregulation of procaspase-10 was detected at ⁇ 58 kDa and of procaspase-2 at ⁇ 48 kDa.
  • the active fragment of caspase-9 was detected at ⁇ 37 kDa, the active fragment of caspase-3 at ⁇ 19 kDa and ⁇ 17 kDa and PARP cleavage at ⁇ 85 kDa.
  • Equal protein loading was controlled by anti- ⁇ -actin antibody.
  • FIG. 15 D,L-methadone enhances doxorubicin uptake and inhibits its efflux.
  • A D,L-methadone enhances doxorubicin-accumulation in the glioblastoma cell line A172. A172 were incubated with 0.3 ⁇ g/mL doxorubicin alone or in combination with 10 ⁇ g/mL D,L-methadone. After 4, 8 and 24h incubation the fluorescence intensity of doxorubicin (Doxo) using flow cytometry analysis were determined. In the graphic the relative doxorubicin-uptake is shown. Columns, mean of triplicates; bars, SD ⁇ 10%. Similar results were obtained in three independent experiments.
  • FIG. 16 1 Doxorubicin enhances opioid receptor expression on the cell surface.
  • A The glioblastoma cell line A172 was treated for 106 h with 0.1 ⁇ g/mL doxorubicin. After staining of doxorubicin-treated (doxorubicin) and untreated cells with naloxone-fluorescein (naloxone) relative fluorescence intensities were determined flowcytometrically.
  • FIG. 17 D,L-methadone sensitizes leukemia cancer cells (Nalm-6), pancreatic cancer cells (Colo357) and ovarian cancer cells (A2780) for etoposide or cisplatin treatment.
  • the Nalm6, Colo357 and A2780 cells were treated with different concentrations of D,L-methadone (10, 3, 1 ⁇ g/mL) alone (Medium, white columns), with 0.03 ⁇ g/mL Etoposide or 0.3 ⁇ g/mL cisplatin alone or with D,L-methadone (10, 3, 1 ⁇ g/mL) in addition to 0.03 ⁇ g/mL Etoposide (0.03 ⁇ g/mL Etoposide, black columns) or 0.3 ⁇ g/mL cisplatin (0.3 ⁇ g/mL cisplatin, black columns).
  • FIG. 18 D,L-methadone sensitizes chronic lymphocytic leukemia (CLL) cells for Fludarabine treatment.
  • the CLL cells were treated with different concentrations of D,L-methadone (10, 3, 1 ⁇ g/mL) alone (Medium, white columns), with 0.1 ⁇ M Fludarabine (0.1 ⁇ M Fludarabine, black columns) alone or with D,L-methadone (30, 10, 5, 3, 1, 0.5, 0.3, 0.1 ⁇ g/mL) in addition to 0.1 ⁇ M Fludarabine (0.1 ⁇ M Fludarabine, black columns).
  • D,L-methadone 30, 10, 5, 3, 1, 0.5, 0.3, 0.1 ⁇ g/mL
  • the percentages of apoptotic cells were measured by hypodiploid DNA analysis. The percentage of specific apoptosis was calculated as described in FIG. 1B . Columns, mean of triplicates; bars, SD ⁇ 10%. Similar results were obtained in three independent experiments.
  • FIG. 19 Cisplatin enhances opioid receptor expression in HL60 cells
  • Cisplatin enhances opioid receptor expression on the surface of the promyelocytic leukemia cell line HL60.
  • the HL60 cell line was treated for 24h with 0.3 ⁇ g/mL cisplatin. After staining of cisplatin-treated (+Cisplatin) and untreated cells ( ⁇ Cisplatin) with naloxone-fluoresceine relative fluorescence intensities were determined flowcytometrically. 2.1-fold increase in opioid receptor expression is shown after subtracting the cells autofluorescence.
  • FIG. 20 The Her2/neu-resistent mamma carcinoma cell line JIMT-1 expresses high levels of the p-opioid receptor
  • the human JIMT-1 cell line derived from a pleural metastasis of a 62-year old patient with breast cancer who was clinically resistant to Herceptin displays opioid-receptors on its cell surface. JIMT-1 cells were stained with naloxone-fluorescein measuring opioid-receptor expression (OR, thick black curve) and analyzed by flow cytometry. Controls (Co) are exhibited as thin black curves.
  • FIG. 21 Cell cycle analysis and apoptosis of the Her2/neu-resistent mamma carcinoma cell line JIMT-1 treated with a combination of D,L-methadone and doxorubicin.
  • the human JIMT-1 cell line was treated with 1, 3 or 10 ⁇ g/mL of methadone alone (1, 3, 10 Met ⁇ Doxo), with 0.015 ⁇ g/mL of doxorubicin (Doxo) or a combination of 0.015 ⁇ g/mL of doxorubicin with 1, 3 or 10 ⁇ g/mL of methadone (1,3,10 Met+Doxo).
  • a FACS analysis of the cells revealed that the combination of both substances dose-dependently increased apoptosis of the JIMT-1 cells at 96 hours after treatment.
  • B A FACS analysis performed at 96 hours (left side of the figure) and 120 hours (right side of the figure) after treatment showed further increased levels of apoptosis due to combination treatment.
  • doxorubicin 0 or 0,015 ⁇ g/ml doxorubicin (black bars), 1 ⁇ g/mL methadone plus 0 or 0,015 ⁇ g/ml doxorubicin (white bars), 3 ⁇ g/mL methadone plus 0 or 0,015 ⁇ g/ml doxorubicin (hatched bars), 10 ⁇ g/mL methadone plus 0 or 0,015 ⁇ g/ml doxorubicin (doted bars).
  • FIG. 22 Cell death induction of JIMT-1 cells using D,L-methadone and doxorubicin cotreatment depends on caspases activation. Inhibition of caspase activation with the broad spectrum caspase inhibitor zVAD.fmk blocks apoptosis induced by cotreatment of D,L-methadone and doxorubicin in JIMT-1 cells.
  • the human cell line JIMT-1 was treated with different concentrations of D,L-methadone (10, 3, 1 ⁇ g/mL) in combination with 0.015 ⁇ g/mL doxorubicin (+0.015 ⁇ g/mL Doxo) in the absence (left sided diagrams) or presence of 50 ⁇ mol/L of zVAD.fmk (diagrams on the right side).
  • D,L-methadone 10, 3, 1 ⁇ g/mL
  • doxorubicin (+0.015 ⁇ g/mL Doxo
  • the cells were analysed using flow cytometry.
  • FIG. 23 Cell death induction of JIMT-1 cells using D,L-methadone and doxorubicin cotreatment depends on caspases activation.
  • D,L-methadone restored deficient caspases activation by doxorubicin in JIMT-1.
  • A172 were treated with different concentrations of D,L-methadone (10, 3, 1 ⁇ g/mL) alone, with 0.015 ⁇ g/mL doxorubicin alone or with different concentrations of D,L-methadone (10, 3, 1 ⁇ g/mL) in addition to 0.015 ⁇ g/ml doxorubicin.
  • After 96h Western blot analyses for caspase--8, -9, -3 and PARP were performed.
  • the active fragment of caspase-8 was detected at ⁇ 43 kDa, the active fragment of caspase-9 was detected at ⁇ 37 kDa, the active fragment of caspase-3 at ⁇ 17 kDa and PARP cleavage at ⁇ 85 kDa.
  • Equal protein loading was controlled by anti- ⁇ -actin antibody.
  • B Downregulation of XIAP and Bcl-x L in JIMT-1 cells by using D,L-methadone in combination with doxorubicin.
  • Mamma carcinoma cells JIMT-1 were treated with different concentrations of D,L-methadone (10, 3, 1 ⁇ g/mL) alone, with 0.015 ⁇ g/mL doxorubicin alone or with D,L-methadone (10, 3, 1 ⁇ g/mL) in addition to doxorubicin (0.015 ⁇ g/mL Doxo) for 96h.
  • Western blot analyses for XIAP and Bcl-x L were performed. XIAP was detected at 57 kDa and Bcl-x L was detected at 21 kDa. Equal protein loading was controlled by anti- ⁇ -actin antibody.
  • FIG. 24 Induction of apoptosis in T-Cell leukemia CEM cells by a combination of doxorubicin and fentanyl.
  • Human T-Cell leukemia CEM cell line (10000 cells/100 ⁇ l) were treated with 30, 10, 5, 3, 1, 0.5, 0.3, 0.1 ⁇ g/mL fentanyl alone (white bars) or in addition to 0.02 ⁇ g/mL doxorubicin (black bars). After 48h and 72h quantification of apoptosis was measured by flow cytometry.
  • FIG. 25 Induction of apoptosis in human acute myeloid leukemia HL-60 cells by a combination of doxorubicin and buprenorphine.
  • Human acute myeloid leukemia HL-60 cell line (5000 cells/100 ⁇ l) were treated with 20, 10, 5, 3, 1, 0.5, 0.3, 0.1 ⁇ g/mL buprenorphine alone (white bars) or in addition of 0.003 ⁇ g/mL doxorubicin (black bars). After 144h or 168h quantification of apoptosis was measured by flow cytometry.
  • FIG. 26 Schematic diagram showing the mutual positive interaction between opioids and anticancer drugs.
  • opioids enhance the cellular uptake and inhibit the efflux of anticancer drugs.
  • anticancer drugs lead to an increased expression of opioid receptors. Hence, both agents can exert their cytotoxic potential to a higher extent.
  • FIG. 27 Opioid receptor expression on different leukemia
  • leukemia cells human T cell leukemia, human acute myeloid leukemia, human B cell precursor leukemia and human B cell leukemia express different moderate number of opioid-receptors on their cell surface.
  • Leukemia cells were stained with naloxone-fluoresceine measuring opioid-receptor expression (OR, thick black curve) and analyzed by flowcytometry. Controls (Co) without naloxone are exhibited as thin black curves.
  • FIG. 28 Effect of combination therapy of opioid receptor agonist and anticancer agent
  • D,L-methadone strongly enhances cisplatin sensitivity of different leukemia cells.
  • Different leukemia cells human T cell leukemia, human acute myeloid leukemia, human B cell precursor leukemia and human B cell leukemia
  • D,L-methadone as indicated
  • ⁇ CDDP white columns
  • D,L-methadone in addition to cisplatin (+CDDP, black columns
  • FSC/SSC-analysis The percentage of specific apoptosis was calculated as described in FIG. 1 B. Columns, mean of triplicates; bars, SD ⁇ 10%.
  • FIG. 29 D,L-methadone in combination with cisplatin restores deficient activation of apoptotic pathways in leukemia cells.
  • D,L-methadone and cisplatin co-treatment provokes caspases activation and induces downregulation and cleavage of XIAP and upregulation of Bax.
  • Different leukemia cells human T cell leukemia, human acute myeloid leukemia, human B cell precursor leukemia
  • FIG. 30 D,L-methadone and cisplatin-induced apoptosis depends on caspase activation.
  • Pre-incubation of different leukemia cells human T cell leukemia, human acute myeloid leukemia, human B cell precursor leukemia
  • zVAD.fmk ⁇ zVAD.fmk, black columns
  • Apoptosis induction was detected after time of incubation by FSC/SSC-analysis. The percentage of specific apoptosis was calculated as described in FIG. 1B . Columns, mean of triplicates; bars, SD ⁇ 10%.
  • FIG. 31 Cisplatin enhances opioid receptor expression.
  • Cisplatin enhances opioid receptor expression on the cells' surface of different leukemia cells (human T cell leukemia, human acute myeloid leukemia and human B cell precursor leukemia) were treated with cisplatin (as indicated). After staining of cisplatin-treated cells (CDDP) and untreated cells (Co) with naloxone-fluoresceine relative fluorescence intensities were determined flowcytometrically. X-fold increase in opioid receptor expression compared to the untreated control group is shown after subtracting the cells' autofluorescence and cisplatin fluorescence.
  • FIG. 32 Combination treatment with D,L-methadone and doxorubicin induced apoptosis depends on opioid-receptor triggering. Inhibition of opioid-receptor triggering inhibits apoptosis induction mediated by combination treatment with D,L-methadone and doxorubicin.
  • Glioblastoma cells were incubated with 60 ⁇ g/mL naloxone (Naloxone), 3 ⁇ g/mL D,L-methadone (D,L-Methadone) and 0.1 ⁇ g/mL doxorubicin (Doxo) alone or in different combinations as indicated by the marks+ and ⁇ . . .
  • FIG. 33 Different duration of effectiveness of different opioids induces different rates of cell death in glioblastoma cells.
  • FIG. 34 Different Duration of Effectiveness of Different Opioids Induces Different Rates of Cell Death in Leukemia Cells
  • FIG. 35 Combination treatment with D,L-methadone and doxorubicin inhibits proliferation and induces S/G2-M cell cycle arrest in glioblastoma cells.
  • Flow cytometric analysis of glioblastoma cells treated with methadone and doxorubicin was shown.
  • Flow cytometric analysis of untreated cells (Untreated cells) (G1 peak is higher than G2 peak), cells treated with 1 ⁇ g/ml methadone (Methadone) (G1 peak is higher than G2 peak) and cells treated with methadone in addition to 0,1 ⁇ g/ml doxorubicin (Methadone+Doxo).
  • G1 peak lower than in untreated cells and G2 peak is higher than in untreated cells was shown in glioblastoma cells treated with methadone in addition to doxorubicin after 96h (A).
  • subG1 peak in front of G1 is the fragmentated DNA (percentage of cell death). Results are representative of 3 independent experiments.
  • FIG. 36 Apoptosis induction and caspase activation depend on opioid receptor activation inducing cAMP downregulation in glioblastoma. Opioid receptor activation triggering downregulation of cAMP plays a critical role in sensitizing glioblastoma cells for doxorubicin treatment.
  • A, B Blocking opioid receptor activation inhibits apoptosis induction. Glioblastoma cell line A172 was incubated with 100 ⁇ g/mL naloxone (Naloxone), 3 ⁇ g/mL D,L-methadone (D,L-Methadone) and 0.3 ⁇ g/mL doxorubicin (Doxo) alone or in different combinations as indicated.
  • the active fragment of caspase-9 was detected at ⁇ 37 kDa, of caspase-3 at ⁇ 19 kDa and ⁇ 17 kDa and PARP cleavage at ⁇ 85 kDa. Equal protein loading was controlled by anti- ⁇ -actin antibody.
  • D Increasing cAMP levels via repression of phosphodiesterase activity inhibits apoptosis. A172 cells were incubated for 120h with 25 ⁇ M 3-lsobutyl-1-methylxanthine (IBMX), 3 ⁇ g/mL D,L-methadone (D,L-Methadone) and 0.3 ⁇ g/mL doxorubicin (Doxo) alone or in different combinations as indicated. After 120h, the percentages of apoptotic cells were measured by hypodiploid DNA analysis.
  • IBMX 3-lsobutyl-1-methylxanthine
  • D,L-Methadone 3 ⁇ g/mL D,L-methadone
  • Doxo
  • FIG. 37 D,L-Methadone inhibits tumour growth of glioblastoma.
  • FIG. 38 D,L-methadone sensitizes ovarian cancer cells for short-term cisplatin treatment.
  • A2780 ovarian cancer cells were treated with different concentrations of D,L-methadone (3, 1, 0 ⁇ g/mL) alone, with 5 or 3 ⁇ g/mL cisplatin alone or in combination with D,L-methadone and 5 ⁇ g/mL cisplatin (+5 ⁇ g/ml CDDP, black columns) or 3 ⁇ g/mL cisplatin (+3 ⁇ g/mL, white columns).
  • the percentages of cell death/apoptotic cells were measured.
  • FIG. 39 D,L-methadone sensitizes ovarian cancer cells for long-term cisplatin treatment.
  • A2780 ovarian cancer cells were treated with different concentrations of D,L-methadone (10, 3, 1, 0 ⁇ g/mL) alone, with cisplatin (2, 1, 0.5, 0.3 ⁇ g/mL) alone or in combination with D,L-methadone and cisplatin.
  • cisplatin 2, 1, 0.5, 0.3 ⁇ g/mL
  • 72h, 96h, 120h and 144h the percentages of cell death/apoptotic cells were measured.
  • FIG. 40 D,L-methadone sensitizes cisplatin-resistant ovarian cancer cells for cisplatin treatment.
  • A2780cis ovarian cancer cells were treated with different concentrations of D,L-methadone (10, 3, 1, 0 ⁇ g/mL) alone, with cisplatin (3, 2, 1 ⁇ g/mL) alone or in combination with D,L-methadone and cisplatin.
  • FIG. 41 D,L-methadone strongly sensitizes ovarian cancer cells for cisplatin treatment.
  • A2780 ovarian cancer cells were treated with different concentrations of D,L-methadone (10, 3, 1, 0 ⁇ g/mL) alone, with 2 ⁇ g/mL cisplatin alone (black column) and with 0.5 or 0.2 ⁇ g/mL cisplatin (hatched and grey columns, respectively) in combination with D,L-methadone as indicated. After 144h, the percentages of cell death/apoptotic cells were measured.
  • FIG. 42 Opioid receptor activation using D,L-methadone sensitizes ovarian cancer cells for cisplatin-induced activation of caspases.
  • D,L-Methadone restored deficient caspases activation by cisplatin in ovarian cancer cells.
  • A2780 ovarian cancer cells were treated with different concentrations of D,L-methadone (3, 1 ⁇ g/mL, ⁇ CDDP) alone, with 5 or 3 ⁇ g/mL cisplatin alone or with D,L-methadone (3, 1 ⁇ g/mL) in addition to 5 or 3 ⁇ g/mL cisplatin (+CDDP).
  • Western blot analyses were performed.
  • the active fragment of caspase-9 was detected at ⁇ 37 kDa, the active fragment of caspase-3 at ⁇ 19 and 17 kDa, active fragment of caspase-8 was detected at ⁇ 43 and 41 kDa and PARP cleavage at ⁇ 85 kDa.
  • Equal protein loading was controlled by anti- ⁇ -actin antibody.
  • FIG. 43 Opioid receptor activation using D,L-methadone plays a critical role in sensitizing breast cancer cells for doxorubicin treatment. Blocking opioid receptor activation using opioid antagonist naloxone strongly inhibits apoptosis induction induced by combination treatment with D,L-methadone and doxorubicin.
  • Breast cancer cells JIMT-1 incubated with 100 ⁇ g/mL naloxone (Naloxone), 10 ⁇ g/mL D,L-methadone (D,L-Methadone) and 0.01 ⁇ g/mL doxorubicin (Doxo) alone or in different combinations as indicated. After 96h, the percentages of cell death/apoptotic cells were measured.
  • FIG. 44 D,L-methadone sensitizes prostate cancer cells for cisplatin treatment.
  • Prostate cancer cells PC-3 were treated with different concentrations of D,L-methadone (3, 1 ⁇ g/mL) alone, with 5 or 3 ⁇ g/mL cisplatin alone or in combination with D,L-methadone and 5 ⁇ g/mL cisplatin (+5 ⁇ g/ml CDDP, black columns) or 3 ⁇ g/mL cisplatin (+3 ⁇ g/mL, white columns).
  • the percentages of apoptotic cells were measured by hypodiploid DNA analysis.
  • FIG. 45 D,L-methadone sensitizes leukemia cells for treatment with different anticancer drugs.
  • Leukemia cells Nalm6 were treated with D,L-methadone (3 ⁇ g/mL) alone, with different anticancer drugs alone as indicated (white columns) or in combination with D,L-methadone and different anticancer drugs (black columns). After 96h, the percentages of cell death/apoptosis were measured.
  • FIG. 46 D,L-methadone sensitizes glioblastoma cells for treatment with different anticancer drugs from the same anti-cancer drug class.
  • Glioblastoma cells A172 were treated with D,L-methadone (3 ⁇ g/mL) alone, with different anticancer drugs alone as indicated (white columns) or in combination with D,L-methadone and different anticancer drugs (black columns). After 120h, the percentages of cell death/apoptosis were measured.
  • FIG. 47 Opioid receptor expression on pancreatic cancer cells.
  • Pancreatic cancer cells Colo357 were stained with naloxone fluorescein measuring opioid receptor expression (OR, thick black curve) by flow cytometry. Controls (Co) are exhibited as thin black curves. A strong expression of opioid receptors on the surface of pancreatic cancer was found.
  • FIG. 48 Opioids such as D,L-methadone increase anti-cancer drug-induced cell death in pancreatic cancer cells.
  • Pancreatic cancer cells Colo 357 were treated with D,L-methadone (10, 3, 1, 0 ⁇ g/mL) alone, with oxaliplatin or cisplatin alone as indicated or with a combination of D,L-methadone and (A) oxaliplatin or (B) cisplatin. After 120h (A) and 144 (B), the percentages of cell death/apoptosis were measured.
  • FIG. 49 Opioid receptor activation using D,L-methadone sensitizes pancreatic cancer cells for cisplatin- and oxaliplatin-induced activation of caspases.
  • D,L-Methadone restored deficient caspases activation by (A) oxaliplatin or (B) cisplatin in pancreatic cancer cells.
  • Pancreatic cancer cells Colo 357 were treated with different concentrations of D,L-methadone (3, 1 ⁇ g/mL) alone, with (A) oxaliplatin (2, 3 ⁇ g/mL) alone, or (B) with cisplatin (0.5 or 0.7 ⁇ g/mL) alone or with (A) D,L-methadone (3, 1 ⁇ g/mL) in addition to 2 or 3 ⁇ g/mL oxaliplatin or with (B) D,L-methadone (3, 1 ⁇ g/mL) in addition to 0.5 or 0.7 ⁇ g/mL cisplatin. After 120h, Western blot analyses were performed.
  • the active fragment of caspase-9 was detected at ⁇ 46 kDa, the active fragment of caspase-3 at ⁇ 19 and 17 kDa, and PARP cleavage at ⁇ 85 kDa.
  • the inhibitory protein of caspases XIAP was detected at ⁇ 57 kDa Equal protein loading was controlled by anti- ⁇ -actin antibody.
  • FIG. 50 Inhibition of caspases activation blocks opioid-sensitized pancreatic cancer cells for oxaliplatin- or cisplatin-induced apoptosis.
  • Pancreatic cancer cells Colo 357 were treated with different concentrations of D,L-methadone (10, 3, 1, 0 ⁇ g/mL) in combination with (A) 3 ⁇ g/mL oxaliplatin, (B) 2 ⁇ g/mL oxaliplatin, (C) 0.7 ⁇ g/mL cisplatin or (D) 0.5 ⁇ g/mL cisplatin without ( ⁇ zVAD.fmk, black columns) or with addition of 50 ⁇ mol/L zVAD.fmk (+zVAD, white columns). After 120h and 144h, the percentages of cell death/apoptotic cells were measured.
  • FIG. 51 Opioids using D,L-methadone sensitizes glioblastoma cells for temozolomide treatment.
  • Glioblastoma cells A172 were treated with different concentrations of D,L-methadone 3, 1, 0 ⁇ g/mL) alone, with temozolomide alone or in combination with D,L-methadone and temozolomide (black columns). After 120h, the percentages of apoptotic cells were measured.
  • FIG. 52 Opioids using D,L-methadone strongly sensitizes pancreatic cancer cells for oxaliplatin and cisplatin treatment.
  • FIG. 53 Opioids using D,L-methadone strongly sensitizes leukemia cells for doxorubicin treatment.
  • FIG. 54 Opioids using D,L-methadone strongly sensitizes breast cancer cells for doxorubicin treatment.
  • FIG. 55 Opioids using D,L-methadone strongly sensitizes glioblastoma cells for doxorubicin treatment.
  • Glioblastoma cells (A172) were treated with different concentrations of D,L-methadone (10, 3, 1, 0 ⁇ g/mL) alone, with 1 ⁇ g/ml doxorubicin (black column) alone, with 0.1 ⁇ g/ml doxorubicin alone or with a combination of 0.1 ⁇ g/mL doxorubicin and D,L-methadone (hatched columns) as indicated. After 144h, the percentages of cell death/apoptotic cells were measured.
  • FIG. 56 Combination treatment of morphine and fentanyl shows a strong synergistic effect for inducing cell death in leukemia cells.
  • HL60 leukemia cells were treated with fentanyl (3, 1 ⁇ g/mL) alone (A) or morphine (3, 1 ⁇ g/mL) alone (A) or with a combination of fentanyl and morphine (B) at concentrations as indicated.
  • fentanyl 3, 1 ⁇ g/mL
  • B a combination of fentanyl and morphine

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Toxicology (AREA)
  • Emergency Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biochemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Analytical Chemistry (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Inorganic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
US14/434,138 2012-10-08 2013-10-08 Combination of opioids and anticancer drugs for cancer treatment Abandoned US20150265594A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP12006946.3 2012-10-08
EP12006946 2012-10-08
EP12007179.0 2012-10-17
EP12007179.0A EP2716291B1 (fr) 2012-10-08 2012-10-17 Combinaison d'opioïdes et médicaments anticancéreux pour le traitement du cancer
PCT/EP2013/070923 WO2014056897A1 (fr) 2012-10-08 2013-10-08 Combinaison d'opioïdes et d'anticancéreux pour le traitement du cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/070923 A-371-Of-International WO2014056897A1 (fr) 2012-10-08 2013-10-08 Combinaison d'opioïdes et d'anticancéreux pour le traitement du cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/000,467 Division US11052082B2 (en) 2012-10-08 2018-06-05 Combination of opioids and anticancer drugs for cancer treatment

Publications (1)

Publication Number Publication Date
US20150265594A1 true US20150265594A1 (en) 2015-09-24

Family

ID=47046323

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/434,138 Abandoned US20150265594A1 (en) 2012-10-08 2013-10-08 Combination of opioids and anticancer drugs for cancer treatment
US16/000,467 Active US11052082B2 (en) 2012-10-08 2018-06-05 Combination of opioids and anticancer drugs for cancer treatment

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/000,467 Active US11052082B2 (en) 2012-10-08 2018-06-05 Combination of opioids and anticancer drugs for cancer treatment

Country Status (12)

Country Link
US (2) US20150265594A1 (fr)
EP (1) EP2716291B1 (fr)
JP (1) JP6472750B2 (fr)
CN (1) CN104902897A (fr)
BR (1) BR112015007866A2 (fr)
CA (1) CA2887043C (fr)
DK (1) DK2716291T3 (fr)
ES (1) ES2779223T3 (fr)
PL (1) PL2716291T3 (fr)
PT (1) PT2716291T (fr)
RU (1) RU2015114786A (fr)
WO (1) WO2014056897A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110090303A (zh) * 2019-05-07 2019-08-06 浙江大学 阿片受体激动剂在制造用于治疗恶性肿瘤的药物中的用途
CN110430889A (zh) * 2017-01-20 2019-11-08 堪萨斯大学 降低c-myc蛋白质水平并减慢癌细胞生长的大环肽
US11007135B2 (en) * 2019-05-13 2021-05-18 Parfums de Coeur, Ltd. Liquid cosmetic
CN114113603A (zh) * 2021-06-30 2022-03-01 四川大学华西医院 Cytl1作为胃癌预后标志物的应用
CN117442737A (zh) * 2023-11-01 2024-01-26 河北医科大学口腔医院 α2-肾上腺素受体激动剂与阿片受体激动剂的联合应用

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RS62297B1 (sr) 2011-11-23 2021-09-30 Therapeuticsmd Inc Prirodne kombinovane hormonske supstitucione formulacije i terapije
US9301920B2 (en) 2012-06-18 2016-04-05 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US20130338122A1 (en) 2012-06-18 2013-12-19 Therapeuticsmd, Inc. Transdermal hormone replacement therapies
US10806740B2 (en) 2012-06-18 2020-10-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10806697B2 (en) 2012-12-21 2020-10-20 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US20150196640A1 (en) 2012-06-18 2015-07-16 Therapeuticsmd, Inc. Progesterone formulations having a desirable pk profile
US10568891B2 (en) 2012-12-21 2020-02-25 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11246875B2 (en) 2012-12-21 2022-02-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10537581B2 (en) 2012-12-21 2020-01-21 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US9180091B2 (en) 2012-12-21 2015-11-10 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US10471072B2 (en) 2012-12-21 2019-11-12 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11266661B2 (en) 2012-12-21 2022-03-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
JP2017516768A (ja) 2014-05-22 2017-06-22 セラピューティックスエムディー インコーポレーテッドTherapeuticsmd, Inc. 天然の併用ホルモン補充療法剤及び療法
WO2016090092A1 (fr) * 2014-12-03 2016-06-09 Nemucore Medical Innovations, Inc. Nano-émulsions thérapeutiques pour l'administration à et l'imagerie du cerveau et des procédés de leur préparation et utilisation
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
RU2018133932A (ru) 2016-04-01 2020-05-12 Терапьютиксмд, Инк. Фармацевтическая композиция стероидного гормона
WO2017173044A1 (fr) 2016-04-01 2017-10-05 Therapeuticsmd Inc. Compositions d'hormones stéroïdes dans des huiles à chaîne moyenne
AU2018220903B2 (en) * 2017-02-14 2021-07-15 Research Triangle Institute Proline-based neuropeptide FF receptor modulators
WO2019197015A1 (fr) * 2018-04-09 2019-10-17 Peter Truog Composition comprenant des dérivés d'acide 4-phénylbutyrique et des opioïdes
WO2020239948A1 (fr) 2019-05-28 2020-12-03 Isanas Ag Agoniste du récepteur μ-opioïde à action centrale en association avec un curcuminoïde s'utilisant dans le traitement d'une tumeur du système nerveux central
CN110638802A (zh) * 2019-10-22 2020-01-03 深圳承启生物科技有限公司 Atca在制备治疗肿瘤的药物中的用途

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9814036D0 (en) * 1998-06-29 1998-08-26 Univ Dundee Materials and methods relating to the induction of apoptosis in target cells
US7923454B2 (en) * 2002-05-17 2011-04-12 Jenken Biosciences, Inc. Opioid and opioid-like compounds and uses thereof
EP1732583A4 (fr) * 2004-02-26 2009-08-12 Penn State Res Found Therapie combinatoires pour le traitement des neoplasies a l'aide des recepteurs du facteur de croissance opioide
US8148338B2 (en) * 2006-02-22 2012-04-03 Supratek Pharma Inc. Doxorubicin formulations for anti-cancer use
WO2009117669A2 (fr) 2008-03-21 2009-09-24 The University Of Chicago Traitement avec des antagonistes des opioïdes et des inhibiteurs mtor
DK2149372T3 (da) * 2008-07-31 2014-07-07 Universität Ulm Anvendelse af opioider af metadongruppen til behandling af patienter med resistent cancer

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Lin et al. International J. of Nanomedicine, 2012, pgs. 4169-4183. *
Maneckjee et al. Proc. Natl. Acad. Sci., 1992, Vol. 89, pgs. 1169-1173. *
McSweeney et al. Experimental and Psychopharmacology, 2005, Vol. 13, No. 3, abstract. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110430889A (zh) * 2017-01-20 2019-11-08 堪萨斯大学 降低c-myc蛋白质水平并减慢癌细胞生长的大环肽
CN110090303A (zh) * 2019-05-07 2019-08-06 浙江大学 阿片受体激动剂在制造用于治疗恶性肿瘤的药物中的用途
US11007135B2 (en) * 2019-05-13 2021-05-18 Parfums de Coeur, Ltd. Liquid cosmetic
CN114113603A (zh) * 2021-06-30 2022-03-01 四川大学华西医院 Cytl1作为胃癌预后标志物的应用
CN117442737A (zh) * 2023-11-01 2024-01-26 河北医科大学口腔医院 α2-肾上腺素受体激动剂与阿片受体激动剂的联合应用

Also Published As

Publication number Publication date
CA2887043A1 (fr) 2014-04-17
EP2716291A1 (fr) 2014-04-09
DK2716291T3 (da) 2020-03-02
US11052082B2 (en) 2021-07-06
JP6472750B2 (ja) 2019-02-20
PL2716291T3 (pl) 2020-06-15
CN104902897A (zh) 2015-09-09
US20180280380A1 (en) 2018-10-04
BR112015007866A2 (pt) 2017-08-08
JP2015532298A (ja) 2015-11-09
RU2015114786A (ru) 2016-11-27
CA2887043C (fr) 2021-06-22
PT2716291T (pt) 2020-03-04
WO2014056897A1 (fr) 2014-04-17
ES2779223T3 (es) 2020-08-14
EP2716291B1 (fr) 2019-11-27

Similar Documents

Publication Publication Date Title
US11052082B2 (en) Combination of opioids and anticancer drugs for cancer treatment
Siveen et al. Thymoquinone overcomes chemoresistance and enhances the anticancer effects of bortezomib through abrogation of NF-κB regulated gene products in multiple myeloma xenograft mouse model
Friesen et al. Cell death sensitization of leukemia cells by opioid receptor activation
JP2015532298A5 (fr)
US20030139352A1 (en) Inhibitors of ABC drug transporters in cancer cells
JP2011520846A (ja) 多発性骨髄腫の治療
US20200123153A1 (en) Combinations for immune-modulation in cancer treatment
EP2320900B1 (fr) Opioïdes pour leur utilisation dans le traitement de patients atteints d'un cancer résistant
Muthiah et al. Overcoming P-glycoprotein–mediated drug resistance with noscapine derivatives
ArulJothi et al. Implications of reactive oxygen species in lung cancer and exploiting it for therapeutic interventions
Hu et al. Kuwanon H Inhibits Melanoma Growth through Cytotoxic Endoplasmic Reticulum Stress and Impaired Autophagy Flux
EP3560497A1 (fr) Compositions et procédés de sensibilisation aux médicaments ou d'inhibition d'une cellule cancéreuse
TW201722422A (zh) 用於治療癌症之合理組合療法
Chen et al. SM-1, a novel PAC-1 derivative, activates procaspase-3 and causes cancer cell apoptosis
JP2019511544A (ja) アジア人患者におけるレスミノスタットの医薬用途
EP3463319A1 (fr) Méthodes et compositions permettant d'éradiquer des cellules leucémiques
CN110267538B (zh) 紫苏醇-3-溴丙酮酸缀合物和治疗癌症的方法
TWI548410B (zh) 使用雙功能性之烷化劑及dna修復抑制劑之組合以治療癌症之方法
CN113710670A (zh) 治疗多发性骨髓瘤的方法

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITAET ULM, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FRIESEN, CLAUDIA, DR.;MILTNER, ERICH, DR.;REEL/FRAME:036616/0829

Effective date: 20150526

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION