WO2013127885A1 - A conjugate of methotrexate and hydroxyethyl starch for use in the treatment cancer - Google Patents

A conjugate of methotrexate and hydroxyethyl starch for use in the treatment cancer Download PDF

Info

Publication number
WO2013127885A1
WO2013127885A1 PCT/EP2013/053974 EP2013053974W WO2013127885A1 WO 2013127885 A1 WO2013127885 A1 WO 2013127885A1 EP 2013053974 W EP2013053974 W EP 2013053974W WO 2013127885 A1 WO2013127885 A1 WO 2013127885A1
Authority
WO
WIPO (PCT)
Prior art keywords
mtx
hes
conjugates
drug
conjugate
Prior art date
Application number
PCT/EP2013/053974
Other languages
French (fr)
Inventor
Tomasz GOSZCZYŃSKI
Janusz BORATYŃSKI
Joanna Wietrzyk
Beata FILIP-PSURSKA
Katarzyna KEMPIŃSKA
Original Assignee
Instytut Immunologii i Terapii Doświadczalnej PAN
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 Instytut Immunologii i Terapii Doświadczalnej PAN filed Critical Instytut Immunologii i Terapii Doświadczalnej PAN
Publication of WO2013127885A1 publication Critical patent/WO2013127885A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/61Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule the organic macromolecular compound being a polysaccharide or a derivative thereof
    • 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

Definitions

  • Subject invention concerns conjugates for use in the treatment of cancer.
  • Synthetic and natural polymers including polyvinyl pyrrolidone) (PVP) [4] polyglutamic acid (PGA) [5], poly(malic acid) [6, 7], poly(ethylene glycol) (PEG) [8], N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers [9], proteins [10] and polysaccharides [1 1 ], may be used as macromolecular carriers.
  • PVP polyvinyl pyrrolidone
  • PGA polyglutamic acid
  • PEG poly(ethylene glycol)
  • HPMA N-(2-hydroxypropyl)methacrylamide copolymers
  • proteins [10] and polysaccharides [1 1 ] may be used as macromolecular carriers.
  • Conjugates of polymers with bioactive compounds may be realized for numerous reasons. One of them is the application of the most popular scheme including hydrophilic polymers with covalently bound bioactive molecules via biodegradable linkers. This kind of approach increases drug solubility, but most importantly modifies its pharmacokinetics in an organism, also on a cellular level, often increasing the therapeutic properties of a drug [12-14]. Bio-distribution of conjugates is to a high degree determined by the properties of a carrier and is realized mainly in an active way (a carrier or its fragment is a structure which potentially may selectively interact with receptors or antigens of target cells) or passive one (enhanced vascular permeability and retention - EPR) [15-19].
  • the EPR effect leads to preferential extravasation and accumulation of macromolecular drug-carrier conjugates (of a hydrodynamic diameter of > 3.5 nm [20, 21 ]) in the tissue of a solid tumor.
  • the drug may be released from the conjugate in an intra- or extracellular space [1 ]. Since macromolecules cannot diffuse freely through the cell membrane, the main mechanism of their infiltration inside the cell is endocytosis [22].
  • attention is focused inter alia on elongated half life time in the blood circulation system and accumulation in tumor tissue, at least as an effect of EPR [23].
  • elimination of a carrier/conjugate needs to take place in order to prevent the occurrence over time of negative results from polymers accumulation [23].
  • it is essential that an equilibrium is achieved assuring such a degree of carrier decomposition that allows gratifying therapeutic effects to be obtained with minimum side effects.
  • Drugs attached to a high molecular carrier may exhibit activity in a form attached to a carrier, as a result of carrier degradation [2] and/or dissociation of a bond between a drug and a carrier [22].
  • the last two processes may occur in an enzymatic or non- enzymatic way as determined by physicochemical conditions in the target site, such as temperature, pH or ionic strength.
  • pH in affected tissues may reach the value of 6.5, i.e. as much as one unit below the pH of blood. This is an effect of hypoxia and intensified necrotic cell death [24, 25].
  • pH in endosomes may reach the value of 5.0 [26].
  • Conjugates with a pH-sensitive bond between a drug and a carrier are used for the therapeutic application of the above facts.
  • An example may be a hydrazone bond subject to hydrolysis in pH between 6 and 5 used in conjugates of HPMA and doxorubicin [27]. These conjugates maintain their stability in blood, while with lower pH values accelerated drug release is observed.
  • the data presented found a therapeutic reflection in this case - in the effectiveness of preparation activity in vivo in an animal model [27].
  • Hydroxyethyl starch is a modified polymer for use in plasma volume replacement [28].
  • the substrate for obtaining HES is natural maize or potato starch which is a polymer of glucose with a-1 -4 glycosidic bonds and numerous branching a-1 -6 glycosidic bonds[29].
  • the degree of branching, i.e. rate of a-1 -6 to a-1 -4 bonds, is usually about 1 :20 [29].
  • the physicochemical properties of HES are determined by its average molecular weight (MW), degree of substitution with hydroxyethyl groups (MS) and the ratio of the degree of substitution of particular sites in the glucose unit determined as the ratio of C2 C6 [30].
  • HES accumulation in an organism is positively correlated with the degree of substitution (MS) [31 , 32].
  • MS degree of substitution
  • the detailed fate of HES in an organism depends first of all on its chemical structure [28].
  • HES polymers, especially those of higher molecular weight and higher degrees of substitution (MS), before they are subject to metabolism, are subject to accumulation in numerous tissues (liver, skin, spleen, lungs, kidneys) [33-37]. Polymers differing slightly in terms of structure may demonstrate significantly different accumulation properties [38].
  • a reducing influence was observed of HES polymers on blood platelets and the process of blood coagulation [39-42], the activity of metalloproteinase-9 (MMP-9) [43], and the expression of proinflammatory cytokines [44].
  • MTX belongs to the group of antifoliates - derivatives of folic acid used in medicine [52]. MTX directly inhibits the activity of DHFR, an enzyme which contributes to the initial reduction of foliates [52, 53], and directly or indirectly affects a range of other molecular targets influencing DNA replication and cell proliferation [54]. MTX has found a wide application in cancer disease treatment and is still used both independently and combined with other chemotherapeutic. It is used in the therapy of, inter alia, acute lymphatic leukemia, osteogenic sarcoma, chorionic epithelioma, and in cases of breast, head and neck cancer [55, 56], as well as in the combined therapy of bladder cancer [57].
  • Nanoconjugates are a new class of therapeutic compounds, which may lead to a new therapeutic quality due to the combination of drugs already used in therapy, as well as new innovative substances with high molecular carriers .
  • One way to verify such a thesis may be the application of a model drug, well described in terms of therapeutic activity, as an active substance.
  • the conjugate and hybrid particle method has significant potential and may enable the elimination of numerous negative features of low molecular drugs.
  • the subject of the invention is a conjugate composed of a carrier and a drug covalently bound thereto, defined by the general formula:
  • n is an integer from 60 to 60000
  • R1 denotes a substituent selected from a group encompassing: hydrogen, - (CH 2 ) n OH, -Drug, -(CH 2 ) n O-Drug,
  • R2 denotes a substituent selected from a group encompassing: hydrogen, - (CH 2 ) n OH, -Drug, -(CH 2 ) n O-Drug, glucose unit,
  • the carrier contains starch with a molecular mass from 10 to 1000 kDa and a degree of substitution with hydroxyalkyl, preferably hydroxyethyl, groups from 0.1 to 0.9 mol/glucose unit, and as the drug it contains a molecule of a known antiinflammatory or anticancer drug which possesses a free carboxyl group, particularly methotrexate,
  • coniugate according to invention may be used in the treatment of cancer, wherein a greater inhibition of tumour growth is attained than when the same amount of said anticancer drug is used in unconiugated form.
  • the effective dose should be understood as a dose of the anticancer drug which in a given patient can elicit the desirable effect, in particular the retardation or inhibition of tumour cell growth.
  • the effective conjugate dose will be at least twofold smaller than the dose of the unconjugated drug.
  • This effect may be observed as a retardation of tumour growth, which may be determined by observing changes in its volume and/or mass.
  • Subject invention discloses application of starch derivatives as effective, high molecular carriers for anticancer, therapeutic substances (e.g. antifoliates).
  • Nanoparticles composed of a starch derivative with a covalently bound anticancer drug (ester bond) were obtained.
  • nanoparticles were obtained of physicochemical properties differing with respect to the carrier.
  • These nanoparticles offered valuable therapeutic features and were composed of two medical substances, which are allowed and currently widely used in medicine.
  • Physicochemical parameters characterizing the changes with respect to the initial polymer were analyzed, as were the biological implications.
  • Biological examinations were performed in vitro with the application of mouse and human cancer cell lines. The anticancer effect in vivo was assessed by testing the activity on animals of lowered immunity enabling examinations with the application of human cancer line cells.
  • the applied solution caused a considerable potentiation (a 10-fold increase in effectiveness when mean volumes of tumors were compared) of the anticancer activity of the conjugates after one intravenous administration of a preparation during a period of advanced cancer progression.
  • the present invention opens the way for the application of modified starch as a drug carrier, especially for anticancer treatments.
  • Figure 5 Debye plot of HES 130/0.4 and HES-MTX conjugates.
  • FIG. 8 Tumor growth kinetics of MV-4-1 1 bearing NOD/CDID mice.
  • the mice were treated either with HES-MTX 130/0.4/59 conjugate (red line) or MTX (green line) alone.
  • the control group received saline (black).
  • Data are presented as mean tumor volume[nnnn 3 ] ⁇ standard deviation.
  • the HES-MTX 130/0.4/59 conjugate diminishes the MV-4-1 1 tumor growth significantly when compared to control or MTX treated group.
  • FIG. 9 Tumor growth kinetics of MV-4-1 1 bearing NOD/CDID mice, treated either with HES-MTX 130/0.4/59 (red), HES-MTX 130/0.4/25 (gray-red) or HES-MTX 130/0.4/1 1 (blue line) conjugate . Data are presented as mean tumor volume [mm 3 ] ⁇ standard deviation.
  • FIG. 10 Tumor growth kinetics of MV-4-1 1 bearing NOD/CDID mice.
  • the mice were treated either with HES-MTX 130/0.4/59 (red line), HES-MTX 130/0.4/25 (gray- red line) or HES-MTX 130/0.4/1 1 (blue line) conjugate, or MTX (green line) alone.
  • the control group received saline (black). Data are presented as mean tumor volume[mm 3 ] ⁇ standard deviation.
  • the HES-MTX 130/0.4/59 conjugate diminishes the MV-4-1 1 tumor growth significantly when compared to control or MTX treated group.
  • Figure 1 1 The influence of HES-MTX 130/0.4/52 conjugate on the tumor growth kinetics of human leukemia MV-4-1 1 bearing mice. The tumor volumes are presented as % of control.
  • FIG. 12 The influence of HES-MTX conjugates on the tumor growth kinetics of human leukemia MV-4-1 1 bearing mice. The tumor volumes are presented as % of control.
  • FIG. 13 Body weight changes of MV-4-1 1 bearing NOD/CDID mice.
  • the mice were treated either with HES-MTX 130/0.4/59, HES-MTX 130/0.4/48, HES-MTX 200/0.5/23, HES-MTX 130/0.4/25 or HES-MTX 130/0.4/1 1 conjugates, or MTX and 3% HES alone.
  • the control group received saline (blue line). Data are presented as mean body weight [g] ⁇ standard deviation. No body weight loss was observed during the experiment.
  • HES in aqueous solutions (6% w/w) was cooled to 4°C in ice bath and pH was adjusted to 10.5 using 1 M solution of Na2CO3. Afterwards cold solution was reacted with MTX anhydride according to the method previously described [1 1]. During conjugation reaction pH was still maintained to 10.5. After adding the total amount of MTX anhydride the pH was immediately adjusted to 7.0 (HCI 1 M) and obtained conjugate were dialyzed against ultrapure water to remove free MTX (dialysis membrane Visking 12kDA (Serva GmbH) for 96 hours or Pellicon® XL (Millipore Corp.) for 3 hours with flow rate 15ml/min). Conjugates with different levels of substitution MTX were synthesized by the same procedure using only a different (HES:MTX anhydride) stoichiometric ratio.
  • the stability of the prepared conjugates was assessed under different conditions and the compounds were stored at -20°C.
  • MS and SL is expressed as number of moles of MTX/hydroxyethyl groups per mole of the glucose unit.
  • Determination of unbound MTX was based on size exclusion chromatography with UV-VIS detection at a wavelength of 302 nm.
  • a Superdex® Peptide column (150 x 4.6 mm) and mobile phase 100 mM sodium bicarbonate with a flow rate of 400 ⁇ / ⁇ was applied [75].
  • the total glucose contents were determined by the phenol-sulphuric acid method[76] with slight modifications.
  • Conjugates were dissolved in a series of various inorganic buffers at pH from 4 to 10 and ionic strength from 10 to 600 mM to a final concentration of 0.8mM (based on the contents of the MTX in the conjugate).
  • the buffer solutions were incubated at 4°C, 20°C and 37°C. At selected time intervals each reaction solution was diluted with NaHCO 3 0.1 M to final concentration 0.2mM (total MTX) and analyzed of unbound MTX[75].
  • Conjugates were dissolved in human plasma to a final concentration of 0.8mM (based on the contents of the MTX in the conjugate). The solutions were incubated at 37°C. At selected time intervals each reaction solution was diluted with NaHCO3 0.1 M to final concentration 0.2mM (total MTX) and analyzed of unbound MTX[75]. DLS and SLS measurements
  • the human biphenotypic B myelomonocytic leukemia MV-4-1 1 cells, human chronic myelogenous leukemia K562 cells, human erythroleukemia HEL92 and Human T lymphoblast CCRF-CEM were obtained from American Type Culture Collection (ATCC, Rockville, MD, USA). The cells were maintained in RPMI-1460 GlutaMAX adjusted to contain 2 mM glutamine and 1 .0 mM sodium pyruvate with fetal bovine serum (10%).
  • Murine leukemia P388 were obtained from the American Type Culture Collection (Rockville, MD, USA) and were maintained in culture or were frozen in the Cell Culture Collection of the Institute of Immunology and Experimental Therapy, Wroclaw, Tru.
  • the cells were plated in 96-well plates (Sarstedt, Germany) at a density of 0.5x10 4 cells per well and were cultured in a mixture of RPMI 1640 and Opti-MEM (1 :1 ) medium, supplemented with 2 mM glutamine (Sigma-Aldrich Chemie GmbH, Germany), 100 mg/ml streptomycin (Polfa, Tarchomin, Tru) and 100 U/ml penicillin (Polfa), 5% fetal bovine serum (Sigma-Aldrich Chemie GmbH, Germany). The cells were cultured at 37°C in a humid atmosphere saturated with 5% CO 2 . Passages of P388 leukemia cells in BDF mice were carried out according to the NIH/NCI standard screening protocols for in vivo assessment [77, 78]
  • mice Male B6D2F1 mice, aged 16-20 weeks were used. The mice were supplied from the Animal Breeding Centre of the Medical Academy, Wroclaw, Tru, and were maintained in standard laboratory conditions. Experiments were performed according to the Guide for the Care and Use of Laboratory Animals (National Academy of Science, National Academy Press, Washington, D.C.) and were approved by the First Local Ethical Committee for the use of Laboratory Animals, Wroclaw, Tru. Male NOD/SCID mice, aged 8- 12 weeks old were used, weighing 20-25 g. The mice were supplied from the JU Children's University Hospital Krakow, Poland, were maintained in standard laboratory conditions. All experiments were performed according to Interdisciplinary Principles and Guidelines for the Use of Animals in Research, (National Academy of Science, National Academy Press, Washington, D.C.). Ad Hoc Committee on Animal Research and were approved by the 1 st Local Committee for Experiments with the Use of Laboratory Animals, Wroclaw, Tru. Design of the in vivo experiments.
  • mice were injected with 10 6 leukemia (P388) cells i.p. (day 0), and 24 h later (day 1 ) each mouse was injected once i.p. with the appropriate agent. All doses (4(Vmol/kg) were based on the contents of the MTX in the conjugate. Body weight and survival data were collected on a daily basis throughout the duration of the experiment.
  • mice were subcutaneously inoculated in the right flank of the abdomen with 7.5-8x10 6 viable MV-4-1 1 tumor cells per mouse in 0.2 ml Hanks solution (Sigma) and when the tumors become measurable (60-100mm 3 ), mice were randomly divided into three (2 nd experiment) or eight (3 rd experiment) groups receiving different HES-MTX conjugates (HES-MTX 130/0.4/52 in 2 nd experiment and all other conjugates in 3 rd experiment) or HES, MTX or 0.9% saline solution alone. All doses (40 ⁇ ) were based on the contents of the MTX in the conjugate. Body weight data were collected on a daily basis throughout the duration of the experiment. At the end of the experiment blood was harvested before subsequent sacrifice of the mice and tumors were collected during autopsy for further analysis.
  • HES-MTX 130/0.4/52 in 2 nd experiment and all other conjugates in 3 rd experiment HES, MTX or 0.9% saline solution alone. All doses (40 ⁇ )
  • IC50 is the concentration of a tested agent which inhibits the proliferation of 50% of the cancer cell population. Average IC50 values for each preparation were calculated using data from at least three independent experiments.
  • Experiment 1 The antileukemic effect in vivo was evaluated as the increase in lifespan (ILS) of treated mice over the control, calculated from the following formula: (MSTT/MSTC) x 100-100, where MSTT is the median survival time of treated animals and MSTC is the median survival time of untreated control mice.
  • the zeta potential of all examined HES-MTX conjugates in water solution receives negative values.
  • the zeta potential value decreases with an increase in the degree of SL substitution, and its distribution is subject to narrowing.
  • Table 3 Hydrodynamic parameters of HES-MTX conjugates.
  • ZP - Zeta potential, Conjugates concentration - 5.5mM, solution - phosphate buffer 10mM pH 7.20.
  • IC50 values for the examined conjugates were about 100 nM (10 nM for free MTX) in the case of MV4-11 line, and within the range of 65-124 nM (7.7 nM for free MTX) in the case of the K562 line. All HES-MTX preparations demonstrated lower in vitro activity when compared to free MTX.
  • HES-MTX 130/0.4/52 preparation elongated the life time of females by 66% when compared to the life time of control mice, while the life time of males was elongated by 45.2% when compared to males in the control group.
  • the life time length of mice in groups treated with the HES-MTX 130/0.4/52 preparation exceeded the life time of mice treated with MTX.
  • HES-MTX 130/0.4/52 conjugate was characterized by a higher than free MTX effectiveness in the inhibition of the growth of experimental MV4- 1 tumors (
  • the control group received saline.
  • the tumor volumes are presented as mean ⁇ standard deviation, % tumor growth inhibition TGI, N- number of mice per group.
  • mice were treated with HES-MTX conjugates, or MTX and 3% HEST alone.
  • the control group received saline.
  • the tumor volumes are presented as mean ⁇ standard deviation, % tumor growth inhibition TGI, N- number of mice per group.
  • HES-MTX 130/0.4/59 and HES-MTX 130/0.4/48 conjugates demonstrated the highest activity among all the examined conjugates (Fig. 8). Both conjugates reduce the volume of MV4-11 tumors to a considerable degree (p ⁇ 0.05) when compared to the control starting from day 13 up to day 29. Comparing the size of tumors in the group which was administered the HES-MTX 130/0.4/59 preparation in one dose, to the size of tumors in groups receiving MTX, significant differences in the size of tumors in days 15 to 29 were noted, while in the HES-MTX 130/0.4/48 group on days 23 to 29 (p ⁇ 0.05) when compared to MTX.
  • HES-MTX 200/0.5/23, 130/0.4/25 and 130/0.4/11 demonstrated a poorer effect in terms of the inhibition of tumor growth when compared to HES-MTX 130/0.4/59 and 130/0.4/48 conjugates (Fig. 9, Fig. 10); however, their activity was higher than those of free MTX (Fig. 1 1 , Fig. 12).
  • TGI parameter tumor growth inhibition
  • mice Based on the analysis of changes in the body weight of mice, no toxicity was observed in the accepted scheme of preparations administration. The average body weight of mice during preparation administration was not lower than on the first day of treatment in any of the groups observed (Fig. 13).
  • HES-MTX conjugates contained hydroxyethyl starch as a carrier substance (HES 130/0.4 and HES 200/0.5). Modifications to HES using MTX were conducted using the anhydride method. The linker between the drug and the polymer was thus glutamic acid (an integral part of the MTX particle) and the connecting bond was an ester bond. Consequently, the following facts should be taken into consideration:
  • HES-MTX conjugates are composed of two medical substances, certified and currently widely used in medicine,
  • esterification is an equilibrium reaction, and a by- product of this reaction is water - during drug release from a conjugate (hydrolysis of esters) we deal with the reconstruction of the substrates of the reaction, i.e. we obtain free MTX and non-modified HES,
  • An additional mechanism of MTX release from a conjugate is HES-MTX hydrolysis by a-amylases. Conjugate subjected to partial enzymatic degradation may exhibit profitable properties. Moreover, digestion of the conjugate may limit MTX release in sites of lowered pH.
  • Enzymatic HES hydrolysis may also be caused by the fact that no toxicity of HES- MTX preparations was observed in in vivo examinations. They are subject, as in the case of HES application in volume therapy, to gradual degradation and removal from an organism.
  • HES-MTX conjugates are characterized by insignificantly higher hydrodynamic diameters when compared to initial HES
  • HES-MTX conjugates are characterized by insignificantly higher polydispersity when compared to initial HES
  • conjugates when compared to initial HES, conjugates are characterized by negative zeta potential of a value and distribution dependent on the degree of MTX polymer substitution,
  • HES-MTX conjugates are characterized by a quite different Debye curve course in comparison to initial HES. This may prove the fact that intermolecular conjugate-conjugate and conjugate-environment interactions are of a different character from interactions between non-modified HES, which may influence the adhesion and distribution of conjugates in an organism.
  • hybrid HES-MTX molecules demonstrate differences in hydrodynamic and electric parameters when compared to non-modified HES polymers. The differences centre on hydrodynamic diameter, polydispersity, charge of molecules and interaction with other molecules of the environment.
  • polymer therapeutics includes natural and synthetic macromolecules used in interdisciplinary studies combining polymer chemistry, biology, pharmacy and medicine for the design of biologically active polymer-drug, polymer-protein conjugates or polymeric micelles [79]. Many polymers with attractive physicochemical features have no possibility to become effective drug carriers due to their biological properties, such as toxicity or immunogenicity [80]. Therefore, selection or design of carrier polymers is a key issue, limiting the process of conjugate formation. Similarly, a significant element is the suitable selection of the linker connecting a polymer with a drug. If it is too stable or not subject to a biodegradation bond connecting the drug with a carrier, it would result in limited drug release and its insufficient concentration in a target site.
  • Ringsdorfs idealistic concept [84], and elements connected to the influence of endocytosis on conjugate behavior in an organism [85] may be useful in the design of polymeric drug-carrier conjugates.
  • Desirable properties of polymers used for the design of therapeutic conjugates are as follows [13, 79, 88]:
  • linkers connecting drugs with polymers should be stable in conditions of conjugate transport and ensure effectiveness in a target site
  • HES polymers as anticancer drug carriers creates large possibilities.
  • an initial polymer naturally starch
  • properties mean molecular weight, number of a-1 ,6- glucoside bonds.
  • These polymers may be designed according to established parameters critical for biological properties, such as MW, MS and ratio of C 2 /C6, with respect to a specified therapeutic substance.
  • the degree of substitution (MS) and its localization may be regulated precisely in the process of modification with hydroxyethyl groups. The whole process may be led in a way enabling the acquisition of a polymer of designed properties adequate to the specified biological targets.
  • HES demonstrates practically all the above-presented desirable properties of polymers used for the design of therapeutic conjugates, and additionally:
  • drugs subjected to conjugation may be any biologically active particle possessing a free carboxyl group (in particular MTX), even if this group is essential for the biological activity of this particle,
  • conjugates may be composed of two or more biologically active substances (including diagnostic substances) revealing new physicochemical and biological properties after conjugation.
  • HES hydroxyethyl starch

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

Conjugate composed of a carrier and a drug covalently bound thereto, for use in the treatment of cancer, wherein the effective dose of anticancer drug used in the form of said conjugate is lower than effective dose of said anticancer drug in unconjugated form.

Description

A CONJUGATE OF METHOTREXATE AND HYDROXYETHYL STARCH FOR
USE IN THE TREATMENTCANCER
Subject invention concerns conjugates for use in the treatment of cancer.
Most currently used medicines belong to the group of low molecular compounds [1 ]. The disadvantages of such kinds of therapeutics include: fast metabolism and excretion from an organism, unprofitable bio-distribution and low selectivity [2]. One way to solve these problems is to combine low molecular therapeutic substances with high molecular carriers [3]. This kind of approach enables an improvement in the pharmacokinetic properties and bio-distribution of both known and innovative drugs. Synthesis of such kinds of hybrid systems is one of the ways to realize the "magic bullet" concept propounded at the beginning of the 20th century by Paul Ehrlich, and which was aimed at achieving the selective deposition and release of therapeutic substances in target tissues [1 ]. This idea was developed and specified in 1975 by Helmut Ringsdorf. Synthetic and natural polymers, including polyvinyl pyrrolidone) (PVP) [4] polyglutamic acid (PGA) [5], poly(malic acid) [6, 7], poly(ethylene glycol) (PEG) [8], N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers [9], proteins [10] and polysaccharides [1 1 ], may be used as macromolecular carriers.
Conjugates of polymers with bioactive compounds may be realized for numerous reasons. One of them is the application of the most popular scheme including hydrophilic polymers with covalently bound bioactive molecules via biodegradable linkers. This kind of approach increases drug solubility, but most importantly modifies its pharmacokinetics in an organism, also on a cellular level, often increasing the therapeutic properties of a drug [12-14]. Bio-distribution of conjugates is to a high degree determined by the properties of a carrier and is realized mainly in an active way (a carrier or its fragment is a structure which potentially may selectively interact with receptors or antigens of target cells) or passive one (enhanced vascular permeability and retention - EPR) [15-19].
The EPR effect leads to preferential extravasation and accumulation of macromolecular drug-carrier conjugates (of a hydrodynamic diameter of > 3.5 nm [20, 21 ]) in the tissue of a solid tumor. The drug may be released from the conjugate in an intra- or extracellular space [1 ]. Since macromolecules cannot diffuse freely through the cell membrane, the main mechanism of their infiltration inside the cell is endocytosis [22]. In the design of high molecular drug-carrier conjugates, attention is focused inter alia on elongated half life time in the blood circulation system and accumulation in tumor tissue, at least as an effect of EPR [23]. However, elimination of a carrier/conjugate needs to take place in order to prevent the occurrence over time of negative results from polymers accumulation [23]. As in many biological systems, it is essential that an equilibrium is achieved assuring such a degree of carrier decomposition that allows gratifying therapeutic effects to be obtained with minimum side effects.
Pharmacokinetic properties and conjugate bio-distribution are also affected by the charge, conformation, hydrophobicity and immunogenicity of the carrier [22].
Drugs attached to a high molecular carrier may exhibit activity in a form attached to a carrier, as a result of carrier degradation [2] and/or dissociation of a bond between a drug and a carrier [22]. The last two processes may occur in an enzymatic or non- enzymatic way as determined by physicochemical conditions in the target site, such as temperature, pH or ionic strength.
The value of pH in affected tissues, especially cancerous ones, may reach the value of 6.5, i.e. as much as one unit below the pH of blood. This is an effect of hypoxia and intensified necrotic cell death [24, 25]. In the case of endocytosis, pH in endosomes may reach the value of 5.0 [26]. Conjugates with a pH-sensitive bond between a drug and a carrier are used for the therapeutic application of the above facts. An example may be a hydrazone bond subject to hydrolysis in pH between 6 and 5 used in conjugates of HPMA and doxorubicin [27]. These conjugates maintain their stability in blood, while with lower pH values accelerated drug release is observed. The data presented found a therapeutic reflection in this case - in the effectiveness of preparation activity in vivo in an animal model [27].
Hydroxyethyl starch is a modified polymer for use in plasma volume replacement [28]. The substrate for obtaining HES is natural maize or potato starch which is a polymer of glucose with a-1 -4 glycosidic bonds and numerous branching a-1 -6 glycosidic bonds[29]. The degree of branching, i.e. rate of a-1 -6 to a-1 -4 bonds, is usually about 1 :20 [29]. The physicochemical properties of HES are determined by its average molecular weight (MW), degree of substitution with hydroxyethyl groups (MS) and the ratio of the degree of substitution of particular sites in the glucose unit determined as the ratio of C2 C6 [30]. HES accumulation in an organism is positively correlated with the degree of substitution (MS) [31 , 32]. The detailed fate of HES in an organism depends first of all on its chemical structure [28]. HES polymers, especially those of higher molecular weight and higher degrees of substitution (MS), before they are subject to metabolism, are subject to accumulation in numerous tissues (liver, skin, spleen, lungs, kidneys) [33-37]. Polymers differing slightly in terms of structure may demonstrate significantly different accumulation properties [38]. A reducing influence was observed of HES polymers on blood platelets and the process of blood coagulation [39-42], the activity of metalloproteinase-9 (MMP-9) [43], and the expression of proinflammatory cytokines [44].
There are literature reports concerning HES conjugates with polypeptides and peptides, especially with erythropoietin (EPO) [45] [46-48], deferoxamine [49, 50]. Also, the application of HES for enzymatic synthesis of polymer-drug and polymer- protein nanoconjugates has been described in the literature [51 ].
MTX belongs to the group of antifoliates - derivatives of folic acid used in medicine [52]. MTX directly inhibits the activity of DHFR, an enzyme which contributes to the initial reduction of foliates [52, 53], and directly or indirectly affects a range of other molecular targets influencing DNA replication and cell proliferation [54]. MTX has found a wide application in cancer disease treatment and is still used both independently and combined with other chemotherapeutic. It is used in the therapy of, inter alia, acute lymphatic leukemia, osteogenic sarcoma, chorionic epithelioma, and in cases of breast, head and neck cancer [55, 56], as well as in the combined therapy of bladder cancer [57].
There have been numerous scientific reports concerning high molecular conjugates containing MTX such as MTX conjugates with albumins [58-63], fibrinogen [64] and glycated fibrinogen [10], mannan [65], dextranes [1 1 , 66] [67-69], hyaluronic acid [70, 71 ] dendrimers [72, 73], and nanotubes [74].
Nanoconjugates are a new class of therapeutic compounds, which may lead to a new therapeutic quality due to the combination of drugs already used in therapy, as well as new innovative substances with high molecular carriers . One way to verify such a thesis may be the application of a model drug, well described in terms of therapeutic activity, as an active substance. The conjugate and hybrid particle method has significant potential and may enable the elimination of numerous negative features of low molecular drugs.
The subject of the invention is a conjugate composed of a carrier and a drug covalently bound thereto, defined by the general formula:
Figure imgf000005_0001
where:
m is an integer from 60 to 60000,
R1 denotes a substituent selected from a group encompassing: hydrogen, - (CH2)nOH, -Drug, -(CH2)nO-Drug,
R2 denotes a substituent selected from a group encompassing: hydrogen, - (CH2)nOH, -Drug, -(CH2)nO-Drug, glucose unit,
wherein as the carrier, it contains starch with a molecular mass from 10 to 1000 kDa and a degree of substitution with hydroxyalkyl, preferably hydroxyethyl, groups from 0.1 to 0.9 mol/glucose unit, and as the drug it contains a molecule of a known antiinflammatory or anticancer drug which possesses a free carboxyl group, particularly methotrexate,
for use in the treatment of cancer, wherein the effective dose of anticancer drug used in the form of said conjugate is lower than effective dose of said anticancer drug in unconiugated form.
In particular embodiments of the invention, coniugate according to invention may be used in the treatment of cancer, wherein a greater inhibition of tumour growth is attained than when the same amount of said anticancer drug is used in unconiugated form.
For the purposes of the present invention, the effective dose should be understood as a dose of the anticancer drug which in a given patient can elicit the desirable effect, in particular the retardation or inhibition of tumour cell growth.
It is expected that in the case of the present invention, the effective conjugate dose will be at least twofold smaller than the dose of the unconjugated drug.
This effect may be observed as a retardation of tumour growth, which may be determined by observing changes in its volume and/or mass.
Subject invention discloses application of starch derivatives as effective, high molecular carriers for anticancer, therapeutic substances (e.g. antifoliates).
Nanoparticles composed of a starch derivative with a covalently bound anticancer drug (ester bond) were obtained. As a result of such a procedure, nanoparticles were obtained of physicochemical properties differing with respect to the carrier. These nanoparticles offered valuable therapeutic features and were composed of two medical substances, which are allowed and currently widely used in medicine. Physicochemical parameters characterizing the changes with respect to the initial polymer were analyzed, as were the biological implications. Biological examinations were performed in vitro with the application of mouse and human cancer cell lines. The anticancer effect in vivo was assessed by testing the activity on animals of lowered immunity enabling examinations with the application of human cancer line cells. The applied solution caused a considerable potentiation (a 10-fold increase in effectiveness when mean volumes of tumors were compared) of the anticancer activity of the conjugates after one intravenous administration of a preparation during a period of advanced cancer progression. The present invention opens the way for the application of modified starch as a drug carrier, especially for anticancer treatments.
LIST OF FIGURES
Figure 1 . Visual scheme of the HES-MTX conjugate.
Figure 2. Scheme of HES modification using MTX. R1 = H, -(CH2)nOH, drug or - (CH2)nO-MTX; R2 = H, -(CH2)nOH, drug, -(CH2)nO-drug or glucose unit; m=60 - 60000; n=1 - 20
Figure 3. LC-ESI-MS analysis of: A - total ion chromatogram of HES-MTX 130/0.4/48, B - total ion chromatogram of MTX pharmaceutical standard, C - ESI-MS spectrum of main peak from A.
Figure 4. Characterization of HES 130/0.4 and HES-MTX 130/0.4/59 conjugates by dynamic light scattering technique. Size distributions are shown according to intensity. Conjugates concentration - 5.5mM, solution - phosphate buffer 10mM pH=7.20.
Figure 5. Debye plot of HES 130/0.4 and HES-MTX conjugates.
Figure 6. Antiproliferative activity in vitro of HES-MTX conjugates in comparison with free MTX. All concentrations were based on the total contents of the MTX.
Figure 7. Antiproliferative activity in vitro of HES-MTX conjugates in comparison with free MTX. All concentrations were based on the total contents of the MTX.
Figure 8. Tumor growth kinetics of MV-4-1 1 bearing NOD/CDID mice. The mice were treated either with HES-MTX 130/0.4/59 conjugate (red line) or MTX (green line) alone. The control group received saline (black). Data are presented as mean tumor volume[nnnn3] ± standard deviation. The HES-MTX 130/0.4/59 conjugate diminishes the MV-4-1 1 tumor growth significantly when compared to control or MTX treated group.
Figure 9. Tumor growth kinetics of MV-4-1 1 bearing NOD/CDID mice, treated either with HES-MTX 130/0.4/59 (red), HES-MTX 130/0.4/25 (gray-red) or HES-MTX 130/0.4/1 1 (blue line) conjugate . Data are presented as mean tumor volume [mm3] ± standard deviation.
Figure 10. Tumor growth kinetics of MV-4-1 1 bearing NOD/CDID mice. The mice were treated either with HES-MTX 130/0.4/59 (red line), HES-MTX 130/0.4/25 (gray- red line) or HES-MTX 130/0.4/1 1 (blue line) conjugate, or MTX (green line) alone. The control group received saline (black). Data are presented as mean tumor volume[mm3] ± standard deviation. The HES-MTX 130/0.4/59 conjugate diminishes the MV-4-1 1 tumor growth significantly when compared to control or MTX treated group.
Figure 1 1 . The influence of HES-MTX 130/0.4/52 conjugate on the tumor growth kinetics of human leukemia MV-4-1 1 bearing mice. The tumor volumes are presented as % of control.
Figure 12. The influence of HES-MTX conjugates on the tumor growth kinetics of human leukemia MV-4-1 1 bearing mice. The tumor volumes are presented as % of control.
Figure 13. Body weight changes of MV-4-1 1 bearing NOD/CDID mice. The mice were treated either with HES-MTX 130/0.4/59, HES-MTX 130/0.4/48, HES-MTX 200/0.5/23, HES-MTX 130/0.4/25 or HES-MTX 130/0.4/1 1 conjugates, or MTX and 3% HES alone. The control group received saline (blue line). Data are presented as mean body weight [g] ± standard deviation. No body weight loss was observed during the experiment.
MATERIALS AND METHODS
Compounds
Methotrexate (EBEWE Pharma, Austria) and HES (Fresenius Kabi, Germany) were certified synthetic materials. DCC and DMF (Sigma-Aldrich) was used without further purification. Samples of the compounds were stored according to the manufacturer indications. In biological experiments, the compounds (HES, MTX and HES-MTX conjugates), prior to usage, were diluted in 0.9% saline solution to reach the required concentrations and administered to mice in a volume 0,01 ml/1 g of body weight.
Conjugates preparation (HES-MTX)
HES in aqueous solutions (6% w/w) was cooled to 4°C in ice bath and pH was adjusted to 10.5 using 1 M solution of Na2CO3. Afterwards cold solution was reacted with MTX anhydride according to the method previously described [1 1]. During conjugation reaction pH was still maintained to 10.5. After adding the total amount of MTX anhydride the pH was immediately adjusted to 7.0 (HCI 1 M) and obtained conjugate were dialyzed against ultrapure water to remove free MTX (dialysis membrane Visking 12kDA (Serva GmbH) for 96 hours or Pellicon® XL (Millipore Corp.) for 3 hours with flow rate 15ml/min). Conjugates with different levels of substitution MTX were synthesized by the same procedure using only a different (HES:MTX anhydride) stoichiometric ratio.
The stability of the prepared conjugates was assessed under different conditions and the compounds were stored at -20°C.
The conjugates were coded as:
HES-MTX 130/0.4/52
HES-MTX → conjugate of hydroxyethyl starch with covalently bound methotrexate
130 → Mean Molecular Weight of HES polymer [kDa]
0.4 → MS - molar substitution by hydroxyethyl groups on glucose units
52 → SL - molar substitution level by MTX on glucose units of the polymer x10"3
MS and SL is expressed as number of moles of MTX/hydroxyethyl groups per mole of the glucose unit.
Analytical procedures
The analysis of total MTX in preparations was based on absorption spectrophotometry in 100 mM sodium bicarbonate at 372 nm by absorption coefficient 8571 M"1cm"1.
Determination of unbound MTX was based on size exclusion chromatography with UV-VIS detection at a wavelength of 302 nm. A Superdex® Peptide column (150 x 4.6 mm) and mobile phase 100 mM sodium bicarbonate with a flow rate of 400μΙ/ηηίη was applied [75]. The total glucose contents were determined by the phenol-sulphuric acid method[76] with slight modifications.
Conjugates stability in aqueous media
Conjugates were dissolved in a series of various inorganic buffers at pH from 4 to 10 and ionic strength from 10 to 600 mM to a final concentration of 0.8mM (based on the contents of the MTX in the conjugate). The buffer solutions were incubated at 4°C, 20°C and 37°C. At selected time intervals each reaction solution was diluted with NaHCO3 0.1 M to final concentration 0.2mM (total MTX) and analyzed of unbound MTX[75].
Conjugates stability in human plasma
Conjugates were dissolved in human plasma to a final concentration of 0.8mM (based on the contents of the MTX in the conjugate). The solutions were incubated at 37°C. At selected time intervals each reaction solution was diluted with NaHCO3 0.1 M to final concentration 0.2mM (total MTX) and analyzed of unbound MTX[75]. DLS and SLS measurements
HES and HES-MTX conjugates were characterized by dynamic light scattering (DLS) and static light scattering (SLS) to obtain hydrodynamic parameters, polydispersity information and ζ potential distribution. The sample solution was illuminated by a 633 nm laser, and the intensity of light scattered at an angle of 173° was measured. The experimental result consists of the overall average size, size distributions by intensity, overall polydispersity index - PDI, ζ potential distribution and Debye plot. All samples were measured using a Zetasizer Nano ZS (Malvern Instruments) in a low volume quartz cuvette (12 μί). All solutions were clear without evidence of insoluble material. The following parameters were used: protein refractive index (1 .450), temperature (25°C), HES and HES-MTX conjugates (5.5mM) were based on the contents of the glucose.
Cell lines
The human biphenotypic B myelomonocytic leukemia MV-4-1 1 cells, human chronic myelogenous leukemia K562 cells, human erythroleukemia HEL92 and Human T lymphoblast CCRF-CEM were obtained from American Type Culture Collection (ATCC, Rockville, MD, USA). The cells were maintained in RPMI-1460 GlutaMAX adjusted to contain 2 mM glutamine and 1 .0 mM sodium pyruvate with fetal bovine serum (10%). Murine leukemia P388 were obtained from the American Type Culture Collection (Rockville, MD, USA) and were maintained in culture or were frozen in the Cell Culture Collection of the Institute of Immunology and Experimental Therapy, Wroclaw, Poland. Twenty-four hours before addition of the tested preparations, the cells were plated in 96-well plates (Sarstedt, Germany) at a density of 0.5x104 cells per well and were cultured in a mixture of RPMI 1640 and Opti-MEM (1 :1 ) medium, supplemented with 2 mM glutamine (Sigma-Aldrich Chemie GmbH, Germany), 100 mg/ml streptomycin (Polfa, Tarchomin, Poland) and 100 U/ml penicillin (Polfa), 5% fetal bovine serum (Sigma-Aldrich Chemie GmbH, Germany). The cells were cultured at 37°C in a humid atmosphere saturated with 5% CO2. Passages of P388 leukemia cells in BDF mice were carried out according to the NIH/NCI standard screening protocols for in vivo assessment [77, 78]
An anti-proliferative assay in vitro.
The in vitro cytotoxic effect was studied after 72-h exposure of the cultured cells to different concentrations of the test preparations (based on the total contents of the MTX). The cells were placed in 96-well flat bottom plates (Sarstedt, Inc., Newton, NC, USA) at a density of 1 x104 cells per well, 24 hours before addition of the tested compounds. The cells were exposed for 72 hours to various concentrations (0.1 ; 1 , 10 and 100 g/ml of MTX and HES-MTX conjugates. The MTT assay for evaluation of the cytostatic effect was performed. The results were calculated as a percent of proliferation inhibition by the tested compounds. The average values were counted using the data from 3-7 repetitions.
Experimental animals:
Male B6D2F1 mice, aged 16-20 weeks were used. The mice were supplied from the Animal Breeding Centre of the Medical Academy, Wroclaw, Poland, and were maintained in standard laboratory conditions. Experiments were performed according to the Guide for the Care and Use of Laboratory Animals (National Academy of Science, National Academy Press, Washington, D.C.) and were approved by the First Local Ethical Committee for the use of Laboratory Animals, Wroclaw, Poland. Male NOD/SCID mice, aged 8- 12 weeks old were used, weighing 20-25 g. The mice were supplied from the JU Children's University Hospital Krakow, Poland, were maintained in standard laboratory conditions. All experiments were performed according to Interdisciplinary Principles and Guidelines for the Use of Animals in Research, (National Academy of Science, National Academy Press, Washington, D.C.). Ad Hoc Committee on Animal Research and were approved by the 1 st Local Committee for Experiments with the Use of Laboratory Animals, Wroclaw, Poland. Design of the in vivo experiments.
Experiment 1 (P388)
Mice were injected with 106 leukemia (P388) cells i.p. (day 0), and 24 h later (day 1 ) each mouse was injected once i.p. with the appropriate agent. All doses (4(Vmol/kg) were based on the contents of the MTX in the conjugate. Body weight and survival data were collected on a daily basis throughout the duration of the experiment.
Experiment 2 and 3
In two subsequent experiments, NOD/SCID male mice were subcutaneously inoculated in the right flank of the abdomen with 7.5-8x106 viable MV-4-1 1 tumor cells per mouse in 0.2 ml Hanks solution (Sigma) and when the tumors become measurable (60-100mm3), mice were randomly divided into three (2nd experiment) or eight (3rd experiment) groups receiving different HES-MTX conjugates (HES-MTX 130/0.4/52 in 2nd experiment and all other conjugates in 3rd experiment) or HES, MTX or 0.9% saline solution alone. All doses (40μηηοΙ^) were based on the contents of the MTX in the conjugate. Body weight data were collected on a daily basis throughout the duration of the experiment. At the end of the experiment blood was harvested before subsequent sacrifice of the mice and tumors were collected during autopsy for further analysis.
Data handling
The in vitro results were presented in terms of IC50 values. The IC50 is the concentration of a tested agent which inhibits the proliferation of 50% of the cancer cell population. Average IC50 values for each preparation were calculated using data from at least three independent experiments.
Experiment 1 : The antileukemic effect in vivo was evaluated as the increase in lifespan (ILS) of treated mice over the control, calculated from the following formula: (MSTT/MSTC) x 100-100, where MSTT is the median survival time of treated animals and MSTC is the median survival time of untreated control mice.
Experiment 2 and 3: The tumor volume was calculated using the formula (a2 x b)/2, where a = shorter tumor diameter in mm and b=longer tumor diameter in mm. The inhibition of tumor growth was calculated from the following formula: TGI [%] = (WT/WC) x 100-100%, where WT was the median tumor volume of treated mice and WC that of the untreated control animals. Statistical evaluation:
Experiment 1 : Survival data in experimental in vivo groups were compared using Cox's F test with the Bonferroni correction for multiple comparisons (p adjusted = p counted χ N, where N = number of pairwise comparisons). P values less than 0.05 were considered significant.
Statistical evaluation.
Experiment 2 and 3: Statistical analysis was performed using STATISTICA version 7.1/10 (StatSoft Inc., USA). The data were analyzed by Kruskal-Wallis ANOVA. Multiple Comparisons p values 2-tailed) and Mann-Whitney test were used in analysis. P values below 0.05 were considered as significant.
RESULTS
Synthesis of conjugates
Synthesis of the HES-MTX conjugate runs according to the SN2 nucleophilic substitution mechanism. Therefore, the highest probability of modification concerns the hydroxyl group by an atom of C2 carbon. However, taking into account the fact that the polymer is primarily modified by hydroxyethyl groups (which may react with MTX as well), it may be assumed that there is a statistical possibility of a reaction with all the hydroxyl groups of the polymer. A visual scheme of the conjugate is presented in Fig. 1 , while the scheme of HES modification using MTX is shown in Fig. 2. Conjugates, according the protocol described in materials and method section, were obtained in two independent syntheses. Conjugates were purified from an excess of free drug and classified in terms of the substitution level of MTX (SL) and the kind of HES polymer applied (Table 1)
Table 1. Physicochemical properties of HES-MTX conjugates. Cg [M] - glucose unit content, C^9 TX - MTX total content in preparations, C¾- - MTX covalently bound to HES, SL - molar substitution level by MTX on glucose units, MTX-(COOH)2 - unbound (free) MTX [%]
Figure imgf000012_0001
Analysis of MTX released from conjugates (Fig. 3)
LC-MS analysis of MTX released from HES-MTX 130/0.4/48 conjugates was performed. The results were compared with an analysis of MTX standard performed in identical conditions. The identity of both samples was demonstrated by comparing the courses of total ion chromatograms and ESI-MS spectra of the main chromatographic peak.
Hydrodynamic characteristics of conjugates.
It may be concluded from the data presented that both the hydrodynamic diameter of conjugates and their polydispersity differ between particular degrees of MTX polymer substitution. All HES-MTX conjugates were characterized by a slightly higher hydrodynamic diameter towards an initial polymer. The polydispersity of conjugates is also characterized by higher values when compared to initial (non-modified) polymers. The increase in hydrodynamic diameter is higher, when the degree of substitution of a given MTX conjugate is higher. This is accompanied by an increase in conjugates polydispersity (Table 2, Fig. 4).
Table 2. Hydrodynamic parameters of HES-MTX conjugates. d(H) - hydrodynamic diameter, Pd - polydispersity. Conjugates concentration - 5.5mM, solution - phosphate buffer 10mM pH=7.20.
Figure imgf000013_0001
The zeta potential of all examined HES-MTX conjugates in water solution receives negative values. The zeta potential value decreases with an increase in the degree of SL substitution, and its distribution is subject to narrowing. The higher the degree of substitution of a given MTX conjugate, the more negative is the zeta potential of conjugates (Table 3). Changes in electric properties of these molecules may be of key significance with respect to their biological properties. Table 3. Hydrodynamic parameters of HES-MTX conjugates. ZP - Zeta potential, Conjugates concentration - 5.5mM, solution - phosphate buffer 10mM pH=7.20.
Figure imgf000014_0001
Measurements of static light dispersion demonstrate drastic changes in conjugate properties in solutions when compared to non-modified polymers. The differences are mainly concerned with the interaction of conjugate molecules with their surroundings (ie solvents). The change in character of the second viral coefficient points to the different character of interactions of free HES and HES-MTX conjugates with the environment, and that the differences are enhanced by the degree of MTX polymer substitution (Table 4, Fig. 5).
Table 4. Hydrodynamic parameters of HES-MTX conjugates. A2 - second virial coefficient, MW - estimated molecular weight, R2 - correlation coefficient of Debye plot. Conjugates concentration - 0.55 - 5.5mM, solution - NaCI 154mM.
Figure imgf000014_0002
Analysis of in vitro stability.
Analyses of stability were performed for HES-MTX 130/0.4/59 conjugate. Table 5 contains the data concerning the stability of the conjugate depending on solution pH, ionic strength and temperature. The cited data demonstrate the properties of bonds between HES and MTX in a manner typical for esters. Hydrolysis of these bonds is catalyzed by bases (fast decomposition in alkaline pH) and strongly depends on temperature.
The stability of the conjugates (half life time) in serum reaches 65 hours (Table 6) and is almost twice as short as the half life time in phosphate buffer of a similar pH. This fact points to the catalytic influence of serum components on the decomposition of HES-MTX conjugate.
Table 5. Conjugate (HES-MTX 130/0.4/59) stability in aqueous media. I - ionic strength, to.5 - half-life after which the release will be half of the total content of MTX in the conjugate.
Figure imgf000015_0001
Table 6. Conjugate (HES-MTX 130/0.4/59) stability in human plasma. I - ionic strength, to.5 - half-life after which the release will be half of the total content of MTX in the conjugate. pH T [°C] to.5 [h]
Human
7.24±0.05 37 65.4714.962
plasma
Phosphate
7.20±0.05 37 121.515.198
buffer 50mM BIOLOGICAL ACTIVITY ANALYSIS
In vitro examinations.
In vitro examinations of the antiproliferative activity of HES-MTX conjugates were performed on the cells of human leukemia lines: MV4-11 , K562, HEL92, CCRF-CEM. IC50 (inhibitory concentration 50%) values were determined in comparison with the reference compound - MTX - which was a positive control. The experimental data are presented in Table 7. Graphic presentation of an activity as mean values of IC50 for the MV4-11 line in Fig. 6 and Fig. 7.
IC50 values for the examined conjugates were about 100 nM (10 nM for free MTX) in the case of MV4-11 line, and within the range of 65-124 nM (7.7 nM for free MTX) in the case of the K562 line. All HES-MTX preparations demonstrated lower in vitro activity when compared to free MTX.
Table 7. Antiproliferative activity in vitro of HES-MTX conjugates in comparison with free MTX. All concentrations were based on the total contents of the MTX.
Figure imgf000016_0001
In vivo examinations.
Model of mouse leukemia P388 (experiment 1).
The experimental data are presented in Table 8. HES-MTX 130/0.4/52 preparation elongated the life time of females by 66% when compared to the life time of control mice, while the life time of males was elongated by 45.2% when compared to males in the control group. The life time length of mice in groups treated with the HES-MTX 130/0.4/52 preparation exceeded the life time of mice treated with MTX. Table 8. Survival data of leukemia-bearing mice (P388) in control group (untreated, n = 8), and mice treated with either free MTX (n = 7) or HES-MTX 130/0.4/52 conjugate (n = 8). All doses were based on the total contents of the MTX.
Figure imgf000017_0001
Model of human leukemia MV-4-11 (experiment 2).
HES-MTX 130/0.4/52 conjugate was characterized by a higher than free MTX effectiveness in the inhibition of the growth of experimental MV4- 1 tumors (
Table 9).
Table 9. The influence of HES-MTX 130/0.4/52 conjugate on the tumor growth kinetics of human leukemia MV-4-11 bearing mice. Mice were treated with HES-MTX conjugates, or MTX and 3% HEST alone. The control group received saline. The tumor volumes are presented as mean ± standard deviation, % tumor growth inhibition TGI, N- number of mice per group.
Figure imgf000017_0002
Model of human leukemia MV-4-11 (experiment 3).
The experiment aimed at:
• confirmation of previous results obtained for the HES-MTX 130/0.4/52 preparation, • examination of the relationship between the therapeutic activity of conjugate and the degree of MTX substitution (SL),
• determination of perspectives for the application of polymers differentiated in terms of MW and MS HES as MTX carriers.
All the examined HES-MTX conjugates were characterized by a higher than free MTX effectiveness in the inhibition of the growth of experimental MV4-11 tumors (Table 10).
Table 10. The influence of HES-MTX conjugates on the tumor growth kinetics of human leukemia MV-4-11 bearing mice. Mice were treated with HES-MTX conjugates, or MTX and 3% HEST alone. The control group received saline. The tumor volumes are presented as mean ± standard deviation, % tumor growth inhibition TGI, N- number of mice per group.
Figure imgf000018_0001
HES-MTX 130/0.4/59 and HES-MTX 130/0.4/48 conjugates demonstrated the highest activity among all the examined conjugates (Fig. 8). Both conjugates reduce the volume of MV4-11 tumors to a considerable degree (p<0.05) when compared to the control starting from day 13 up to day 29. Comparing the size of tumors in the group which was administered the HES-MTX 130/0.4/59 preparation in one dose, to the size of tumors in groups receiving MTX, significant differences in the size of tumors in days 15 to 29 were noted, while in the HES-MTX 130/0.4/48 group on days 23 to 29 (p<0.05) when compared to MTX.
Preparations HES-MTX 200/0.5/23, 130/0.4/25 and 130/0.4/11 demonstrated a poorer effect in terms of the inhibition of tumor growth when compared to HES-MTX 130/0.4/59 and 130/0.4/48 conjugates (Fig. 9, Fig. 10); however, their activity was higher than those of free MTX (Fig. 1 1 , Fig. 12).
Analyzing the TGI parameter (tumor growth inhibition) on the 29th day of the experiment, the highest inhibition in tumor growth was 83.3% in the HES-MTX 130/0.4/59 group, then TGI values were 74.8% for HES-MTX 130/0.4/48, 61.6% for HES-MTX 200/0.5/23, 45.8% and 41.8% for HES-MTX 130/0.4/11 and HES-MTX 130/0.4/25, respectively. For a comparison, the TGI value for free MTX on day 29 was 28.97%.
Based on the analysis of changes in the body weight of mice, no toxicity was observed in the accepted scheme of preparations administration. The average body weight of mice during preparation administration was not lower than on the first day of treatment in any of the groups observed (Fig. 13).
DISCUSSION AND CONCLUSIONS
Physicochemical properties of HES-MTX conjugates and their biological activity. The obtained HES-MTX conjugates contained hydroxyethyl starch as a carrier substance (HES 130/0.4 and HES 200/0.5). Modifications to HES using MTX were conducted using the anhydride method. The linker between the drug and the polymer was thus glutamic acid (an integral part of the MTX particle) and the connecting bond was an ester bond. Consequently, the following facts should be taken into consideration:
• HES-MTX conjugates are composed of two medical substances, certified and currently widely used in medicine,
• a linker in a conjugate is an integral part of the drug particle - there is a lack of additional linking substances or groups,
• esterification is an equilibrium reaction, and a by- product of this reaction is water - during drug release from a conjugate (hydrolysis of esters) we deal with the reconstruction of the substrates of the reaction, i.e. we obtain free MTX and non-modified HES,
• identity of the MTX released from the conjugate with the pharmaceutical MTX standard was demonstrated,
• the kinetics of MTX release from conjugates strongly depends on environmental pH and temperature. It should thus be expected that drug release would occur with a differentiated rate depending on the pH observed in a given tissue (blood, cancer tissue, endosome). There is a possibility that the described feature may lead to conjugate deposition in sites of lowered pH value.
An additional mechanism of MTX release from a conjugate is HES-MTX hydrolysis by a-amylases. Conjugate subjected to partial enzymatic degradation may exhibit profitable properties. Moreover, digestion of the conjugate may limit MTX release in sites of lowered pH.
Enzymatic HES hydrolysis may also be caused by the fact that no toxicity of HES- MTX preparations was observed in in vivo examinations. They are subject, as in the case of HES application in volume therapy, to gradual degradation and removal from an organism.
The examinations of hydrodynamic parameters revealed the following facts:
• HES-MTX conjugates are characterized by insignificantly higher hydrodynamic diameters when compared to initial HES,
• HES-MTX conjugates are characterized by insignificantly higher polydispersity when compared to initial HES,
• when compared to initial HES, conjugates are characterized by negative zeta potential of a value and distribution dependent on the degree of MTX polymer substitution,
• HES-MTX conjugates are characterized by a quite different Debye curve course in comparison to initial HES. This may prove the fact that intermolecular conjugate-conjugate and conjugate-environment interactions are of a different character from interactions between non-modified HES, which may influence the adhesion and distribution of conjugates in an organism.
In summary, hybrid HES-MTX molecules demonstrate differences in hydrodynamic and electric parameters when compared to non-modified HES polymers. The differences centre on hydrodynamic diameter, polydispersity, charge of molecules and interaction with other molecules of the environment.
An antiproliferation study in vitro of HES-MTX conjugates performed on the control (MTX) revealed an expected effect - antiproliferation activity was an order lower than that for the free drug. A lack of the presence of the enzymatic apparatus of an organism in this type of experiment means that mainly MTX released as a result of chemical hydrolysis is responsible for an antiproliferative effect.
Biological study in vivo provided the following information:
• anticancer activity in in vivo experiments was the most effective when cancer developed in the form of a tumor,
• all synthesized conjugates profitably influenced the kinetics of the growth of experimental cancer tumors in mice, and all were more effective than free MTX (in an accepted administration scheme),
the best effects were obtained for conjugates of the highest degree of MTX substitution, obtaining over 90% reduction in tumor mass, and in individual cases their temporary atrophy was noted,
• conjugates of the lowest degree of substitution - HES-MTX 130/0,4/11 - demonstrated the lowest activity among the examined conjugates, but this activity was higher than that for free MTX.
Comparison of the effectiveness of two HES types as carriers did not reveal any considerable differences - it only gave a suggestion that HES with higher values of MW, MS and ratio of C2/C6 (when compared to HES 130/0,4) may be more effective carriers of therapeutic substances. There is a probability that 1st and 2nd generation HES used years ago in medicine will return to use drug carriers, since their negative features in volume therapy, such as increased accumulation in some tissues, may appear positive in the case of oncological drug administration.
Perspectives for HES application as high molecular carriers of anticancer drugs.
The term "polymer therapeutics" includes natural and synthetic macromolecules used in interdisciplinary studies combining polymer chemistry, biology, pharmacy and medicine for the design of biologically active polymer-drug, polymer-protein conjugates or polymeric micelles [79]. Many polymers with attractive physicochemical features have no possibility to become effective drug carriers due to their biological properties, such as toxicity or immunogenicity [80]. Therefore, selection or design of carrier polymers is a key issue, limiting the process of conjugate formation. Similarly, a significant element is the suitable selection of the linker connecting a polymer with a drug. If it is too stable or not subject to a biodegradation bond connecting the drug with a carrier, it would result in limited drug release and its insufficient concentration in a target site. In turn, quick degradation of a connecting bond may lead to excessive removal of a dissociated drug by the kidneys [13] Attention should be paid to the fact that bio-distribution of compounds in an organism never occurs entirely on one selected path. In fact, we observe an effect as being a result of a range of physiological, physicochemical factors and overlapping interactions. Thus, when designing drug-carrier systems, attention should be paid to all factors which may affect the realization of the assumed effect.
The application of natural polymers as drug carriers seems to the safest; however, modifications of these polymers enabling therapeutic settlement are not biological property neutral. [81-83].
Ringsdorfs idealistic concept [84], and elements connected to the influence of endocytosis on conjugate behavior in an organism [85] may be useful in the design of polymeric drug-carrier conjugates.
Combining low molecular drugs with high molecular carriers can inter alia:
• regulate drug transport in an organism and cellular transport determining preferential transport to target sites, limiting concurrently passive transport to healthy tissues [86],
• cause accumulation of conjugates in serum or/and tissues making use of the EPR effect [18]
• give the possibility of the application of targeted therapy, for example by modification of polymers with antibodies [87],
Desirable properties of polymers used for the design of therapeutic conjugates, according to the current opinions, are as follows [13, 79, 88]:
• polymers should be non-toxic and non-immunogenic,
• mean molar mass of polymers should be large enough to ensure a suitably long period of conjugate circulation in an organism; however, for polymers not subject to biodegradation they must be optimized in such a manner as to ensure gradual elimination from an organism,
• polymers should ensure the possibility of the attachment of a suitable amount of the drug,
• linkers connecting drugs with polymers should be stable in conditions of conjugate transport and ensure effectiveness in a target site,
The application of HES polymers as anticancer drug carriers creates large possibilities. Depending on its origin, an initial polymer (natural starch) is characterized by different properties (mean molecular weight, number of a-1 ,6- glucoside bonds). These polymers may be designed according to established parameters critical for biological properties, such as MW, MS and ratio of C2/C6, with respect to a specified therapeutic substance. The degree of substitution (MS) and its localization may be regulated precisely in the process of modification with hydroxyethyl groups. The whole process may be led in a way enabling the acquisition of a polymer of designed properties adequate to the specified biological targets. These facts mean that HES is a very interesting polymer with medical applications, and currently its utilization in volume therapy may be considered as only the beginning of its "medical career".
HES demonstrates practically all the above-presented desirable properties of polymers used for the design of therapeutic conjugates, and additionally:
• drugs subjected to conjugation may be any biologically active particle possessing a free carboxyl group (in particular MTX), even if this group is essential for the biological activity of this particle,
• conjugates may be composed of two or more biologically active substances (including diagnostic substances) revealing new physicochemical and biological properties after conjugation.
Finally, examination and then control of structural and functional differences of high molecular polymers in connection with continuous examinations of molecular mechanisms of various complex diseases, including cancers, has the chance to lead to a new generation of polymer drugs based on HES.
REFERENCES
[1] R. Haag, F. Kratz, Polymer therapeutics: concepts and applications, Angew Chem Int Ed Engl, 45 (2006) 1198-1215.
[2] T.M. Allen, P.R. Cullis, Drug delivery systems: entering the mainstream, Science, 303 (2004) 1818-1822.
[3] R. Duncan, M.J. Vicent, F. Greco, R.I. Nicholson, Polymer-drug conjugates: towards a novel approach for the treatment of endrocine-related cancer, Endocrine-related cancer, 12 Suppl 1 (2005) S189-199.
[4] H. Kamada, Y. Tsutsumi, Y. Yamamoto, T. Kihira, Y. Kaneda, Y. Mu, H. Kodaira, S.I. Tsunoda, S. Nakagawa, T. Mayumi, Antitumor activity of tumor necrosis factor-alpha conjugated with polyvinylpyrrolidone on solid tumors in mice, Cancer research, 60 (2000) 6416-6420.
[5] S.D. Chipman, F.B. Oldham, G. Pezzoni, J.W. Singer, Biological and clinical characterization of paclitaxel poliglumex (PPX, CT-2103), a macromolecular polymer-drug conjugate, International journal of nanomedicine, 1 (2006) 375-383. [6] J.Y. Ljubimova, M. Fujita, A.V. Ljubimov, V.P. Torchilin, K.L. Black, E. Holler, Poly(malic acid) nanoconjugates containing various antibodies and oligonucleotides for multitargeting drug delivery, Nanomedicine (Lond), 3 (2008) 247-265.
[7] J.Y. Ljubimova, M. Fujita, N.M. Khazenzon, B.S. Lee, S. Wachsmann-Hogiu, D.L. Farkas, K.L. Black, E. Holler, Nanoconjugate based on polymalic acid for tumor targeting, Chemico-biological interactions, 171 (2008) 195-203.
[8] G. Pasut, F.M. Veronese, PEGylation for improving the effectiveness of therapeutic biomolecules, Drugs Today (Bare), 45 (2009) 687-695.
[9] J. Kopecek, Biomaterials and drug delivery: past, present, and future, Molecular pharmaceutics, 7 (2010) 922- 925.
[10] U. Kanska, M.S. Omar, R. Budzynska, D. Nevozhay, M. Jagiello, A. Opolski, J. Boratynski, Antileukemic activity of glycated fibrinogen-methotrexate conjugates, Anticancer Res, 25 (2005) 2229-2234.
[11] D. Nevozhay, R. Budzynska, U. Kanska, M. Jagiello, M.S. Omar, J. Boratynski, A. Opolski, Antitumor properties and toxicity of dextran-methotrexate conjugates are dependent on the molecular weight of the carrier, Anticancer Res, 26 (2006) 1135-1143.
[12] M.J. Vicent, R. Duncan, Polymer conjugates: nanosized medicines for treating cancer, Trends Biotechnol, 24 (2006) 39-47.
[13] R. Duncan, Polymer conjugates as anticancer nanomedicines, Nat Rev Cancer, 6 (2006) 688-701.
[14] F. Canal, J. Sanchis, M.J. Vicent, Polymer-drug conjugates as nano-sized medicines, Curr Opin Biotech, 22
(2011) 894-900.
[15] H. Maeda, Tumor-selective delivery of macromolecular drugs via the EPR effect: background and future prospects, Bioconjugate chemistry, 21 (2010) 797-802.
[16] H. Maeda, G.Y. Bharate, J. Daruwalla, Polymeric drugs for efficient tumor-targeted drug delivery based on EPR-effect, Eur J Pharm Biopharm, 71 (2009) 409-419.
[17] A.K. Iyer, G. Khaled, J. Fang, H. Maeda, Exploiting the enhanced permeability and retention effect for tumor targeting, Drug discovery today, 11 (2006) 812-818.
[18] Y. Matsumura, H. Maeda, A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs, Cancer research, 46 (1986) 6387-6392.
[19] J.A. Nagy, L. Benjamin, H. Zeng, A.M. Dvorak, H.F. Dvorak, Vascular permeability, vascular hyperpermeability and angiogenesis, Angiogenesis, 11 (2008) 109-119.
[20] J. Fang, H. Nakamura, H. Maeda, The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect, Advanced drug delivery reviews, 63 (2011) 136- 151.
[21] M.R. Kano, Y. Bae, C. Iwata, Y. Morishita, M. Yashiro, M. Oka, T. Fujii, A. Komuro, K. Kiyono, M. Kaminishi, K. Hirakawa, Y. Ouchi, N. Nishiyama, K. Kataoka, K. Miyazono, Improvement of cancer-targeting therapy, using nanocaniers for intractable solid tumors by inhibition of TGF-β signaling, Proceedings of the National Academy of Sciences, 104 (2007) 3460-3465.
[22] Y. Takakura, M. Hashida, Macromolecular carrier systems for targeted drug delivery: pharmacokinetic considerations on biodistribution, Pharmaceutical research, 13 (1996) 820-831.
[23] N. Larson, H. Ghandehari, Polymeric conjugates for drug delivery, Chemistry of materials : a publication of the American Chemical Society, 24 (2012) 840-853.
[24] P. Vaupel, F. Kallinowski, P. Okunieff, Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review, Cancer research, 49 (1989) 6449-6465.
[25] J.L. Wike-Hooley, J. Haveman, H.S. Reinhold, The relevance of tumour pH to the treatment of malignant disease, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology, 2 (1984) 343-366.
[26] L.E. Gerweck, K. Seetharaman, Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer, Cancer research, 56 (1996) 1194-1198.
[27] K. Ulbrich, T. Etrych, P. Chytil, M. Jelinkova, B. Rihova, HPMA copolymers with pH-controlled release of doxorubicin: in vitro cytotoxicity and in vivo antitumor activity, Journal of controlled release : official journal of the Controlled Release Society, 87 (2003) 33-47.
[28] M. Westphal, M.F. James, S. Kozek-Langenecker, R. Stacker, B. Guidet, H. Van Aken, Hydroxyethyl starches: different products-different effects, Anesthesiology, 111 (2009) 187-202.
[29] J. Boldt, Modern rapidly degradable hydroxyethyl starches: current concepts, Anesthesia and analgesia, 108 (2009) 1574-1582.
[30] S.D. Gandhi, R.B. Weiskopf, C. Jungheinrich, R. Koom, D. Miller, R.E. Shangraw, D.S. Prough, D. Baus, F.
Bepperling, D.C. Waritier, Volume replacement therapy during major orthopedic surgery using Voluven
(hydroxyethyl starch 130/0.4) or hetastarch, Anesthesiology, 106 (2007) 1120-1127.
[31] B. Weidler, B. von Bormann, K. Sommermeyer, E. Lohmann, J. Peil, G. Hempelmann, [Pharmacokinetic parameters as criteria for clinical use of hydroxyethyl starch preparations], Arzneimittel-Forschung, 41 (1991)
494-498.
[32] A. Yacobi, R.G. Stall, C.Y. Sum, CM. Lai, S.D. Gupta, J.D. Hulse, Pharmacokinetics of hydroxyethyl starch in normal subjects, Journal of clinical pharmacology, 22 (1982) 206-212.
[33] F. Jesch, G. Hubner, V. Zumtobel, M. Zimmermann, K. Messmer, [Hydroxyethyl starch (HAS 450/0.7) in human plasma and liver. Course of concentration and histological changes], Infusionstherapie und klinische Ernahrung, 6 (1979) 112-117.
[34] C. Sirtl, H. Laubenthal, V. Zumtobel, D. Kraft, W. Jurecka, Tissue deposits of hydroxyethyl starch (HES): dose-dependent and time-related, British journal of anaesthesia, 82 (1999) 510-515.
[35] J.D. Hulse, A. Yacobi, Hetastarch: an overview of the colloid and its metabolism, Drug intelligence & clinical pharmacy, 17 (1983) 334-341. [36] S. Stander, Z. Szepfalusi, B. Bohle, H . Stander, D. Kraft, T.A. Luger, D. Metze, Differential storage of hydroxyethyl starch (HES) in the skin: an immunoelectron-microscopical long-term study, Cell and tissue research, 304 (2001 ) 261 -269.
[37] D. Metze, S. Reimann, Z. Szepfalusi, B. Bohle, D. Kraft, T.A. Luger, Persistent pruritus after hydroxyethyl starch infusion therapy: a result of long-term storage in cutaneous nerves, The British journal of dermatology, 136 (1997) 553-559.
[38] J. Leuschner, J. Opitz, A. Winkler, R. Scharpf, F. Bepperling, Tissue storage of 14C-labelled hydroxyethyl starch (HES) 130/0.4 and HES 200/0.5 after repeated intravenous administration to rats, Drugs in R&D, 4 (2003) 331 -338.
[39] Y. Claes, J. Van Hemelrijck, M. Van Gerven, J. Arnout, J. Vermylen, B. Weidler, H. Van Aken, Influence of hydroxyethyl starch on coagulation in patients during the perioperative period, Anesthesia and analgesia, 75 (1992) 24-30.
[40] M. Jamnicki, A. Zollinger, B. Seifert, D. Popovic, T. Pasch, D.R. Spahn, Compromised blood coagulation: an in vitro comparison of hydroxyethyl starch 130/0.4 and hydroxyethyl starch 200/0.5 using thrombelastography, Anesthesia and analgesia, 87 (1998) 989-993.
[41 ] S.A. Kozek-Langenecker, Effects of hydroxyethyl starch solutions on hemostasis, Anesthesiology, 103 (2005) 654-660.
[42] C. Madjdpour, N. Dettori, P. Frascarolo, M. Burki, M. Boll, A. Fisch, T. Bombeli, D.R. Spahn, Molecular weight of hydroxyethyl starch: is there an effect on blood coagulation and pharmacokinetics?, British journal of anaesthesia, 94 (2005) 569-576.
[43] C.A. Volta, V. Alvisi, M. Campi, E. Marangoni, R. Alvisi, M. Castellazzi, E. Fainardi, M.C. Manfrinato, F. Dallocchio, T. Bellini, Influence of different strategies of volume replacement on the activity of matrix metalloproteinases: an in vitro and in vivo study, Anesthesiology, 106 (2007) 85-91.
[44] K. Lang, S. Suttner, J. Boldt, B. Kumle, D. Nagel, Volume replacement with HES 130/0.4 may reduce the inflammatory response in patients undergoing major abdominal surgery, Canadian journal of anaesthesia = Journal canadien d'anesthesie, 50 (2003) 1009-1016.
[45] W.A.d.L. Eichner, 35510 Butzbach, DE), Lutterbeck, Katharina (Mainzer Tor Anlage 10, 61 169 Friedberg, DE), Zander, Norbert (Zellbergsheideweg 45, 38527 Meine, DE), Frank, Ronald (Heegblick 1 , 38527 Meine- Grassel, DE), Knoller, Helmut (Ritterstrasse 15, 61 169 Friedberg, DE), Conradt, Harald (Okerstrasse 1 1 , 38100 Braunschweig, DE), Conjugates Of Hydroxyethyl Starch And Erythropoietin., in, Fresenius Kabi Deutschland GmbH (Else-Kroner-Strasse 1 , 61352 Bad Homburg, DE)„ 2007.
[46] K. Sommermeyer, Process for the production of conjugates from polysaccharides and polynucleotides. , in, vol. WO 2005/074993 08 02, Supramol Parenteral Colloid GMBH NOXXON PHARMA AG 2009.
[47] K.R. Sommermeyer, DE), Eichner, Wolfram (Butzbach, DE), Haemoglobin-hydroxyethyl starch conjugates as oxygen carriers, in, Fresenius AG (Oberusel, DE), U nited States, 2000.
[48] N. Zander, R. Frank, Conjugates of Hydroxyalkyl Starch and a Protein., in, Fresenius Kabi DE GMBH, 2005. [49] P. Harmatz, R.W. Grady, P. Dragsten, E. Vichinsky, P. Giardina, J. Madden, M. Jeng, B. Miller, G. Hanson, B. Hedlund, Phase lb clinical trial of starch-conjugated deferoxamine (40SD02): a novel long-acting iron chelator, British journal of haematology, 138 (2007) 374-381.
[50] P.R. Dragsten, P.E. Hallaway, G.J. Hanson, A.E. Berger, B. Bernard, B.E. Hedlund, First human studies with a high-molecular-weight iron chelator, The Journal of laboratory and clinical medicine, 135 (2000) 57-65.
[51] A. Besheer, T.C. Hertel, J. Kressler, K. Mader, M. Pietzsch, Enzymatically catalyzed HES conjugation using microbial transglutaminase: Proof of feasibility, Journal of pharmaceutical sciences, 98 (2009) 4420-4428.
[52] M. Visentin, R.B. Zhao, I.D. Goldman, The Antifolates, Hematol Oncol Clin N, 26 (2012) 629-+.
[53] L. Genestier, R. Paillot, L. Quemeneur, K. Izeradjene, J. P. Revillard, Mechanisms of action of methotrexate,
Immunopharmacology, 47 (2000) 247-257.
[54] J.R. Bertino, Cancer research: from folate antagonism to molecular targets, Best practice & research. Clinical haematology, 22 (2009) 577-582.
[55] M.G. Fury, D.G. Pfister, Current recommendations for systemic therapy of recurrent and/or metastatic head and neck squamous cell cancer, Journal of the National Comprehensive Cancer Network : JNCCN, 9 (2011) 681 - 689.
[56] Y. Molin, J. Fayette, Current chemotherapies for recurrent/metastatic head and neck cancer, Anti-cancer drugs, 22 (2011) 621-625.
[57] N. Ismaili, M. Amzerin, A. Flechon, Chemotherapy in advanced bladder cancer: current status and future, Journal of hematology & oncology, 4 (2011) 35.
[58] E. Neumann, E. Frei, D. Funk, M.D. Becker, H.H. Schrenk, U. Muller-Ladner, C. Fiehn, Native albumin for targeted drug delivery, Expert opinion on drug delivery, 7 (2010) 915-925.
[59] A.N. Vis, A. van der Gaast, B.W. van Rhijn, T.K. Catsburg, C. Schmidt, G.H. Mickisch, A phase II trial of methotrexate-human serum albumin (MTX-HSA) in patients with metastatic renal cell carcinoma who progressed under immunotherapy, Cancer chemotherapy and pharmacology, 49 (2002) 342-345.
[60] A. Wunder, G. Stehle, H.H. Schrenk, G. Hartung, D.L. Heene, W. Maier-Borst, H. Sinn, Antitumor activity of methotrexate-albumin conjugates in rats bearing a Walker-256 carcinoma, International journal of cancer. Journal international du cancer, 76 (1998) 884-890.
[61] G. Stehle, H. Sinn, A. Wunder, H.H. Schrenk, S. Schutt, W. Maier-Borst, D.L. Heene, The loading rate determines tumor targeting properties of methotrexate-albumin conjugates in rats, Anti-cancer drugs, 8 (1997) 677-685.
[62] L. Bures, A. Lichy, J. Bostik, M. Spundova, The use of protein as a carrier of methotrexate for experimental cancer chemotherapy. V. Alternative method for preparation of serum albumin-methotrexate derivative, Neoplasma, 37 (1990) 225-231.
[63] G.W. Halbert, AT. Florence, J.F. Stuart, Characterization of in-vitro drug release and biological activity of methotrexate-bovine serum albumin conjugates, The Journal of pharmacy and pharmacology, 39 (1987) 871-876.
[64] J. Boratynski, A. Opolski, J. Wietrzyk, A. Gorski, C. Radzikowski, Cytotoxic and antitumor effect of fibrinogen- methotrexate conjugate, Cancer Lett, 148 (2000) 189-195. [65] R. Budzynska, D. Nevozhay, U. Kanska, M. Jagiello, A. Opolski, J. Wietrzyk, J. Boratynski, Antitumor activity of mannan-methotrexate conjugate in vitro and in vivo, Oncology research, 16 (2007) 415-421.
[66] D. Nevozhay, R. Budzynska, M. Jagiello, U. Kanska, M.S. Omar, A. Opolski, J. Wietrzyk, J. Boratynski, The effect of the substitution level of some dextran-methotrexate conjugates on their antitumor activity in experimental cancer models, Anticancer Res, 26 (2006) 2179-2186.
[67] Y. Chau, N.M. Dang, F.E. Tan, R. Langer, Investigation of targeting mechanism of new dextran-peptide- methotrexate conjugates using biodistribution study in matrix-metalloproteinase-overexpressing tumor xenograft model, Journal of pharmaceutical sciences, 95 (2006) 542-551.
[68] Y. Chau, R.F. Padera, N.M. Dang, R. Langer, Antitumor efficacy of a novel polymer-peptide-drug conjugate in human tumor xenograft models, International Journal of Cancer, 118 (2006) 1519-1526.
[69] Y. Chau, F.E. Tan, R. Langer, Synthesis and characterization of dextran-peptide-methotrexate conjugates for tumor targeting via mediation by matrix metalloproteinase II and matrix metalloproteinase IX, Bioconjugate chemistry, 15 (2004) 931-941.
[70] A. Homma, H. Sato, T. Tamura, A. Okamachi, T. Emura, T. Ishizawa, T. Kato, T. Matsuura, S. Sato, Y. Higuchi, T. Watanabe, H. Kitamura, K. Asanuma, T. Yamazaki, M. Ikemi, H. Kitagawa, T. Morikawa, H. Ikeya, K. Maeda, K. Takahashi, K. Nohmi, N. Izutani, M. Kanda, R. Suzuki, Synthesis and optimization of hyaluronic acid- methotrexate conjugates to maximize benefit in the treatment of osteoarthritis, Bioorganic & medicinal chemistry, 18 (2010) 1062-1075.
[71] A. Homma, H. Sato, A. Okamachi, T. Emura, T. Ishizawa, T. Kato, T. Matsuura, S. Sato, T. Tamura, Y. Higuchi, T. Watanabe, H. Kitamura, K. Asanuma, T. Yamazaki, M. Ikemi, H. Kitagawa, T. Morikawa, H. Ikeya, K. Maeda, K. Takahashi, K. Nohmi, N. Izutani, M. Kanda, R. Suzuki, Novel hyaluronic acid-methotrexate conjugates for osteoarthritis treatment, Bioorganic & medicinal chemistry, 17 (2009) 4647-4656.
[72] Y.H. Zhang, TP. Thomas, K.H. Lee, M.H. Li, H. Zong, A.M. Desai, A. Kotlyar, B.H. Huang, M.M.B. Holl, J.R. Baker, Polyvalent saccharide-functionalized generation 3 poly(amidoamine) dendrimer-methotrexate conjugate as a potential anticancer agent, Bioorganic & medicinal chemistry, 19 (2011) 2557-2564.
[73] A. Myc, J. Kukowska-Latallo, P. Cao, B. Swanson, J. Battista, T. Dunham, J.R. Baker, Jr., Targeting the efficacy of a dendrimer-based nanotherapeutic in heterogeneous xenograft tumors in vivo, Anti-cancer drugs, 21 (2010) 186-192.
[74] C. Samori, H. Ali-Boucetta, R. Sainz, C. Guo, F.M. Toma, C. Fabbro, T. da Ros, M. Prato, K. Kostarelos, A. Bianco, Enhanced anticancer activity of multi-walled carbon nanotube-methotrexate conjugates using cleavable linkers, Chemical Communications, 46 (2010) 1494-1496.
[75] J. Ciekot, T. Goszczynski, J. Boratynskit, Methods for methotrexate determination in macromolecular conjugates drug carrier, Acta poloniae pharmaceutica, 69 (2012) 1342-1346.
[76] T. Masuko, A. Minami, N. Iwasaki, T. Majima, S. Nishimura, Y.C. Lee, Carbohydrate analysis by a phenol- sulfuric acid method in microplate format, Analytical biochemistry, 339 (2005) 69-72.
[77] S.N. Khleif, G.A. Curt, Animal models in drug development., Lea & Febiger, London, 1993. [78] M. Dunitz, Experimental Therapeutics Program. In Vivo Cancer Models., in: M. Dunitz (Ed.) N IH Publication, vol. 84-2635, Bethesda, 2003, pp. 1976-1982.
[79] R. Duncan, Polymer therapeutics as nanomedicines: new perspectives, Curr Opin Biotech, 22 (201 1) 492- 501.
[80] B. Rihova, Biocompatibility of biomaterials: Hemocompatibility, immunocompatibility and biocompatibility of solid polymeric materials and soluble targetable polymeric carriers, Advanced drug delivery reviews, 21 (1996) 157-176.
[81 ] S. Danhauserriedl, E. Hausmann, H.D. Schick, R. Bender, H. Dietzfelbinger, J. Rastetter, A.R. Hanauske, Phase-I Clinical and Pharmacokinetic Trial of Dextran Conjugated Doxorubicin (Ad-70, Dox-Oxd), Invest New Drug, 1 1 (1993) 187-195.
[82] E. Kumazawa, Y. Ochi, DE-310, a novel macromolecular carrier system for the camptothecin analog DX- 8951f: Potent antitumor activities in various murine tumor models, Cancer Sci, 95 (2004) 168-175.
[83] C.H. Takimoto, P.M. Lorusso, L. Forero, G.H. Schwartz, A.W. Tolcher, A. Patnaik, L.A. Hammond, S. Syed, C. Simmons, M. Ducharme, R. De Jager, E.K. Rowinsky, A phase I and pharmacokinetic study of DE-310 administered as a 3 hour infusion every 4 weeks (wks) to patients (pts) with advanced solid tumors or lymphomas., Clinical Cancer Research, 9 (2003) 6105s-6105s.
[84] H. Ringsdorf, Structure and Properties of Pharmacologically Active Polymers, J Polym Sci Pol Sym, (1975) 135-153.
[85] C. Deduve, T. Debarsy, B. Poole, A. Trouet, P. Tulkens, F. Vanhoof, Lysosomotropic Agents, Biochemical pharmacology, 23 (1974) 2495-&.
[86] J. Kopecek, P. Kopeckova, T. Minko, Z. Lu, H PMA copolymer-anticancer drug conjugates: design, activity, and mechanism of action, Eur J Pharm Biopharm, 50 (2000) 61 -81.
[87] R. Duncan, The dawning era of polymer therapeutics, Nature reviews. Drug discovery, 2 (2003) 347-360.
[88] R. Duncan, M.J. Vicent, Polymer therapeutics-prospects for 21st century: The end of the beginning, Advanced drug delivery reviews, (2012).

Claims

Claims
1 . A conjugate composed of a carrier and a drug covalently bound thereto, defined by the general formula:
Figure imgf000030_0001
where:
m is an integer from 60 to 60000,
R1 denotes a substituent selected from a group encompassing: hydrogen, -(CH2)nOH, -Drug, -(CH2)nO-Drug,
R2 denotes a substituent selected from a group encompassing: hydrogen, -(CH2)nOH, -Drug, -(CH2)nO-Drug, glucose unit,
wherein as the carrier, it contains starch with a molecular mass from 10 to 1000 kDa and a degree of substitution with hydroxyalkyl, preferably hydroxyethyl, groups from 0.1 to 0.9 mol/glucose unit, and as the drug it contains a molecule of a known antiinflammatory or anticancer drug which possesses a free carboxyl group, particularly methotrexate,
for use in the treatment of cancer, wherein the effective dose of anticancer drug used in the form of said conjugate is lower than effective dose of said anticancer drug in unconjugated form.
2. A conjugate according to claim 1 for use in the treatment of cancer, wherein a greater inhibition of tumour growth is attained than when the same amount of said anticancer drug is used in unconjugated form.
PCT/EP2013/053974 2012-02-27 2013-02-27 A conjugate of methotrexate and hydroxyethyl starch for use in the treatment cancer WO2013127885A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PL398253A PL228735B1 (en) 2012-02-27 2012-02-27 A conjugate comprising a carrier and covalently tied with the drug and the use thereof
PLP.398253 2012-02-27

Publications (1)

Publication Number Publication Date
WO2013127885A1 true WO2013127885A1 (en) 2013-09-06

Family

ID=48193243

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/053974 WO2013127885A1 (en) 2012-02-27 2013-02-27 A conjugate of methotrexate and hydroxyethyl starch for use in the treatment cancer

Country Status (2)

Country Link
PL (1) PL228735B1 (en)
WO (1) WO2013127885A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112094320A (en) * 2019-06-18 2020-12-18 首都医科大学 His-Gly-Glu modified methotrexate, synthesis, antitumor activity and application thereof
US10874753B2 (en) 2014-09-26 2020-12-29 The South African Nuclear Energy Corporation Limited Radiopharmaceutical conjugate of a metabolite and an EPR agent, for targeting tumour cells
CN113041359A (en) * 2021-03-26 2021-06-29 湖南师范大学 Glutathione-responsive osteosarcoma-resistant prodrug nanoparticle and preparation method and application thereof
WO2022260545A1 (en) * 2021-06-11 2022-12-15 Uniwersytet Medyczny Im. Piastów Śląskich We Wrocławiu Synthesis of a novel glucose-conjugated methotrexate and its application for the treatment and prevention of neoplasms

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005000074A1 (en) 2003-06-09 2005-01-06 Aero International Products, Inc. Reversible inflation system
EP2070951A1 (en) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Method for producing a hydroxyalkyl starch derivatives with two linkers
EP2070950A1 (en) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Hydroxyalkyl starch derivatives and process for their preparation

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005000074A1 (en) 2003-06-09 2005-01-06 Aero International Products, Inc. Reversible inflation system
EP2070951A1 (en) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Method for producing a hydroxyalkyl starch derivatives with two linkers
EP2070950A1 (en) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Hydroxyalkyl starch derivatives and process for their preparation

Non-Patent Citations (89)

* Cited by examiner, † Cited by third party
Title
"National Academy of Science", NATIONAL ACADEMY PRESS
A. BESHEER; T.C. HERTEL; J. KRESSLER; K. MADER; M. PIETZSCH: "Enzymatically catalyzed HES conjugation using microbial transglutaminase: Proof of feasibility", JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 98, 2009, pages 4420 - 4428
A. HOMMA; H. SATO; A. OKAMACHI; T. EMURA; T. ISHIZAWA; T. KATO; T. MATSUURA; S. SATO; T. TAMURA; Y. HIGUCHI: "Novel hyaluronic acid-methotrexate conjugates for osteoarthritis treatment", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 17, 2009, pages 4647 - 4656
A. HOMMA; H. SATO; T. TAMURA; A. OKAMACHI; T. EMURA; T. ISHIZAWA; T. KATO; T. MATSUURA; S. SATO; Y. HIGUCHI: "Synthesis and optimization of hyaluronic acid-methotrexate conjugates to maximize benefit in the treatment of osteoarthritis", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 18, 2010, pages 1062 - 1075
A. MYC; J. KUKOWSKA-LATALLO; P. CAO; B. SWANSON; J. BATTISTA; T. DUNHAM; J.R. BAKER, JR.: "Targeting the efficacy of a dendrimer-based nanotherapeutic in heterogeneous xenograft tumors in vivo", ANTI-CANCER DRUGS, vol. 21, 2010, pages 186 - 192
A. WUNDER; G. STEHLE; H.H. SCHRENK; G. HARTUNG; D.L. HEENE; W. MAIER-BORST; H. SINN: "Antitumor activity of methotrexate-albumin conjugates in rats bearing a Walker-256 carcinoma, International journal of cancer", JOURNAL INTERNATIONAL DU CANCER, vol. 76, 1998, pages 884 - 890
A. YACOBI; R.G. STOLL; C.Y. SUM; C.M. LAI; S.D. GUPTA; J.D. HULSE: "Pharmacokinetics of hydroxyethyl starch in normal subjects", JOURNAL OF CLINICAL PHARMACOLOGY, vol. 22, 1982, pages 206 - 212
A.K. LYER; G. KHALED; J. FANG; H. MAEDA: "Exploiting the enhanced permeability and retention effect for tumor targeting", DRUG DISCOVERY TODAY, vol. 11, 2006, pages 812 - 818
A.N. VIS; A. VAN DER GAAST; B.W. VAN RHIJN; T.K. CATSBURG; C. SCHMIDT; G.H. MICKISCH: "A phase II trial of methotrexate-human serum albumin (MTX-HSA) in patients with metastatic renal cell carcinoma who progressed under immunotherapy", CANCER CHEMOTHERAPY AND PHARMACOLOGY, vol. 49, 2002, pages 342 - 345
B. RIHOVA: "Biocompatibility of biomaterials: Hemocompatibility, immunocompatibility and biocompatibility of solid polymeric materials and soluble targetable polymeric carriers", ADVANCED DRUG DELIVERY REVIEWS, vol. 21, 1996, pages 157 - 176
B. WEIDLER; B. VON BORMANN; K. SOMMERMEYER; E. LOHMANN; J. PEIL; G. HEMPELMANN: "Pharmacokinetic parameters as criteria for clinical use of hydroxyethyl starch preparations", ARZNEIMITTEL-FORSCHUNG, vol. 41, 1991, pages 494 - 498
BUDZYNSKA RENATA ET AL: "Antitumor activity of mannan-methotrexate conjugate in vitro and in vivo", ONCOLOGY RESEARCH, vol. 16, no. 9, 2007, pages 415 - 421, XP008163320, ISSN: 0965-0407 *
C. DEDUVE; T. DEBARSY; B. POOLE; A. TROUET; P. TULKENS; F. VANHOOF: "Lysosomotropic Agents", BIOCHEMICAL PHARMACOLOGY, vol. 23, 1974, pages 2495
C. MADJDPOUR; N. DETTORI; P. FRASCAROLO; M. BURKI; M. BOLL; A. FISCH; T. BOMBELI; D.R. SPAHN: "Molecular weight of hydroxyethyl starch: is there an effect on blood coagulation and pharmacokinetics?", BRITISH JOURNAL OF ANAESTHESIA, vol. 94, 2005, pages 569 - 576
C. SAMORI; H. ALI-BOUCETTA; R. SAINZ; C. GUO; F.M. TOMA; C. FABBRO; T. DA ROS; M. PRATO; K. KOSTARELOS; A. BIANCO: "Enhanced anticancer activity of multi-walled carbon nanotube-methotrexate conjugates using cleavable linkers", CHEMICAL COMMUNICATIONS, vol. 46, 2010, pages 1494 - 1496
C. SIRTL; H. LAUBENTHAL; V. ZUMTOBEL; D. KRAFT; W. JURECKA: "Tissue deposits of hydroxyethyl starch (HES): dose-dependent and time-related", BRITISH JOURNAL OF ANAESTHESIA, vol. 82, 1999, pages 510 - 515
C.A. VOLTA; V. ALVISI; M. CAMPI; E. MARANGONI; R. ALVISI; M. CASTELLAZZI; E. FAINARDI; M.C. MANFRINATO; F. DALLOCCHIO; T. BELLINI: "Influence of different strategies of volume replacement on the activity of matrix metalloproteinases: an in vitro and in vivo study", ANESTHESIOLOGY, vol. 106, 2007, pages 85 - 91
C.H. TAKIMOTO; P.M. LORUSSO; L. FORERO; G.H. SCHWARTZ; A.W. TOLCHER; A. PATNAIK; L.A. HAMMOND; S. SYED; C. SIMMONS; M. DUCHARME: "A phase I and pharmacokinetic study of DE-310 administered as a 3 hour infusion every 4 weeks (wks) to patients (pts) with advanced solid tumors or lymphomas.", CLINICAL CANCER RESEARCH, vol. 9, 2003, pages 6105S - 6105S
D. METZE; S. REIMANN; Z. SZEPFALUSI; B. BOHLE; D. KRAFT; T.A. LUGER: "Persistent pruritus after hydroxyethyl starch infusion therapy: a result of long-term storage in cutaneous nerves", THE BRITISH JOURNAL OF DERMATOLOGY, vol. 136, 1997, pages 553 - 559
D. NEVOZHAY; R. BUDZYNSKA; M. JAGIELLO; U. KANSKA; M.S. OMAR; A. OPOLSKI; J. WIETRZYK; J. BORATYNSKI: "The effect of the substitution level of some dextran-methotrexate conjugates on their antitumor activity in experimental cancer models", ANTICANCER RES, vol. 26, 2006, pages 2179 - 2186
D. NEVOZHAY; R. BUDZYNSKA; U. KANSKA; M. JAGIELLO; M.S. OMAR; J. BORATYNSKI; A. OPOLSKI: "Antitumor properties and toxicity of dextran-methotrexate conjugates are dependent on the molecular weight of the carrier", ANTICANCER RES, vol. 26, 2006, pages 1135 - 1143
E. KUMAZAWA; Y. OCHI: "DE-310, a novel macromolecular carrier system for the camptothecin analog DX-8951f: Potent antitumor activities in various murine tumor models", CANCER SCI, vol. 95, 2004, pages 168 - 175
E. NEUMANN; E. FREI; D. FUNK; M.D. BECKER; H.H. SCHRENK; U. MULLER-LADNER; C. FIEHN: "Native albumin for targeted drug delivery", EXPERT OPINION ON DRUG DELIVERY, vol. 7, 2010, pages 915 - 925
F. CANAL; J. SANCHIS; M.J. VICENT: "Polymer-drug conjugates as nano-sized medicines", CURR OPIN BIOTECH, vol. 22, 2011, pages 894 - 900
F. JESCH; G. HUBNER; V. ZUMTOBEL; M. ZIMMERMANN; K. MESSMER: "Hydroxyethyl starch (HAS 450/0.7) in human plasma and liver. Course of concentration and histological changes", INFUSIONSTHERAPIE UND KLINISCHE ERNAHRUNG, vol. 6, 1979, pages 112 - 117
G. PASUT; F.M. VERONESE: "PEGylation for improving the effectiveness of therapeutic biomolecules", DRUGS TODAY (BARC, vol. 45, 2009, pages 687 - 695
G. STEHLE; H. SINN; A. WUNDER; H.H. SCHRENK; S. SCHUTT; W. MAIER-BORST; D.L. HEENE: "The loading rate determines tumor targeting properties of methotrexate-albumin conjugates in rats", ANTI-CANCER DRUGS, vol. 8, 1997, pages 677 - 685
G.W. HALBERT; A.T. FLORENCE; J.F. STUART: "Characterization of in-vitro drug release and biological activity of methotrexate-bovine serum albumin conjugates", THE JOURNAL OF PHARMACY AND PHARMACOLOGY, vol. 39, 1987, pages 871 - 876
H. KAMADA; Y. TSUTSUMI; Y. YAMAMOTO; T. KIHIRA; Y. KANEDA; Y. MU; H. KODAIRA; S.I. TSUNODA; S. NAKAGAWA; T. MAYUMI: "Antitumor activity of tumor necrosis factor-alpha conjugated with polyvinylpyrrolidone on solid tumors in mice", CANCER RESEARCH, vol. 60, 2000, pages 6416 - 6420
H. MAEDA: "Tumor-selective delivery of macromolecular drugs via the EPR effect: background and future prospects", BIOCONJUGATE CHEMISTRY, vol. 21, 2010, pages 797 - 802
H. MAEDA; G.Y. BHARATE; J. DARUWALLA: "Polymeric drugs for efficient tumor-targeted drug delivery based on EPR-effect", EUR J PHARM BIOPHARM, vol. 71, 2009, pages 409 - 419
H. RINGSDORF: "Structure and Properties of Pharmacologically Active Polymers", J POLYM SCI POL SYM, 1975, pages 135 - 153
J. BOLDT: "Modern rapidly degradable hydroxyethyl starches: current concepts", ANESTHESIA AND ANALGESIA, vol. 108, 2009, pages 1574 - 1582
J. BORATYNSKI; A. OPOLSKI; J. WIETRZYK; A. GORSKI; C. RADZIKOWSKI: "Cytotoxic and antitumor effect of fibrinogen-methotrexate conjugate", CANCER LETT, vol. 148, 2000, pages 189 - 195
J. CIEKOT; T. GOSZCZYNSKI; J. BORATYNSKIT: "Methods for methotrexate determination in macromolecular conjugates drug carrier", ACTA POLONIAE PHARMACEUTICA, vol. 69, 2012, pages 1342 - 1346
J. FANG; H. NAKAMURA; H. MAEDA: "The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect", ADVANCED DRUG DELIVERY REVIEWS, vol. 63, 2011, pages 136 - 151
J. KOPECEK, BIOMATERIALS AND DRUG DELIVERY: PAST, PRESENT, AND FUTURE, MOLECULAR PHARMACEUTICS, vol. 7, 2010, pages 922 - 925
J. KOPECEK; P. KOPECKOVA; T. MINKO; Z. LU: "HPMA copolymer-anticancer drug conjugates: design, activity, and mechanism of action", EUR J PHARM BIOPHARM, vol. 50, 2000, pages 61 - 81
J. LEUSCHNER; J. OPITZ; A. WINKLER; R. SCHARPF; F. BEPPERLING: "Tissue storage of 14C-labelled hydroxyethyl starch (HES) 130/0.4 and HES 200/0.5 after repeated intravenous administration to rats", DRUGS IN R&D, vol. 4, 2003, pages 331 - 338
J.A. NAGY; L. BENJAMIN; H. ZENG; A.M. DVORAK; H.F. DVORAK: "Vascular permeability, vascular hyperpermeability and angiogenesis", ANGIOGENESIS, vol. 11, 2008, pages 109 - 119
J.D. HULSE; A. YACOBI: "Hetastarch: an overview of the colloid and its metabolism", DRUG INTELLIGENCE & CLINICAL PHARMACY, vol. 17, 1983, pages 334 - 341
J.L. WIKE-HOOLEY; J. HAVEMAN; H.S. REINHOLD: "The relevance of tumour pH to the treatment of malignant disease", RADIOTHERAPY AND ONCOLOGY : JOURNAL OF THE EUROPEAN SOCIETY FOR THERAPEUTIC RADIOLOGY AND ONCOLOGY, vol. 2, 1984, pages 343 - 366
J.R. BERTINO: "Cancer research: from folate antagonism to molecular targets", BEST PRACTICE & RESEARCH. CLINICAL HAEMATOLOGY, vol. 22, 2009, pages 577 - 582
J.Y. LJUBIMOVA; M. FUJITA; A.V. LJUBIMOV; V.P. TORCHILIN; K.L. BLACK; E. HOLLER: "Poly(malic acid) nanoconjugates containing various antibodies and oligonucleotides for multitargeting drug delivery", NANOMEDICINE (LOND, vol. 3, 2008, pages 247 - 265
J.Y. LJUBIMOVA; M. FUJITA; N.M. KHAZENZON; B.S. LEE; S. WACHSMANN-HOGIU; D.L. FARKAS; K.L. BLACK; E. HOLLER: "Nanoconjugate based on polymalic acid for tumor targeting", CHEMICO-BIOLOGICAL INTERACTIONS, vol. 171, 2008, pages 195 - 203
K. LANG; S. SUTTNER; J. BOLDT; B. KUMLE; D. NAGEL: "Volume replacement with HES 130/0.4 may reduce the inflammatory response in patients undergoing major abdominal surgery", CANADIAN JOURNAL OF ANAESTHESIA = JOURNAL CANADIEN D'ANESTHESIE, vol. 50, 2003, pages 1009 - 1016
K. ULBRICH; T. ETRYCH; P. CHYTIL; M. JELINKOVA; B. RIHOVA: "HPMA copolymers with pH-controlled release of doxorubicin: in vitro cytotoxicity and in vivo antitumor activity", JOURNAL OF CONTROLLED RELEASE : OFFICIAL JOURNAL OF THE CONTROLLED RELEASE SOCIETY, vol. 87, 2003, pages 33 - 47
L. BURES; A. LICHY; J. BOSTIK; M. SPUNDOVA: "The use of protein as a carrier of methotrexate for experimental cancer chemotherapy. V. Alternative method for preparation of serum albumin-methotrexate derivative", NEOPLASMA, vol. 37, 1990, pages 225 - 231
L. GENESTIER; R. PAILLOT; L. QUEMENEUR; K. IZERADJENE; J.P. REVILLARD: "Mechanisms of action of methotrexate", IMMUNOPHARMACOLOGY, vol. 47, 2000, pages 247 - 257
L.E. GERWECK; K. SEETHARAMAN: "Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer", CANCER RESEARCH, vol. 56, 1996, pages 1194 - 1198
M. DUNITZ: "Vivo Cancer Models.", vol. 84-2635, 2003, NIH PUBLICATION, article "Experimental Therapeutics Program", pages: 1976 - 1982
M. JAMNICKI; A. ZOLLINGER; B. SEIFERT; D. POPOVIC; T. PASCH; D.R. SPAHN: "Compromised blood coagulation: an in vitro comparison of hydroxyethyl starch 130/0.4 and hydroxyethyl starch 200/0.5 using thrombelastography", ANESTHESIA AND ANALGESIA, vol. 87, 1998, pages 989 - 993
M. VISENTIN; R.B. ZHAO; I.D. GOLDMAN, THE ANTIFOLATES, HEMATOL ONCOL CLIN N, vol. 26, 2012, pages 629
M. WESTPHAL; M.F. JAMES; S. KOZEK-LANGENECKER; R. STOCKER; B. GUIDET; H. VAN AKEN: "Hydroxyethyl starches: different products--different effects", ANESTHESIOLOGY, vol. 111, 2009, pages 187 - 202
M.G. FURY; D.G. PFISTER: "Current recommendations for systemic therapy of recurrent and/or metastatic head and neck squamous cell cancer", JOURNAL OF THE NATIONAL COMPREHENSIVE CANCER NETWORK : JNCCN, vol. 9, 2011, pages 681 - 689
M.J. VICENT; R. DUNCAN: "Polymer conjugates: nanosized medicines for treating cancer", TRENDS BIOTECHNOL, vol. 24, 2006, pages 39 - 47
M.R. KANO; Y. BAE; C. IWATA; Y. MORISHITA; M. YASHIRO; M. OKA; T. FUJII; A. KOMURO; K. KIYONO; M. KAMINISHI: "Improvement of cancer-targeting therapy, using nanocarriers for intractable solid tumors by inhibition of TGF-B signaling", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 104, 2007, pages 3460 - 3465
N. ISMAILI; M. AMZERIN; A. FLECHON: "Chemotherapy in advanced bladder cancer: current status and future", JOURNAL OF HEMATOLOGY & ONCOLOGY, vol. 4, 2011, pages 35
N. LARSON; H. GHANDEHARI: "Chemistry of materials : a publication of the American Chemical Society", vol. 24, 2012, article "Polymeric conjugates for drug delivery", pages: 840 - 853
NEVOZHAY DMITRY ET AL: "Antitumor properties and toxicity of dextran-methotrexate conjugates are dependent on the molecular weight of the carrier", ANTICANCER RESEARCH, vol. 26, no. 2A, March 2006 (2006-03-01), pages 1135 - 1143, XP008163318, ISSN: 0250-7005 *
P. HARMATZ; R.W. GRADY; P. DRAGSTEN; E. VICHINSKY; P. GIARDINA; J. MADDEN; M. JENG; B. MILLER; G. HANSON; B. HEDLUND: "Phase Ib clinical trial of starch-conjugated deferoxamine (40SD02): a novel long-acting iron chelator", BRITISH JOURNAL OF HAEMATOLOGY, vol. 138, 2007, pages 374 - 381
P. VAUPEL; F. KALLINOWSKI; P. OKUNIEFF: "Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review", CANCER RESEARCH, vol. 49, 1989, pages 6449 - 6465
P.R. DRAGSTEN; P.E. HALLAWAY; G.J. HANSON; A.E. BERGER; B. BERNARD; B.E. HEDLUND: "First human studies with a high-molecular-weight iron chelator", THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE, vol. 135, 2000, pages 57 - 65
QING LUO ET AL: "A novel 5-fluorouracil prodrug using hydroxyethyl starch as a macromolecular carrier for sustained release", CARBOHYDRATE POLYMERS, APPLIED SCIENCE PUBLISHERS, LTD. BARKING, GB, vol. 87, no. 4, 13 November 2011 (2011-11-13), pages 2642 - 2647, XP028349373, ISSN: 0144-8617, [retrieved on 20111122], DOI: 10.1016/J.CARBPOL.2011.11.039 *
R. BUDZYNSKA; D. NEVOZHAY; U. KANSKA; M. JAGIELLO; A. OPOLSKI; J. WIETRZYK; J. BORATYNSKI: "Antitumor activity of mannan-methotrexate conjugate in vitro and in vivo", ONCOLOGY RESEARCH, vol. 16, 2007, pages 415 - 421
R. DUNCAN: "Polymer conjugates as anticancer nanomedicines", NAT REV CANCER, vol. 6, 2006, pages 688 - 701
R. DUNCAN: "Polymer therapeutics as nanomedicines: new perspectives", CURR OPIN BIOTECH, vol. 22, 2011, pages 492 - 501
R. DUNCAN: "The dawning era of polymer therapeutics", NATURE REVIEWS. DRUG DISCOVERY, vol. 2, 2003, pages 347 - 360
R. DUNCAN; M.J. VICENT: "Polymer therapeutics-prospects for 21st century: The end of the beginning", ADVANCED DRUG DELIVERY REVIEWS, 2012
R. DUNCAN; M.J. VICENT; F. GRECO; R.I. NICHOLSON: "Polymer-drug conjugates: towards a novel approach for the treatment of endrocine-related cancer", ENDOCRINE-RELATED CANCER, vol. 12, 2005, pages 189 - 199
R. HAAG; F. KRATZ: "Polymer therapeutics: concepts and applications", ANGEW CHEM INT ED ENGL, vol. 45, 2006, pages 1198 - 1215
S. DANHAUSERRIEDL; E. HAUSMANN; H.D. SCHICK; R. BENDER; H. DIETZFELBINGER; J. RASTETTER; A.R. HANAUSKE: "Phase-I Clinical and Pharmacokinetic Trial of Dextran Conjugated Doxorubicin (Ad-70, Dox-Oxd", INVEST NEW DRUG, vol. 11, 1993, pages 187 - 195
S. STANDER; Z. SZEPFALUSI; B. BOHLE; H. STANDER; D. KRAFT; T.A. LUGER; D. METZE: "Differential storage of hydroxyethyl starch (HES) in the skin: an immunoelectron-microscopical long-term study", CELL AND TISSUE RESEARCH, vol. 304, 2001, pages 261 - 269
S.A. KOZEK-LANGENECKER: "Effects of hydroxyethyl starch solutions on hemostasis", ANESTHESIOLOGY, vol. 103, 2005, pages 654 - 660
S.D. CHIPMAN; F.B. OLDHAM; G. PEZZONI; J.W. SINGER: "Biological and clinical characterization of paclitaxel poliglumex (PPX, CT-21 03), a macromolecular polymer-drug conjugate", INTERNATIONAL JOURNAL OF NANOMEDICINE, vol. 1, 2006, pages 375 - 383
S.D. GANDHI; R.B. WEISKOPF; C. JUNGHEINRICH; R. KOORN; D. MILLER; R.E. SHANGRAW; D.S. PROUGH; D. BAUS; F. BEPPERLING; D.C. WARLTIE: "Volume replacement therapy during major orthopedic surgery using Voluven (hydroxyethyl starch 130/0.4) or hetastarch", ANESTHESIOLOGY, vol. 106, 2007, pages 1120 - 1127
S.N. KHLEIF; G.A. CURT: "Animal models in drug development.", 1993, LEA & FEBIGER
T. MASUKO; A. MINAMI; N. IWASAKI; T. MAJIMA; S. NISHIMURA; Y.C. LEE: "Carbohydrate analysis by a phenol- sulfuric acid method in microplate format", ANALYTICAL BIOCHEMISTRY, vol. 339, 2005, pages 69 - 72
T.M. ALLEN; P.R. CULLIS: "Drug delivery systems: entering the mainstream", SCIENCE, vol. 303, 2004, pages 1818 - 1822
U. KANSKA; M.S. OMAR; R. BUDZYNSKA; D. NEVOZHAY; M. JAGIELLO; A. OPOLSKI; J. BORATYNSKI: "Antileukemic activity of glycated fibrinogen-methotrexate conjugates", ANTICANCER RES, vol. 25, 2005, pages 2229 - 2234
WHITELEY J M ET AL: "Folate antagonists covalently linked to carbohydrates: Synthesis, properties, and use in the purification of dihydrofolate reductases", ARCHIVES OF BIOCHEMISTRY AND BIOPHYSICS, ACADEMIC PRESS, US, vol. 150, no. 1, 1 May 1972 (1972-05-01), pages 15 - 22, XP024809404, ISSN: 0003-9861, [retrieved on 19720501], DOI: 10.1016/0003-9861(72)90004-5 *
Y. CHAU; F.E. TAN; R. LANGER: "Synthesis and characterization of dextran-peptide-methotrexate conjugates for tumor targeting via mediation by matrix metalloproteinase II and matrix metalloproteinase IX", BIOCONJUGATE CHEMISTRY, vol. 15, 2004, pages 931 - 941
Y. CHAU; N.M. DANG; F.E. TAN; R. LANGER: "Investigation of targeting mechanism of new dextran-peptide-methotrexate conjugates using biodistribution study in matrix-metalloproteinase-overexpressing tumor xenograft model", JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 95, 2006, pages 542 - 551
Y. CHAU; R.F. PADERA; N.M. DANG; R. LANGER: "Antitumor efficacy of a novel polymer-peptide-drug conjugate in human tumor xenograft models", INTERNATIONAL JOURNAL OF CANCER, vol. 118, 2006, pages 1519 - 1526
Y. CLAES; J. VAN HEMELRIJCK; M. VAN GERVEN; J. ARNOUT; J. VERMYLEN; B. WEIDLER; H. VAN AKEN: "Influence of hydroxyethyl starch on coagulation in patients during the perioperative period", ANESTHESIA AND ANALGESIA, vol. 75, 1992, pages 24 - 30
Y. MATSUMURA; H. MAEDA: "A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs", CANCER RESEARCH, vol. 46, 1986, pages 6387 - 6392
Y. MOLIN; J. FAYETTE: "Current chemotherapies for recurrent/metastatic head and neck cancer", ANTI-CANCER DRUGS, vol. 22, 2011, pages 621 - 625
Y. TAKAKURA; M. HASHIDA: "Macromolecular carrier systems for targeted drug delivery: pharmacokinetic considerations on biodistribution", PHARMACEUTICAL RESEARCH, vol. 13, 1996, pages 820 - 831
Y.H. ZHANG; T.P. THOMAS; K.H. LEE; M.H. LI; H. ZONG; A.M. DESAI; A. KOTLYAR; B.H. HUANG; M.M.B. HOLL; J.R. BAKER: "Polyvalent saccharide-functionalized generation 3 poly(amidoamine) dendrimer-methotrexate conjugate as a potential anticancer agent", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 19, 2011, pages 2557 - 2564

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10874753B2 (en) 2014-09-26 2020-12-29 The South African Nuclear Energy Corporation Limited Radiopharmaceutical conjugate of a metabolite and an EPR agent, for targeting tumour cells
CN112094320A (en) * 2019-06-18 2020-12-18 首都医科大学 His-Gly-Glu modified methotrexate, synthesis, antitumor activity and application thereof
CN112094320B (en) * 2019-06-18 2022-08-02 首都医科大学 His-Gly-Glu modified methotrexate, synthesis, antitumor activity and application thereof
CN113041359A (en) * 2021-03-26 2021-06-29 湖南师范大学 Glutathione-responsive osteosarcoma-resistant prodrug nanoparticle and preparation method and application thereof
CN113041359B (en) * 2021-03-26 2022-05-20 湖南师范大学 Glutathione-responsive osteosarcoma-resistant prodrug nanoparticle and preparation method and application thereof
WO2022260545A1 (en) * 2021-06-11 2022-12-15 Uniwersytet Medyczny Im. Piastów Śląskich We Wrocławiu Synthesis of a novel glucose-conjugated methotrexate and its application for the treatment and prevention of neoplasms

Also Published As

Publication number Publication date
PL398253A1 (en) 2013-09-02
PL228735B1 (en) 2018-04-30

Similar Documents

Publication Publication Date Title
Fan et al. pH sensitive polymeric complex of cisplatin with hyaluronic acid exhibits tumor-targeted delivery and improved in vivo antitumor effect
Nakamura et al. Two step mechanisms of tumor selective delivery of N-(2-hydroxypropyl) methacrylamide copolymer conjugated with pirarubicin via an acid-cleavable linkage
Shiah et al. Biodistribution and antitumour efficacy of long-circulating N-(2-hydroxypropyl) methacrylamide copolymer–doxorubicin conjugates in nude mice
Mezghrani et al. Hepatocellular carcinoma dually-targeted nanoparticles for reduction triggered intracellular delivery of doxorubicin
EP0662002B1 (en) Alginate-bioactive agent conjugates
CA2616957C (en) Antitumoral bioconjugates of hyaluronic acid or its derivatives obtained by indirect chemical conjugation
US20040028745A1 (en) Hydrogels and water soluble polymeric carriers for durg delivery
US20060127310A1 (en) Amplification of biotin-mediated targeting
Lin et al. Improved targeting of platinum chemotherapeutics: the antitumour activity of the HPMA copolymer platinum agent AP5280 in murine tumour models
Cheng et al. Construction and evaluation of PAMAM–DOX conjugates with superior tumor recognition and intracellular acid-triggered drug release properties
CN107096038B (en) Preparation method of cross-linked nano-drug based on active reaction type one-step method
Zhang et al. An intelligent cell-selective polymersome-DM1 nanotoxin toward triple negative breast cancer
WO2013127885A1 (en) A conjugate of methotrexate and hydroxyethyl starch for use in the treatment cancer
Park et al. Self-assembled nanocomplex of PEGylated protamine and heparin–suramin conjugate for accumulation at the tumor site
Xie et al. Targeted nanoparticles from xyloglucan–doxorubicin conjugate loaded with doxorubicin against drug resistance
Gaál et al. Low toxicity and high antitumour activity of daunomycin by conjugation to an immunopotential amphoteric branched polypeptide
Ciekot et al. Hydroxyethylcellulose as a methotrexate carrier in anticancer therapy
EA007353B1 (en) Ph-sensitive polymeric conjugates of anthracycline cancerostatic active agents for targeted therapy
US9192677B2 (en) Biodegradable, water soluble and pH responsive poly(organo)phosphazenes
Park et al. Ternary biomolecular nanoparticles for targeting of cancer cells and anti-angiogenesis
KR20200015037A (en) A photothermal nanocomplex comprising photothermal nanoparticle, anticancer drug and conjugate of hyaluronic acid and PEG
JP2003504312A (en) Biologically active material
JP2018070459A (en) Medical drug composite, polymer carrier, method for producing same, and therapeutic agent of cancer, rheumatism or inflammation
CN111920959B (en) ROS (reactive oxygen species) -responsive heparin polysaccharide nano prodrug compound based on click reaction, nano preparation, and preparation method and application thereof
WO2003094929A2 (en) Preblocking with non-ha gags increases effectiveness of ha conjugated anticancer agents

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13719221

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13719221

Country of ref document: EP

Kind code of ref document: A1