US20030148933A1 - Derivatives of dehydrodidemnin B - Google Patents

Derivatives of dehydrodidemnin B Download PDF

Info

Publication number
US20030148933A1
US20030148933A1 US10/357,759 US35775903A US2003148933A1 US 20030148933 A1 US20030148933 A1 US 20030148933A1 US 35775903 A US35775903 A US 35775903A US 2003148933 A1 US2003148933 A1 US 2003148933A1
Authority
US
United States
Prior art keywords
cells
dehydrodidemnin
ddb
alkyl
acyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/357,759
Inventor
Kenneth Rinehart
Anna Lithgow-Bertelloni
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pharmamar SA
Original Assignee
Pharmamar SA
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
Priority claimed from PCT/GB1990/001495 external-priority patent/WO1991004985A1/en
Application filed by Pharmamar SA filed Critical Pharmamar SA
Priority to US10/357,759 priority Critical patent/US20030148933A1/en
Publication of US20030148933A1 publication Critical patent/US20030148933A1/en
Priority to US11/136,622 priority patent/US7348311B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K11/00Depsipeptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K11/02Depsipeptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof cyclic, e.g. valinomycins ; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • C07K5/0205Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link containing the structure -NH-(X)3-C(=0)-, e.g. statine or derivatives thereof

Definitions

  • This invention relates to dehydrodidemnin B and, in particular, to the isolation of dehydrodidemnin B, a cyclic depsipeptide, from a tunicate of the Ascidiacea class.
  • This novel compound has been shown to have antiviral, antitumoral and cytotoxic activities.
  • the didemnins form a class of cyclic depsipeptides which have been isolated from various species of the Trididemnum genus. They have been shown to have potent activity against viruses and tumor cells (Rinehart, Jr., et al., J. Am. Chem. Soc., 103, 1857-59 (1981). Didemnin B, up to now the most active compound of this class, has been shown to have potent immunosuppressive activity (Montgomery et al., Transplantation, 40, 49-56 (1985) and a more potent inhibition of binding of prolactin to human lymphocytes than other didemnin compounds (Montgomery et al., Fed. Prac., 44, 634 (1987).
  • This invention provides a novel and more active compound of this class, unexpectedly isolated from the Mediterranean tunicate Alpidium albicans , namely dehydrodidemnin B (or “DDB”), having the formula:
  • R is hydrogen; and derivatives thereof with the same class of biological activity, i.e., where R is Acyl, Alkyl or Aryl.
  • FIGS. 1A and 1B each illustrate a scheme by which DDB can be isolated from tunicates of the Ascidiacea class, subphylum Urochordata;
  • FIGS. 2 - 5 are mass spectra of compounds disclosed herein;
  • FIGS. 6 and 7- 9 are 1 H NMR spectra of compounds disclosed herein.
  • FIGS. 10 and 11- 14 are 13 C NMR spectrum of compounds disclosed herein;
  • DDB The compound of the present invention, is characterized by the following properties, considering also that, in solution, two conformers (at least) are possible:
  • TLC R f 0.4; 0.35 (Silica gel, 2:3, CH 2 Cl 2 /EtOAc); 0.5; 0.44 (Silica gel; 9:1, CHCl 3 /MeOH);
  • DDB The structure determination of DDB was accomplished by comparison of mass spectrometry low and high resolution FABMS (Rinehart, Jr. et al., Pure and Appl. Chem., 54, 2409-2424 (1982)) and NMR data with other didemnin data, and confirmed by synthesis of DDB involving coupling of natural didemnin A with the appropriate side chain.
  • the low resolution FAB mass spectra showed peaks at m/z 1110 (M+H), 816 (M+2H ⁇ side chain) and 295 (side chain).
  • the compound of this invention has been shown to inhibit in vitro L1210 and P-388 mouse leukemia cells; L-929, mouse areolar and adipose tissue, B-16, mouse melanoma cells; A-549, human lung carcinoma cells; HeLa, human cervix epithelioid carcinoma cells and KB, human cervix epithelioid carcinoma cells and KB, human oral epidermoid carcinoma cells, and in vivo, P-338, mouse leukemia cells, Lewis lung carcinoma cells and B-16 melanoma cells.
  • DDB is useful as an antitumor agent and therefore is useful inhibiting the growth of tumor cells in mammals exhibiting such tumor cells.
  • Dehydrodidemnin B like didemnin B, (Montgomery et al., Transplantation, 40, 49-56 (1985)), is a powerful immunomodulator.
  • Dehydrodidemnin B has also shown activity against Herpes simplex virus, type 1, in CV-1 cells (monkey kidney cells); thus it is also useful as an antiviral agent.
  • the IC 50 determined was 60 ng/mL (e.g., 10 fold greater than for L-1210 cells) and 1 ⁇ g/mL, respectively.
  • the compound of present invention is preferably presented for administration to humans and animals in unit dosage form in pharmaceutically appropriate carriers containing the active ingredient in the appropriate quantity.
  • dosage levels of the administered active ingredient can be intravenous 0.05 to about 50 mg/Kg, intraperitoneal, subcutaneous and intramuscular 1 to 100 mg/Kg; oral 1 to 150 mg/Kg of animal (body) weight.
  • DDB is useful to inhibit the growth of cancer cells in animals or humans bearing a neoplastic disease, for example, acute myelocytic leukemia, acute lymphocytic leukemia, malignant melanoma, adenocarcinoma of the lung, small cell carcinoma of the lung, and the like.
  • a neoplastic disease for example, acute myelocytic leukemia, acute lymphocytic leukemia, malignant melanoma, adenocarcinoma of the lung, small cell carcinoma of the lung, and the like.
  • the compound can be isolated from tunicates of the genus Aplidium, and more especially from the species Aplidium albicans .
  • the species is found in the Iberian Mediterranean Coast as well as in the Balearic Islands. The species has been also found in Great Britain, English Channel as well as in the Africa Coast and Portugal. It seems to prefer detritic, coralligenic and sciafilae algae communities. They also can be found in more photophilic habitats.
  • Colonies of the tunicate are generally flat and lobed (2.5 cm diameter). It is jelly like, totally encrusted with sand which confers a sandy color to the colony. Zooides are of a whitish color 10 mm long; the oral siphon has 6 lobes, and the cloacal languaget is trifid, which is a species characteristic. Generally there are 10-11 rows of stigmas. The stomach has 6 marked folds. Gonads are of the family type with one or several ovocites below the digestive track and numerous testicular follicles forming one or double row in the post abdomen. Larvae are incubated in the number of 1 to 9 in the atrial cavity; they have 3 cupping-glasses and several vesicular formations in the anterior part.
  • isolation method generally comprises alcoholic extraction of the homogenized tunicate and selective purification of the desired DDB.
  • the tunicate was extracted with MeOH, filtered and dissolved in MeOH: Toluene 3:1 and partitioned with 10% NaNO 3 .
  • the aqueous layer was successively extracted with CH 2 Cl 2 , EtOAc and n-BuOH.
  • the organic fractions were combined after monitoring by normal-phase TLC developed CHCl 3 :MeOH 9:1, affording a 2:1 (v/v) and the activity was concentrated in the methanolic layer.
  • the polar fraction is passed through Silica gel Step-gradient Chromatography.
  • the last fraction is further purified by reverse phase HPLC at a flow rate of 2 ml/min. Two mean peaks were collected and readily interconverted to a mixture of I and II, to establish an approximately 1:1 ratio.
  • the DDB can also be prepared by total synthesis, or semisynthesis from natural Didemnin A, following in both cases standard procedures of protection and activation in peptide chemistry.
  • Pro-OBzl in DMF is mixed with pyruvic acid and HOBt, and DCC in CH 2 Cl 2 added.
  • the reaction product can be purified and shows the chemical and physical properties corresponding to Pyruvyl-Pro-OBzl.
  • DDB Apart from DDB itself, the present invention extends to derivatives of DDB, comprising acylated, alkylated or arylated derivatives of DDB, where R could be a group COR′ or R′, where R′ represents the following substituents:
  • R 1 R 2 , R 3 could be alkyl (either linear or branched), aryl or alkylaryl, the aryl groups, bearing or not the substituents described under R′.
  • the residues R 1 , R 2 , R 3 could be either the same or different.
  • the derivatives can be more preferably alkyl, aryl or acyl-derivatives, where R′ is an aliphatic or aromatic group, more preferably a 1-6 carbon atom residue.
  • the acyl derivatives can be obtained by treatment of the parent compound with the corresponding carboxylic anhydride in the presence of pyridine or other nitrogenated organic base; by reaction of DDB with the respective acylchloride; or by dehydration with DCC from DDB and the corresponding carboxylic acid.
  • alkyl or aryl derivatives (R/R′) they can be obtained by reaction of DDB with the corresponding halide, in the presence of an alkaline-organic weak base or by dehydration between DDB and alkyl or aryl hydroxy derivative by an organic dehydrating agent.
  • NMR spectra were obtained with a General Electric QE-300 (300 MHz, 1 H), a Nicolet NT-360 (360 MHz, 1 H) or a General Electric GN 500 (500 MHz, 1 H) at the University of Illinois or a Varian Unity 300 (300 MHz, 1 H and 75 MHz, 13 C at PharmaMar, S. A. (Madrid, Spain) Chemical shifts are reported in ppm referenced to the chloroform peak at ⁇ 7.26 ppm for 1 H. FABMS were measured on a VG Analytical ZAB at the Mass Spectrometry laboratory of the University of Illinois.
  • GC analyses were carried out using a Varian GC (Model 3700) equipped with an Alltech Associates, Inc., Chirasil-Val II capillary column (25 m ⁇ 0.32 mm) with Helium gas carrier at a flow rate of 1.2 ml/min with programmed oven temperature (90° C., 4° C./min, 180° C.).
  • Reversed-phrase HPLC was performed on a system equipped with an Altex pump (Model 110 A) and a Waters Associates differential refractometer (Model R-401) and an Alltech Spherisorb C18 column (25 cm ⁇ 1 cm, particle size 10 ⁇ m) with MeOH: H 2 O 0:1 as the solvent system.
  • DDB The structure of DDB has been determined by physical and spectroscopic methods.
  • TLC R f 0.4; 0.35 (Silica gel, 2:3, CH 2 Cl 2 /EtOAc); 0,5; 0.44 (Silica gel; 9:1, CHCl 3 /MeOH);
  • dehydrodidemnin B can be confirmed also by comparison of the acetylation product with the acetyl derivative of didemnin B.
  • DDB The structure of DDB can also be determined by identification of the individual subunits by total hydrolysis and conversion of the amino acids to their N-trifluoroacetyl methyl esters and analysis by GC.
  • amino acids were identified by their retention times and comparison of authentic samples obtained from the conversion of didemnin B to the N-trifluoroacetyl methyl esters of the amino acids.
  • L-1210 cells were seeded into 16 mm wells at 1 ⁇ 10 4 cells per well in 1 mL aliquots of MEN 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. Cells were counted after three drugs was counted daily to ensure that the cells remained in exponential growth over the period of observation. Growth Inhibition of L-1210 cells by DDB net increase in cell ng/mL DDB number % Inhibition 0 2.9 ⁇ 10 5 0 0.05 2.7 ⁇ 10 5 7 0.1 2.7 ⁇ 10 5 7 0.2 2.1 ⁇ 10 5 28 0.5 1.0 ⁇ 10 5 66 1 2.5 ⁇ 10 4 91 2 6.3 ⁇ 10 3 98
  • P-388 cells were seeded into 16 mm wells at 1 ⁇ 10 4 cells per well in 1 mL aliquots of MEM 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. Cells were counted after three days of incubation. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period of observation. Growth Inhibition of P-388 cells by DDB net increase in cell ng/mL DDB number % Inhibition 0 5.63 ⁇ 10 5 0 0.12 3.97 ⁇ 10 5 29 0.25 1.27 ⁇ 10 5 77 0.5 4.47 ⁇ 10 5 92
  • L-929 cells were seeded into 16 mm wells at 1 ⁇ 10 4 cells per well in 1 mL aliquots of MEM 10C. The following day, medium was replaced with 0.5 mL aliquots of MEM 10C. The following day, the medium was replaced with 0.5 mL aliquots of MEM 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period of observation. Cells were trypsinized and counted 4 days after seeding. Growth Inhibition of L-929 cells by DDB net increase in cell ng/mL DDB number % Inhibition 0 3.17 ⁇ 10 5 0 1 2.31 ⁇ 10 5 27 2.5 1.13 ⁇ 10 5 64 5 5 ⁇ 10 5 84
  • B-16 cells were seeded into 16 mm wells at 1 ⁇ 10 4 cells per well in 1 mL aliquots of MEM 10C determinations were carried out in triplicate. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period for observation. Cells were trypsinized and counted 4 days after seeding. Growth Inhibition of B-16 cells by DDB net increase in cell ng/mL DDB number % Inhibition 0 1.71 ⁇ 10 5 0 0.16 1.71 ⁇ 10 5 0 0.12 1.27 ⁇ 10 5 25 0.25 8.25 ⁇ 10 4 54 0.5 4.50 ⁇ 10 4 74 1.0 2.88 ⁇ 10 4 83
  • A-549 cells were seeded into 16 mm wells at 1 ⁇ 10 4 cells per well in 1 mL aliquots of MEM 10C. The following day, the medium was replaced with 0.5 mL aliquots of MEM 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period of observation. Cells were trypsinated and counted 4 days after seeding.
  • HeLa cells were seeded into 16 mm wells at 1 ⁇ 10 4 cells per well in 1 mL aliquots of MEM 10C. The following day, the medium was replaced with 0.5 mL aliquots of MEM 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period of observation. Cells were trypsinized and counted 4 days after seeding. Growth Inhibition of KB cells by DDB net increase in cell ng/mL DDB number % Inhibition 0 4.50 ⁇ 10 4 0 2.5 4.57 ⁇ 10 4 0 5 2.40 ⁇ 10 4 46 10 1.02 ⁇ 10 4 77
  • Dehydrodidemnin B is active as an immunosuppressive agent. In the mixed lymphocyte reaction it suppresses the immune reaction of murine cells. It also inhibits the growth of murine T-cells and B-cells.
  • a white solitary tunicate was collected near Ibiza in the Balearic Islands (Spain) and was identified by Dr. Xavier Turon of the Universitat de Barcelona, Barcelona (Spain) as Aplidium albicans .
  • a sample is preserved at Centre d'Etudes Avancats, Blanes (Germona, Spain).
  • Preliminary tests on shipboard indicated antiviral activity against VSV-1 (Vesicular stomatitis virus). Further studies in the laboratory confirmed the antiviral activity against Herpes simplex virus, type 1 (HSV-1) in monkey kidney cells (CV-1) and also showed cytotoxicity against mouse lymphoid leukemia in vitro (L1210 line cells).
  • the frozen tunicate was extracted with methanol. Solvent partitioning of the residue afforded three active fractions which were combined according to their similarity in TLC (Thin Layer Chromatography). The crude active fraction was portioned and the activity concentrated in the methanolic layer. The methanol layer was chromatographed by silica gel gravity column (chloroform and chloroform-methanol mixtures), affording one active fraction which was further purified by Reversed-Phase High-Performance Liquid Chromatography (RPC 18 HPLC), affording two peaks (I and II). Analysis by TLC revealed two identical spots in each HPLC fraction. Re-injection of each individual fraction led to two peaks with the same retention times as I and II. Co-injection of I and II confirmed the presence of two identical peaks (possible conformers) in each fraction suggesting a rapid interconversion of I to II and vice versa.
  • Dehydrodidemnin B can also be obtained and its structure confirmed by comparison with a semisynthetic sample prepared by coupling of the appropriate side chain to natural didemnin A.
  • the data obtained for the semisynthetic sample totally agreed with data for natural DDB.
  • R f 0.4163 (19:1:0.5, CHCl 3 /MeOH/AcOH;
  • EDC (1-ethyl-3-(3-dimethylaminopropyl)carbodiimide, 4.27 g, 22.3 mmol) was added to a solution of Pyrvu-Pro (8.2 g, 44.5 mmol) in dry CH 2 Cl 2 (40 mL) at 10° C. with stirring. The mixture was stirred for 2 h at 10° C. and then cooled to 0° C. Didemnin A (1.4 g, 1.48 mmol) in CH 2 Cl 2 -DMF (10 mL, 4:1) was added, and the clear solution was stirred at 0° C. for 2 h and then left in the refrigerator overnight.

Abstract

Dehydrodidemnin with useful biological activity has formula (1). It can be isolated from natural sources or synthesized, and it forms active derivatives.
Figure US20030148933A1-20030807-C00001

Description

    FIELD OF THE INVENTION
  • This invention relates to dehydrodidemnin B and, in particular, to the isolation of dehydrodidemnin B, a cyclic depsipeptide, from a tunicate of the Ascidiacea class. This novel compound has been shown to have antiviral, antitumoral and cytotoxic activities. [0001]
  • BACKGROUND OF THE INVENTION
  • The didemnins form a class of cyclic depsipeptides which have been isolated from various species of the Trididemnum genus. They have been shown to have potent activity against viruses and tumor cells (Rinehart, Jr., et al., [0002] J. Am. Chem. Soc., 103, 1857-59 (1981). Didemnin B, up to now the most active compound of this class, has been shown to have potent immunosuppressive activity (Montgomery et al., Transplantation, 40, 49-56 (1985) and a more potent inhibition of binding of prolactin to human lymphocytes than other didemnin compounds (Montgomery et al., Fed. Prac., 44, 634 (1987).
  • SUMMARY OF THE INVENTION
  • This invention provides a novel and more active compound of this class, unexpectedly isolated from the Mediterranean tunicate [0003] Alpidium albicans, namely dehydrodidemnin B (or “DDB”), having the formula:
    Figure US20030148933A1-20030807-C00002
  • where R is hydrogen; and derivatives thereof with the same class of biological activity, i.e., where R is Acyl, Alkyl or Aryl.[0004]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. 1A and 1B each illustrate a scheme by which DDB can be isolated from tunicates of the Ascidiacea class, subphylum Urochordata; [0005]
  • FIGS. [0006] 2-5 are mass spectra of compounds disclosed herein;
  • FIGS. 6 and 7-[0007] 9 are 1H NMR spectra of compounds disclosed herein; and
  • FIGS. 10 and 11-[0008] 14 are 13C NMR spectrum of compounds disclosed herein;
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The compound of the present invention, DDB, is characterized by the following properties, considering also that, in solution, two conformers (at least) are possible: [0009]
  • TLC R[0010] f=0.4; 0.35 (Silica gel, 2:3, CH2Cl2/EtOAc); 0.5; 0.44 (Silica gel; 9:1, CHCl3/MeOH);
  • RP-HPLC t[0011] R=10.7; 11.9 min (Spherisorb C18 column, 250 mm×10 mm, 10 μm particle size, 9.1, MeOH/H2); 2 mL/min);
  • [α][0012] D 25°=−86° (c 1, MeOH);
  • HR FABMS (M+M) C[0013] 57H88N7O15 m/z calcd. 1110.6366; (M−side chain+H): C42H66N5O11 m/z calcd. 816.4781 (found 816.4755); (M−chain): C15H23N2O4 m/z calcd. 295.1657 (found 295.1657);
  • IR (CHCl[0014] 3) vmax cm−1 3680, 3600, 2970, 2940, 2880, 1740, 1650, 1605, 1540, 1510;
  • [0015] 1H NMR (CDCl3, δ, ppm): 7.82 (d, J=9 Hz, 1H); 7.79 (d, J=9 Hz, 1H); 7.62 (d, J=6 Hz, 1H); 7.21 (d, J=9 Hz, 1H); 7.19 (d, J-9 Hz, 1H); 7.08 (d, J=8.5 Hz, 2H); 6.85 (d, J=8.5 Hz, 2H); 3.77 (s, 3H); 3.13 (s, 3H); 3.08 (s, 3H); 2.54 (s, 3H); 2.50 (s, 3H); 2.1 (s, 3H); 2.02 (s, 3H); 0.82-0.88 (overlapped d and t, 3OH); and
  • [0016] 13C NMR (CDCl3, δ, ppm): 204.93 (s); 204.77 (s); 201.23 (s); 197.55 (s); 173.05 (s); 173.05 (s); 172.36 (s); 171.84 (s); 171.21 (s); 171.16 (s); 170.59 (s); 169.58 (s); 169.51 (s); 169.35 (s); 168.36 (s); 168.28 (s); 161.31 (s); 161.06 (s); 158.64 (s); 158.62 (s), 130.31 (d); 114.12 (d); 114.01 (d); 81.47 (d), 81.43 (d); 70.68 (d); 70.33 (d); 67.97 (d); 67.76 (d); 66.38 (d); 66.22 (d); 60.39 (t); 50.88 (d); 57.80 (d); 66.38 (d); 66.22 (d); 60.39 (t); 50.88 (d); 57.80 (d); 57.45 (d); 57.26 (d); 57.18 (d); 57.12 (d); 55.61 (d); 55.57 (d); 55.26 (q); 54.65 (d); 49.55 (d); 49.49 (d); 48.85 (t); 48.41 (t); 46.98 (t); 41.29 (t); 41.24 (t); 38.78 (q); 38.74 (q); 38.68 (q); 36.42 (t); 36.22 (t); 34.06 (d); 33.99 (d); 33.96 (t); 31.57 (d); 31.38 (q); 31.34 (q); 31.30 (q); 30.69 (d); 29.68 (t); 29.64 (d); 27.28 (t); 27.94 (t); 27.30 (t); 27.17 (t); 27.08 (t); 25.91 (t); 25.87 (t); 25.73 (d); 25.68 (d); 25.63 (d); 25.52 (d); 25.48 (d); 24.80 (q); 24.70 (q); 24.44 (q); 24.31 (q); 22.21 (q); 22.12 (q); 21.92 (q); 21.79 (q); 21.76 (q); 19.46 (q); 17.76 (q); 17.72 (q); 17.18 (q); 16.87 (q); 16.08 (q); 15.62 (q); 15.48 (q); 15.05 (q); 12.55 (q); 12.50 (q).
  • The structure determination of DDB was accomplished by comparison of mass spectrometry low and high resolution FABMS (Rinehart, Jr. et al., [0017] Pure and Appl. Chem., 54, 2409-2424 (1982)) and NMR data with other didemnin data, and confirmed by synthesis of DDB involving coupling of natural didemnin A with the appropriate side chain. The low resolution FAB mass spectra showed peaks at m/z 1110 (M+H), 816 (M+2H−side chain) and 295 (side chain). The lack of two mass units in the molecular ion and side chain peaks, in addition to the same m/z ratio for the ring, suggested that the difference between dehydrodidemnin B and didemnin B was represented by one more degree of unsaturation in the side chain. The molecular formula deduced from high resolution FABMS was C57H88N7O15 (M+H, Δ2.8 mmu); and for the fragment ions corresponding to the ring and the side chain; C42H66N5O11 (Δ0.4 mmu) and C15H23N2O4 (Δ2.6 mmu), respectively. Tandem mass spectrometry on these peaks showed the typical cleavage pattern of didemnins.
  • From the NMR data, the presence of peptide linkages was indicated by peaks near δ8 ppm and the methyl signals corresponding to the amino acid residues. Even though some of these peaks are doubled or tripled due to the presence of, at least two main conformers in solution at room temperature, these peaks are very similar to those of didemnins. The main difference observed between DDB and didemnin B is the methyl singlet peak at 2.04 ppm which could be assigned to a methyl ketone and the absence of the signal corresponding to the α-proton of the hydroxyl group in the lactyl moiety at 4.3 ppm. [0018]
  • Biological Activities [0019]
  • The compound of this invention has been shown to inhibit in vitro L1210 and P-388 mouse leukemia cells; L-929, mouse areolar and adipose tissue, B-16, mouse melanoma cells; A-549, human lung carcinoma cells; HeLa, human cervix epithelioid carcinoma cells and KB, human cervix epithelioid carcinoma cells and KB, human oral epidermoid carcinoma cells, and in vivo, P-338, mouse leukemia cells, Lewis lung carcinoma cells and B-16 melanoma cells. Thus, DDB is useful as an antitumor agent and therefore is useful inhibiting the growth of tumor cells in mammals exhibiting such tumor cells. [0020]
  • The following table summarizes the IC[0021] 50 values for each line cells in vitro:
    TABLE 1
    Cell Line IC50 (ng/mL)
    L-1210 0.3
    P-388 0.175
    L-929 1.9
    B-16 0.225
    A-549 0.5
    HeLa 0.5
    KB 5.6
  • The following table shows the % T/C in vivo after administration of DDB: [0022]
    TABLE 2
    Dehydrodidemnin B in vivo Activity
    Compound Control DDB DDB
    Dose (μg/kg/injn) 160 80
    Schedule and Route QD 1-9, IF QD 1-9, IP QD 1-9, IP
    P-388, Median 10.0 21.0 19.5
    Survival Time, Days 100 210 195
    P-388 % T/Ca
    Lewis Lung, Mean 1512 0 189
    Tumor Volume mm3 1.00 0.00 0.13
    Lewis Lung
    % T/Cb
    3-16 Melanoma 17.0 >27.0 >27.0
    Median Survival Time
    Days
    B-16 Melanoma 100 >158 >158
    % T/Ca
  • Dehydrodidemnin B, like didemnin B, (Montgomery et al., [0023] Transplantation, 40, 49-56 (1985)), is a powerful immunomodulator.
  • Dehydrodidemnin B has also shown activity against [0024] Herpes simplex virus, type 1, in CV-1 cells (monkey kidney cells); thus it is also useful as an antiviral agent. The IC50 determined was 60 ng/mL (e.g., 10 fold greater than for L-1210 cells) and 1 μg/mL, respectively.
  • The compound of present invention is preferably presented for administration to humans and animals in unit dosage form in pharmaceutically appropriate carriers containing the active ingredient in the appropriate quantity. [0025]
  • Illustratively, dosage levels of the administered active ingredient can be intravenous 0.05 to about 50 mg/Kg, intraperitoneal, subcutaneous and intramuscular 1 to 100 mg/Kg; oral 1 to 150 mg/Kg of animal (body) weight. [0026]
  • The administration of DDB is useful to inhibit the growth of cancer cells in animals or humans bearing a neoplastic disease, for example, acute myelocytic leukemia, acute lymphocytic leukemia, malignant melanoma, adenocarcinoma of the lung, small cell carcinoma of the lung, and the like. [0027]
  • The compound can be isolated from tunicates of the genus Aplidium, and more especially from the species [0028] Aplidium albicans. The species is found in the Iberian Mediterranean Coast as well as in the Balearic Islands. The species has been also found in Great Britain, English Channel as well as in the Africa Coast and Portugal. It seems to prefer detritic, coralligenic and sciafilae algae communities. They also can be found in more photophilic habitats.
  • Colonies of the tunicate are generally flat and lobed (2.5 cm diameter). It is jelly like, totally encrusted with sand which confers a sandy color to the colony. Zooides are of a [0029] whitish color 10 mm long; the oral siphon has 6 lobes, and the cloacal languet is trifid, which is a species characteristic. Generally there are 10-11 rows of stigmas. The stomach has 6 marked folds. Gonads are of the family type with one or several ovocites below the digestive track and numerous testicular follicles forming one or double row in the post abdomen. Larvae are incubated in the number of 1 to 9 in the atrial cavity; they have 3 cupping-glasses and several vesicular formations in the anterior part.
  • Thus in a typical procedure in accordance with the present invention, isolation method generally comprises alcoholic extraction of the homogenized tunicate and selective purification of the desired DDB. [0030]
  • As shown in FIG. 1, the tunicate was extracted with MeOH, filtered and dissolved in MeOH: Toluene 3:1 and partitioned with 10% NaNO[0031] 3. The aqueous layer was successively extracted with CH2Cl2, EtOAc and n-BuOH. The organic fractions were combined after monitoring by normal-phase TLC developed CHCl3:MeOH 9:1, affording a 2:1 (v/v) and the activity was concentrated in the methanolic layer. The polar fraction is passed through Silica gel Step-gradient Chromatography. The last fraction is further purified by reverse phase HPLC at a flow rate of 2 ml/min. Two mean peaks were collected and readily interconverted to a mixture of I and II, to establish an approximately 1:1 ratio.
  • The DDB can also be prepared by total synthesis, or semisynthesis from natural Didemnin A, following in both cases standard procedures of protection and activation in peptide chemistry. [0032]
  • Pyruvic acid+L-Pro→Side Chain
  • Side Chain+Didemnin A→Dihydrodidemnin B
  • Thus for example, Pro-OBzl, in DMF is mixed with pyruvic acid and HOBt, and DCC in CH[0033] 2Cl2 added. The reaction product can be purified and shows the chemical and physical properties corresponding to Pyruvyl-Pro-OBzl.
  • To a solution of this last product in CH[0034] 2Cl2, EDC and then Didemnin A was added. The evaporated residue is purified yielding DDB having chemical, physical, spectroscopical and biological characteristics in accord with natural Dehydrodidemnin B.
  • Apart from DDB itself, the present invention extends to derivatives of DDB, comprising acylated, alkylated or arylated derivatives of DDB, where R could be a group COR′ or R′, where R′ represents the following substituents: [0035]
  • CH[0036] 3, CH2R1, CH2R1, CHR1R2R3 or C6H5— where R1 R2, R3, could be alkyl (either linear or branched), aryl or alkylaryl, the aryl groups, bearing or not the substituents described under R′. The residues R1, R2, R3 could be either the same or different.
  • The derivatives can be more preferably alkyl, aryl or acyl-derivatives, where R′ is an aliphatic or aromatic group, more preferably a 1-6 carbon atom residue. [0037]
  • In general, such derivatives from DDB of this kind, are expected to show similar biological activity to that of DDB itself, including specifically antitumoral, antiviral, cytotoxic and immunosuppressive activity. [0038]
  • The acyl derivatives can be obtained by treatment of the parent compound with the corresponding carboxylic anhydride in the presence of pyridine or other nitrogenated organic base; by reaction of DDB with the respective acylchloride; or by dehydration with DCC from DDB and the corresponding carboxylic acid. [0039]
  • In the case of the alkyl or aryl derivatives (R/R′), they can be obtained by reaction of DDB with the corresponding halide, in the presence of an alkaline-organic weak base or by dehydration between DDB and alkyl or aryl hydroxy derivative by an organic dehydrating agent. [0040]
  • Instrumentation, Material and Methods [0041]
  • NMR spectra were obtained with a General Electric QE-300 (300 MHz, [0042] 1H), a Nicolet NT-360 (360 MHz, 1H) or a General Electric GN 500 (500 MHz, 1H) at the University of Illinois or a Varian Unity 300 (300 MHz, 1H and 75 MHz, 13C at PharmaMar, S. A. (Madrid, Spain) Chemical shifts are reported in ppm referenced to the chloroform peak at δ7.26 ppm for 1H. FABMS were measured on a VG Analytical ZAB at the Mass Spectrometry laboratory of the University of Illinois. GC analyses were carried out using a Varian GC (Model 3700) equipped with an Alltech Associates, Inc., Chirasil-Val II capillary column (25 m×0.32 mm) with Helium gas carrier at a flow rate of 1.2 ml/min with programmed oven temperature (90° C., 4° C./min, 180° C.). Reversed-phrase HPLC was performed on a system equipped with an Altex pump (Model 110 A) and a Waters Associates differential refractometer (Model R-401) and an Alltech Spherisorb C18 column (25 cm×1 cm, particle size 10 μm) with MeOH: H2O 0:1 as the solvent system.
  • The following examples illustrate the invention. [0043]
  • EXAMPLE 1
  • 1. Structure Determination [0044]
  • The structure of DDB has been determined by physical and spectroscopic methods. [0045]
  • 1.1 Spectroscopic Data [0046]
  • TLC R[0047] f=0.4; 0.35 (Silica gel, 2:3, CH2Cl2/EtOAc); 0,5; 0.44 (Silica gel; 9:1, CHCl3/MeOH);
  • RP-HPLC t[0048] r=10, 7; 11.9 min (Spherisorb C18 column, 250 mm×10 mm, 10 μm particle size, 9:1, MeOH/H2O; 2 mL/min);
  • [α][0049] D 25=86° (C 1, MeOH);
  • HR FABMS (M+H) C[0050] 57H88N7O15 m/z calcd. 1110.6382 (found 1110.6366); (M−side chain+H): C42H66N5O11 m/z calcd. 816.4781 (found 816.4755): (M−side chain): C15H23N2O4 m/z calcd. 295.1657 (found 295.1657);
  • IR (CHCl[0051] 3) vmax cm−1: 3680, 3600, 2970, 2940, 2880, 1740, 1650, 1605, 1540, 1510;
  • [0052] 1H NMR (CDCl3, δ, ppm): 7.82 (d, J=9 Hz, 1 H); 7.79 (d, J=9 Hz, 1H); 7.62 (d, J=6 Hz, 1H); 7.21 (d, J=9 Hz, 1H); 7.19 (d, J-9 Hz, 1H); 7.08 (d, J=8.5 H, 2H; 6.85 (d, J=8.5 Hz, 2H); 3.77 (s, 3H); 3.13 (s, 3H); 3.08 (s, 3H); 2.54 (s, 3H); 2.50 (s, 3H); 2.1 (s, 3H); 2.02 (s, 3H); 0.82-0.88 (overlapped d and t, 3OH);
  • [0053] 13C NMR (CDCl3, δ, ppm): 204.93 (s); 204.77 (s); 201.23 (s); 197.55 (s); 173.05 (s); 173.05 (s); 172.36 (s); 171.16 (s); 170.59 (s); 169.58 (s); 169.35 (s); 168.36 (s); 168.28 (s); 161.31 (s); 161.06 (s); 158.64 (s); 158.62 (s), 130.31 (d); 114.12 (d); 114.10 (d); 81.47 (d), 81.43 (d); 70.68 (d); 70.33 (d); 67.97 (d); 67.76 (d); 66.38 (d); 66.22 (d); 60.39 (t); 50.88 (d); 57.80 (d); 66.38 (d); 66.22 (d); 60.39 (t); 50.88 (d); 57.80 (d); 57.45 (d); 57.26 (d); 57.18 (d); 57.12 (d); 55.61 (d); 55.57 (d); 55.26 (q); 54.65 (d); 49.55 (d); 49.49 (d); 48.85 (t); 48.41 (t); 46.98 (t); 41.29 (t); 41.24 (t); 38.78 (q); 38.74 (q); 38.68 (q); 36.42 (t); 36.22 (t); 34.06 (d); 33.99 (d); 31.57 (d); 31.38 (q); 31.34 (q); 31.30 (q); 30.69 (d); 29.68 (t); 29.64 (d); 27.28 (t); 27.94 (t); 27.30 (t); 27.17 (t); 27.08 (t); 25.91 (t); 25.87 (t); 25.87 (t); 25.73 (d); 25.68 (d); 25.63 (d); 25.52 (d); 25.48 (d); 24.80 (q); 24.70 (q); 24.44 (q); 24.31 (q); 24.44 (q); 22.21 (q); 22.12 (q); 21.92 (q); 21.79 (q); 21.76 (q); 19.46 (q); 17.76 (q); 17.72 (q); 17.18 (q); 16.87 (q); 16.08 (q); 15.62 (q); 15.48 (q); 15.05 (q); 12.55 (q); 12.50 (q).
  • 1.2 Acetylation of DDB [0054]
  • The structure of dehydrodidemnin B can be confirmed also by comparison of the acetylation product with the acetyl derivative of didemnin B. [0055]
  • Acetylation of DDB with acetic anhydride and pyridine gave a monoacetyl derivative. [0056]
  • Low resolution mass spectrum showed peaks at ml z 1153.5 (M+H), 859.0 (M+2H−side chain) and 295.4 (side chain), indicating the loss of one of the two possible sites of acetylation with respect to didemnin B, and that the missing site is the hydroxyl group of the lactyl moiety in the side chain. [0057]
  • 1.3 N-Trifluoroacetyl methyl esters of amino acid Residues [0058]
  • The structure of DDB can also be determined by identification of the individual subunits by total hydrolysis and conversion of the amino acids to their N-trifluoroacetyl methyl esters and analysis by GC. [0059]
  • The amino acids were identified by their retention times and comparison of authentic samples obtained from the conversion of didemnin B to the N-trifluoroacetyl methyl esters of the amino acids. [0060]
  • t[0061] R (min): L-Threonine (1.23); D-N-Me-Leucine (1.70); L-Leucine (2.05); L-Proline (2.38); (3S,4R,5S)-Isostatine (3.15, 4.13, 4.77); L-N, O-Me2-Tyrosine (6.75).
  • A mixture of DDB and glass-distilled HCl was heated during 18 hours at 110° C. in a sealed Teflon-lined screw-capped vial. The solvent was removed under a stream of N[0062] 2 gas.
  • The hydrolysate was treated with MeOH/Acetyl chloride during 1 hour at 110° C. The solution was cooled to room temperature, the solvent was removed under a stream of N[0063] 2 gas. The solid was treated with a mixture of TFAA/TFA during 15 min at 100° C. The solution, was cooled and the solvent evaporated. The residue was dissolved in 2-propanol for GC analysis.
  • EXAMPLE 2
  • Biological Activity Assays [0064]
  • 2.1 Assay Against L-1210 Cells (Ascetic Fluid from DBA/2mouse) [0065]
  • L-1210 cells were seeded into 16 mm wells at 1×10[0066] 4 cells per well in 1 mL aliquots of MEN 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. Cells were counted after three drugs was counted daily to ensure that the cells remained in exponential growth over the period of observation.
    Growth Inhibition of L-1210 cells by DDB
    net increase in cell
    ng/mL DDB number % Inhibition
    0 2.9 × 105 0
    0.05 2.7 × 105 7
    0.1 2.7 × 105 7
    0.2 2.1 × 105 28
    0.5 1.0 × 105 66
    1 2.5 × 104 91
    2 6.3 × 103 98
  • 2.2 Assay Against P-388 Cells (Lymphoid Neoplasm from DBA/2mouse) [0067]
  • P-388 cells were seeded into 16 mm wells at 1×10[0068] 4 cells per well in 1 mL aliquots of MEM 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. Cells were counted after three days of incubation. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period of observation.
    Growth Inhibition of P-388 cells by DDB
    net increase in cell
    ng/mL DDB number % Inhibition
    0 5.63 × 105 0
    0.12 3.97 × 105 29
    0.25 1.27 × 105 77
    0.5 4.47 × 105 92
  • 2.3 Assay Against L-929 Cells (Mouse Areolar and Adipose Tissue) [0069]
  • L-929 cells were seeded into 16 mm wells at 1×10[0070] 4 cells per well in 1 mL aliquots of MEM 10C. The following day, medium was replaced with 0.5 mL aliquots of MEM 10C. The following day, the medium was replaced with 0.5 mL aliquots of MEM 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period of observation. Cells were trypsinized and counted 4 days after seeding.
    Growth Inhibition of L-929 cells by DDB
    net increase in cell
    ng/mL DDB number % Inhibition
    0 3.17 × 105 0
    1 2.31 × 105 27
    2.5 1.13 × 105 64
    5   5 × 105 84
  • 2.4 Assay Against B-16 Cells (Mouse Melanoma) [0071]
  • B-16 cells were seeded into 16 mm wells at 1×10[0072] 4 cells per well in 1 mL aliquots of MEM 10C determinations were carried out in triplicate. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period for observation. Cells were trypsinized and counted 4 days after seeding.
    Growth Inhibition of B-16 cells by DDB
    net increase in cell
    ng/mL DDB number % Inhibition
    0 1.71 × 105 0
    0.16 1.71 × 105 0
    0.12 1.27 × 105 25
    0.25 8.25 × 104 54
    0.5 4.50 × 104 74
    1.0 2.88 × 104 83
  • 2.5 Assay Against A-549 Cells (Human Lung Carcinoma) [0073]
  • A-549 cells were seeded into 16 mm wells at 1×10[0074] 4 cells per well in 1 mL aliquots of MEM 10C. The following day, the medium was replaced with 0.5 mL aliquots of MEM 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period of observation. Cells were trypsinated and counted 4 days after seeding.
    Growth Inhibition of A-549 cells by DDB
    net increase in cell
    ng/mL DDB number % Inhibition
    0 8.16 × 104 0
    0.25 4.80 × 104 41
    0.50 4.00 × 104 50
    1.0 2.50 × 104 68
    2.5 1.30 × 104 84
  • 2.6 Assay Against HeLa Cells (Human Cervix Epitheliod Carcinoma) [0075]
  • HeLa cells were seeded into 16 mm wells at 1×10[0076] 4 cells per well in 1 mL aliquots of MEM 10C. The following day, the medium was replaced with 0.5 mL aliquots of MEM 10C containing the indicated concentrations of drug. All determinations were carried out in triplicate. A separate set of cultures without drug was counted daily to ensure that the cells remained in exponential growth over the period of observation. Cells were trypsinized and counted 4 days after seeding.
    Growth Inhibition of KB cells by DDB
    net increase in cell
    ng/mL DDB number % Inhibition
    0 4.50 × 104 0
    2.5 4.57 × 104 0
    5 2.40 × 104 46
    10 1.02 × 104 77
  • 2.8 Assay Against HSV-1 (Herpes Simplex Virus Type-1) [0077]
  • 16 mm diameter wells were seeded each with 2×10[0078] 5 CV-1 cells in 1 mL aliquots of MEM 10C. Four days after, cells were infected with HSV-1 at 10C PFU per well. After adsorption for 1.5 hours, the inoculum was replaced in pairs of wells with 0.5 mL aliquots of MEM 5C containing the indicated concentrations of drug. Cells from two wells without drug were scraped into the medium and frozen 4 hours after infection to provide a baseline for calculating new virus production. The average of these samples was 2.5×105+1.2×106 PFU per mL. The remaining samples were collected 24 hours after infection.
    Inhibition of HSV-1 replication by DDB
    net virus produced
    ng/mL DDB (PFU/mL) % Inhibition
    0 4.5 × 108 0
    0.03 3.8 × 108 16
    0.1 1.5 × 108 67
    0.3 1.9 × 108 96
    1 0 100
  • 2.9 Immunosuppressive Activity [0079]
  • Dehydrodidemnin B is active as an immunosuppressive agent. In the mixed lymphocyte reaction it suppresses the immune reaction of murine cells. It also inhibits the growth of murine T-cells and B-cells. [0080]
  • EXAMPLE 3
  • Extraction and Isolation [0081]
  • A white solitary tunicate was collected near Ibiza in the Balearic Islands (Spain) and was identified by Dr. Xavier Turon of the Universitat de Barcelona, Barcelona (Spain) as [0082] Aplidium albicans. A sample is preserved at Centre d'Etudes Avancats, Blanes (Germona, Spain). Preliminary tests on shipboard indicated antiviral activity against VSV-1 (Vesicular stomatitis virus). Further studies in the laboratory confirmed the antiviral activity against Herpes simplex virus, type 1 (HSV-1) in monkey kidney cells (CV-1) and also showed cytotoxicity against mouse lymphoid leukemia in vitro (L1210 line cells).
  • The frozen tunicate was extracted with methanol. Solvent partitioning of the residue afforded three active fractions which were combined according to their similarity in TLC (Thin Layer Chromatography). The crude active fraction was portioned and the activity concentrated in the methanolic layer. The methanol layer was chromatographed by silica gel gravity column (chloroform and chloroform-methanol mixtures), affording one active fraction which was further purified by Reversed-Phase High-Performance Liquid Chromatography (RPC[0083] 18HPLC), affording two peaks (I and II). Analysis by TLC revealed two identical spots in each HPLC fraction. Re-injection of each individual fraction led to two peaks with the same retention times as I and II. Co-injection of I and II confirmed the presence of two identical peaks (possible conformers) in each fraction suggesting a rapid interconversion of I to II and vice versa.
  • EXAMPLE 4
  • Semisynthesis of DDB from Didemnin A [0084]
  • Dehydrodidemnin B can also be obtained and its structure confirmed by comparison with a semisynthetic sample prepared by coupling of the appropriate side chain to natural didemnin A. The data obtained for the semisynthetic sample totally agreed with data for natural DDB. [0085]
  • 4.1 Synthesis of Pyruvyl-Pro-)Bzl [0086]
  • The hydrochloride salt of Pro-OBzl (10.2 g, 42 mmol) was dissolved in dry DMF (30 ml), neutralized with NMM (N-methylmorpholine, 4.7 mL, 42 mmol) at 0° C., and the solution was mixed with pyruvic acid (8.8 g, 100 mmol) and HOBt (1-hydroxybenzotriazole, 16.8 g, 110 mmol) in CH[0087] 2Cl2-DMF (90 mL, 8:1). DCC (dicyclohexylcarbodiimide, 22.6 g, 110 mmol) in CH2Cl2 (35 mL) was added to the above mixture at 0° C. with stirring. The reaction mixture was stirred for 2 hours at 0° C. and left overnight at room temperature. DCCl was filtered off and washed with CH2Cl2 (20 mL). The filtrate was evaporated to dryness, the residue taken up in EtOAc and washed successively with 5% citric acid, water, 5% NaHCO3 and finally with water to neutral pH. The organic layer was dried (Na2SO) and concentrated. The residue was chromatographed on SiO2 with hexane-EtOAc (2:1) to give the title compound (11 g, 95%).
  • [α][0088] D 25=−78.57 (c 0.14, CHCl3);
  • R[0089] f+0.63 (19:1, CHCl3/MeOH);
  • Anal. Calcd. for C[0090] 15H18NO4 (M+H): 276.1235;
  • Found: 276.1235 (M+H, HRFABMS). [0091]
  • 4.2 Synthesis of Pyruvyl-Proline [0092]
  • The protected dipeptide from the previous synthesis (11.0 g, 40 mmol) was dissolved in EtOAc (75 mL) and stirred under hydrogen over Pd/C for 2 h. The catalyst was then filtered off and the filtrate was evaporated to dryness. The residue was crystallized from EtoAc-hexane to give the unprotected peptide (6.9 g, 93): [0093]
  • [α][0094] D 25=−103.99 (c 0.124, CHCl3);
  • R[0095] f=0.4163 (19:1:0.5, CHCl3/MeOH/AcOH;
  • Anal. Calcd. for C[0096] 8H12NO4 (M+H): 186.0766;
  • Found: 186.0765 (M+H, HRFABMS). [0097]
  • 4.3 Synthesis of Dehydrodidemnin B [0098]
  • EDC (1-ethyl-3-(3-dimethylaminopropyl)carbodiimide, 4.27 g, 22.3 mmol) was added to a solution of Pyrvu-Pro (8.2 g, 44.5 mmol) in dry CH[0099] 2Cl2 (40 mL) at 10° C. with stirring. The mixture was stirred for 2 h at 10° C. and then cooled to 0° C. Didemnin A (1.4 g, 1.48 mmol) in CH2Cl2-DMF (10 mL, 4:1) was added, and the clear solution was stirred at 0° C. for 2 h and then left in the refrigerator overnight.
  • DMAP (4-dimethylaminopyridine, 25 mg) was added to the reaction mixture, and it was again left in the refrigerator for 48 h. The solvent was evaporated to dryness, and the residue was taken up in EtOAc and washed with 5% NaHCO[0100] 3 and water to neutral pH. The organic layer was dried (Na2SO4) and concentrated. The residue so obtained was chromatographed on silica gel using CHCl3-MeOH (19:1) to give dehydrodidemnin B (1.4 g, 84%, 2 spots on TLC):
  • [α][0101] D 25=−95.384 (c 0.06, MeOH)3);
  • R[0102] f=0.51 and 0.44 (19:1, CHCl3/MeOH);
  • Anal. Calcd. for C[0103] 57H88N7O15 (M+H): 1110.6338;
  • Found: 1110.6355 (M+H, HRFABMS). [0104]
  • The same series of reactions can be carried out with slight modifications, in particular EDC can be replaced by DDC with slightly lower yield. [0105]
  • The present invention has been described in detail, including the preferred embodiments thereof. However, it will be appreciated that those skilled in the art, upon consideration of the present disclosure, may make modifications and/or improvements on this invention and still be within the scope and spirit of this invention as set forth in the following claims. [0106]

Claims (6)

What is claimed is:
1. The substantially pure compound, Dehydrodidemnin B, essentially free of the cellular debris of the Mediterranean tunicate, Aplidium albicans, said compound having the formula:
Figure US20030148933A1-20030807-C00003
2. An acylated, alkylated or arylated derivative of dehydrodidemnin B wherein the acyl, alkyl, or aryl groups are selected from C1 to C6 alkyl, C1 to C6 acyl, phenyl, benzyl, or C1 to C6 alkylphenyl.
3. A pharmaceutical composition which comprises dehydrodidemnin B or an active acylated, alkylated or arylated derivative thereof, together with a pharmaceutically acceptable carrier, wherein the acyl, alkyl, or aryl groups are selected from either C1 to C6 alkyl, C1 to C6 acyl, phenyl, benzyl, or C1 to C6 alkylphenyl.
4. A semisynthetic process for the preparation of dehydrodidemnin B, which comprises the coupling of pyruvyl-proline to didemnin A.
5. A method for isolation of dehydrodidemnin B which comprises extraction from a dehydrodidemnin B-containing tunicate.
6. The monoacetyl derivative of dehydrodidemnin B.
US10/357,759 1989-09-29 2003-02-04 Derivatives of dehydrodidemnin B Abandoned US20030148933A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/357,759 US20030148933A1 (en) 1990-10-01 2003-02-04 Derivatives of dehydrodidemnin B
US11/136,622 US7348311B2 (en) 1989-09-29 2005-05-23 Derivatives of dehydrodidemnin B

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
WOPCT/GB90/01495 1990-10-01
PCT/GB1990/001495 WO1991004985A1 (en) 1989-09-29 1990-10-01 Dehydrodidemnin b
US84456792A 1992-04-24 1992-04-24
US08/280,110 US5834586A (en) 1989-09-29 1994-07-25 Dehydrodidemnin B
US18268898A 1998-10-30 1998-10-30
US09/619,354 US6710029B1 (en) 1989-09-29 2000-07-19 Dehydrodidemnin B
US10/357,759 US20030148933A1 (en) 1990-10-01 2003-02-04 Derivatives of dehydrodidemnin B

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/619,354 Continuation US6710029B1 (en) 1989-09-29 2000-07-19 Dehydrodidemnin B

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/136,622 Division US7348311B2 (en) 1989-09-29 2005-05-23 Derivatives of dehydrodidemnin B

Publications (1)

Publication Number Publication Date
US20030148933A1 true US20030148933A1 (en) 2003-08-07

Family

ID=27671034

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/357,759 Abandoned US20030148933A1 (en) 1989-09-29 2003-02-04 Derivatives of dehydrodidemnin B
US11/136,622 Expired - Fee Related US7348311B2 (en) 1989-09-29 2005-05-23 Derivatives of dehydrodidemnin B

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/136,622 Expired - Fee Related US7348311B2 (en) 1989-09-29 2005-05-23 Derivatives of dehydrodidemnin B

Country Status (1)

Country Link
US (2) US20030148933A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023274145A1 (en) * 2021-06-28 2023-01-05 浙江珲达生物科技有限公司 Method for preparing dehydrodidemnin b compound

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE363910T1 (en) * 1999-11-15 2007-06-15 Pharma Mar Sa TREATMENT OF CANCER DISEASES BY APLIDINE
KR20060002778A (en) * 2003-03-12 2006-01-09 파르마 마르, 에스.에이. Improved antitumoral treatments
ES2575518T3 (en) * 2006-02-28 2016-06-29 Pharma Mar S.A. Improved treatment of multiple myeloma
AU2008313627A1 (en) * 2007-10-19 2009-04-23 Pharma Mar, S.A. Improved antitumoral treatments
CN101965191A (en) * 2008-03-07 2011-02-02 法马马有限公司 Improved anticancer treatments
JOP20190254A1 (en) 2017-04-27 2019-10-27 Pharma Mar Sa Antitumoral compounds

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4493796A (en) * 1980-09-12 1985-01-15 Board Of Trustees, Univ. Of Ill. Didemnins A, B, C, and derivatives thereof, as antiviral agents
US4670262A (en) * 1982-09-23 1987-06-02 Farmitalia Carlo Erba S.P.A. Novel pharmacological compositions based on Cisplatinum and method for obtaining same
US4952399A (en) * 1985-09-20 1990-08-28 Cernitin Sa Pharmaceutical composition which inhibits the growth of a tumor
US5294603A (en) * 1980-09-12 1994-03-15 The Board Of Trustees Of The University Of Illinois Pharmaceutical compositions containing didemnins
US5294608A (en) * 1991-12-12 1994-03-15 Hoechst Aktiengesellschaft Guanidinoalkyl-1,1-bisphosphonic acid derivatives, process for their preparation and their use
US5462726A (en) * 1993-12-17 1995-10-31 Bristol-Myers Squibb Company Method of inhibiting side effects of solvents containing ricinoleic acid or castor oil or derivatives thereof employing a thromboxane A2 receptor antagonist and pharmaceutical compositions containing such solvents
US5834586A (en) * 1989-09-29 1998-11-10 Pharma Mar, S.A. Dehydrodidemnin B
US6034058A (en) * 1997-04-15 2000-03-07 Rinehart; Kenneth L. Semi-synthetic alanyl dilemnin analogs
US6156724A (en) * 1996-06-07 2000-12-05 Rinehart; Kenneth L. Uses of didemnins as immunomodulating agents

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0048149B1 (en) 1980-09-12 1983-11-30 University of Illinois Foundation Novel antibiotics, derivatives thereof, processes for their extraction, and compositions containing them
US4948791A (en) 1989-04-10 1990-08-14 The Board Of Trustees Of The University Of Illinois Novel Cytotoxic cyclic depsipeptides from the tunicate trididemnum solidum
DE4120327A1 (en) 1991-06-20 1992-12-24 Basf Ag NEW PEPTIDES, THEIR PREPARATION AND USE
ES2102322B1 (en) 1995-07-13 1998-03-01 Pharma Mar Sa DIDEMNINE PREPARATION PROCEDURE A.
JP2001503746A (en) 1996-10-24 2001-03-21 ザ・ボード・オブ・トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・イリノイ Semisynthetic method for didemnin analogues
DE69723728T2 (en) 1996-10-24 2004-06-03 The Board Of Trustees For The University Of Illinois, Urbana TOTAL SYNTHESIS OF THE AMINO HIP ANALOG OF DIDEMNIN A
GB9803448D0 (en) 1998-02-18 1998-04-15 Pharma Mar Sa Pharmaceutical formulation
ATE363910T1 (en) 1999-11-15 2007-06-15 Pharma Mar Sa TREATMENT OF CANCER DISEASES BY APLIDINE
WO2001076616A1 (en) 2000-04-07 2001-10-18 The Trustees Of The University Of Pennsylvania Tamandarin and didemnin analogs and methods of making and using them
UA76718C2 (en) 2000-06-30 2006-09-15 Фарма Мар, С.А. Anticancer aplidine derivatives
AU9402401A (en) 2000-10-12 2002-04-22 Pharma Mar Sa Treatment of cancers

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4493796A (en) * 1980-09-12 1985-01-15 Board Of Trustees, Univ. Of Ill. Didemnins A, B, C, and derivatives thereof, as antiviral agents
US5294603A (en) * 1980-09-12 1994-03-15 The Board Of Trustees Of The University Of Illinois Pharmaceutical compositions containing didemnins
US4670262A (en) * 1982-09-23 1987-06-02 Farmitalia Carlo Erba S.P.A. Novel pharmacological compositions based on Cisplatinum and method for obtaining same
US4952399A (en) * 1985-09-20 1990-08-28 Cernitin Sa Pharmaceutical composition which inhibits the growth of a tumor
US5834586A (en) * 1989-09-29 1998-11-10 Pharma Mar, S.A. Dehydrodidemnin B
US6153731A (en) * 1989-09-29 2000-11-28 Pharma Mar, S.A. Dehydrodidemnin B
US6710029B1 (en) * 1989-09-29 2004-03-23 Pharma Mar S.A. Dehydrodidemnin B
US5294608A (en) * 1991-12-12 1994-03-15 Hoechst Aktiengesellschaft Guanidinoalkyl-1,1-bisphosphonic acid derivatives, process for their preparation and their use
US5462726A (en) * 1993-12-17 1995-10-31 Bristol-Myers Squibb Company Method of inhibiting side effects of solvents containing ricinoleic acid or castor oil or derivatives thereof employing a thromboxane A2 receptor antagonist and pharmaceutical compositions containing such solvents
US6156724A (en) * 1996-06-07 2000-12-05 Rinehart; Kenneth L. Uses of didemnins as immunomodulating agents
US6034058A (en) * 1997-04-15 2000-03-07 Rinehart; Kenneth L. Semi-synthetic alanyl dilemnin analogs

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023274145A1 (en) * 2021-06-28 2023-01-05 浙江珲达生物科技有限公司 Method for preparing dehydrodidemnin b compound
CN115594739A (en) * 2021-06-28 2023-01-13 浙江珲达生物科技有限公司(Cn) Preparation method of dehydrodidemnin B compound

Also Published As

Publication number Publication date
US7348311B2 (en) 2008-03-25
US20050222015A1 (en) 2005-10-06

Similar Documents

Publication Publication Date Title
USRE39887E1 (en) Dehydrodidemnin B
RU2060998C1 (en) Method of synthesis of peptides, peptides, immunomodulating composition and a method of regulation of insufficient or excessive function of t-cells in patient
CA1270596A (en) Peptide derivatives having a nitrogenous polycyclic structure and process for their preparation
US7348311B2 (en) Derivatives of dehydrodidemnin B
DD213917A5 (en) METHOD FOR PRODUCING NEW PEPTIDE DERIVATIVES
JP2002513402A (en) Isolation and structure determination of the cytostatic linear and cyclic depsipeptides dolastatin 16, dolastatin 17, and dolastatin 18
EP0011283B1 (en) New lactyl tetrapeptide, processes for preparation thereof and pharmaceutical compositions containing it
US6156724A (en) Uses of didemnins as immunomodulating agents
JPH0123479B2 (en)
US4965250A (en) Peptide compounds having a nitrogenous polycyclic structure
US4442031A (en) Immunopotentiating peptides
CA1242703A (en) Tuftsinyl-tuftsin
EP0880541A1 (en) Derivatives of antibiotic ge2270 factors
IE42369B1 (en) Peptide derivatives having an antihypertensive effect and process for their manufacture
US4851446A (en) Immunosuppressing method
HU193528B (en) Process for preparing a-219780 cyclic peptide-acil derivatives
US4487764A (en) New peptides and a process for their preparation
EP0337731A2 (en) Peptide antibiotics
EP0452447A1 (en) Reduced irreversible bombesin antagonists
EP0894092B1 (en) A cyclic hepta-peptide derivative from colonial ascidians, lissoclinum sp.
GB2227244A (en) Immunosuppressive fluorinated cyclosporin analogs
JPH0672152B2 (en) Thymosin α1-fragment and immunomodulator containing the compound
EP0050856B1 (en) New peptide, process for its preparation and pharmaceutical composition containing it
CA2128552A1 (en) Derivatives of 6,7-dihydroxy-4-thiaheptanoic acid and their use
US5656601A (en) Acylated splenopentins, methods for their synthesis and their use

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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