In Vivo Antitumor Activity of Bis(4,7-dimethyl-1,10-phenanthroline) Sulfatooxovanadium(IV) METVAN VO(SO4)(Me2-Phen)2

Page created by Kimberly Roberts
 
CONTINUE READING
In Vivo Antitumor Activity of Bis(4,7-dimethyl-1,10-phenanthroline) Sulfatooxovanadium(IV) METVAN VO(SO4)(Me2-Phen)2
2124 Vol. 7, 2124 –2133, July 2001                                                                                      Clinical Cancer Research

     In Vivo Antitumor Activity of Bis(4,7-dimethyl-1,10-phenanthroline)
     Sulfatooxovanadium(IV) {METVAN [VO(SO4)(Me2-Phen)2]}

     Rama Krishna Narla, Chun-Lin Chen,                                      after tumor cell inoculation than those of the vehicle-treated
     Yanhong Dong, and Fatih M. Uckun1                                       control SCID mice (174 ⴞ 29 mm3 versus 487 ⴞ 82 mm3;
                                                                             P ⴝ 0.002). The favorable in vivo pharmacodynamic fea-
     Parker Hughes Cancer Center, Departments of Oncology [R. K. N.,
     F. M. U.], Pharmaceutical Sciences [C-L. C.], and Chemistry [Y. D.],    tures and antitumor activity of METVAN warrants further
     and Drug Discovery Program [R. K. N., C-L. C., Y. D., F. M. U.],        development of this novel oxovanadium compound as a
     Parker Hughes Institute, St. Paul, Minnesota 55113                      potential new anticancer agent.

                                                                             INTRODUCTION
     ABSTRACT
                                                                                   The use of vanadium compounds and vanadium salts for
          The compound bis(4,7-dimethyl-1,10-phenanthroline)
                                                                             clinical applications received renewed interest in the late 1970s
     sulfatooxovanadium(IV) {METVAN [VO(SO 4 )(Me 2 -
                                                                             and early 1980s because of the discovery that vanadate(V)
     Phen)2]}, exhibits potent cytotoxicity against human cancer
                                                                             solutions produced insulin-like effects in rat diaphragms and
     cells at low micromolar concentrations. At concentrations
                                                                             isolated adipocytes in vitro (1–3). Subsequently, the adminis-
     >1 ␮M, METVAN treatment was associated with a nearly
                                                                             tration of vanadate solutions to diabetic rats was shown to lower
     complete loss of the adhesive, migratory, and invasive prop-
                                                                             blood glucose levels (4). Despite a long history of vanadium
     erties of the treated tumor cell populations. METVAN did
                                                                             compounds being used in the treatment of human diseases,
     not cause acute or subacute toxicity in mice at dose levels
                                                                             however, only a small number of organometallic compounds
     ranging from 12.5 mg/kg to 100 mg/kg. Therapeutic plasma
                                                                             containing vanadium have been tested for antitumor activity
     concentrations >5 ␮M were rapidly achieved and main-
                                                                             (5–11); a few of these were shown to possess anticancer activity
     tained in mice for at least 24 h after i.p. bolus injection of a
                                                                             in vitro as well as in vivo (6, 8, 12–14).
     single 10 mg/kg nontoxic dose of METVAN. At this dose
                                                                                   Vanadium can be found in both anionic and cationic forms
     level, the maximum plasma METVAN concentration was
                                                                             with oxidation states ranging from ⫺1 to ⫹5 (15–18). Vana-
     37.0 ␮M, which was achieved with a tmax of 21.4 min. Plasma
                                                                             dium complexes with oxidation states ⫹4 (IV) and ⫹5 (V) have
     samples (diluted 1:16) from METVAN-treated mice killed
                                                                             been shown to modulate cellular redox potential, regulate enzy-
     85% of human breast cancer cells in vitro. METVAN was
                                                                             matic phosphorylation, and exert various other effects in mul-
     slowly eliminated with an apparent plasma t1/2 of 17.5 h and
                                                                             tiple biological systems (19 –25). In addition to the ability of
     systemic clearance of 42.1 ml/h/kg. In accordance with its
                                                                             vanadium to assume various oxidation states, its coordination
     potent in vitro activity and favorable in vivo pharmacokinet-
                                                                             chemistry also plays a key role in its interactions with various
     ics, METVAN exhibited significant antitumor activity and
                                                                             biomolecules. As part of a systematic effort aimed at developing
     delayed tumor progression in CB.17 severe combined im-
                                                                             compounds with anticancer activity, we recently identified
     munodeficient (SCID) mouse xenograft models of human
                                                                             METVAN, {bis(4,7-dimethyl-1,10-phenanthroline) sulfatooxo-
     glioblastoma and breast cancer. In these experiments,
                                                                             vanadium(IV) [VO(SO4)(Me2-Phen)2]} as an active apoptosis-
     METVAN was administered in daily injections of a single
                                                                             inducing agent with potent in vitro antitumor activity (26 –28).
     nontoxic 10 mg/kg i.p. dose on 5 consecutive days per week
                                                                             At nanomolar to low micromolar concentrations, METVAN
     for 4 consecutive weeks beginning the day after the s.c.
                                                                             induced apoptosis in leukemic cell lines, multiple myeloma cell
     inoculation of U87 glioblastoma or MDA-MB-231 breast
                                                                             lines, and solid tumor cell lines derived from breast cancer,
     cancer cells. At 40 days after the inoculation of tumor cells,
                                                                             glioblastoma, and testicular cancer patients (26 –28). Apoptosis
     the U87 tumor xenografts in the vehicle-treated control
                                                                             induced by treatment with METVAN is mediated by the gen-
     SCID mice were much larger than those of the mice treated
                                                                             eration of reactive oxygen species, depletion of glutathione, and
     with METVAN (4560 ⴞ 654 mm3 versus 1688 ⴞ 571 mm3;
                                                                             depolarization of mitochondrial membranes (29). Furthermore,
     P ⴝ 0.003). Similarly, the MDA-MB-231 tumors in SCID
                                                                             METVAN inhibited the constitutive expression of MMP2-9
     mice treated with METVAN were much smaller 40 days
                                                                             protein and its gelatinolytic activity in HL-60 acute myeloid
                                                                             leukemia cells and MMP-2 as well as of MMP-9 gelatinolytic
                                                                             activities in leukemic cells from acute lymphoblastic leukemia,

     Received 2/14/01; revised 3/28/01; accepted 3/30/01.
     The costs of publication of this article were defrayed in part by the
                                                                             2
     payment of page charges. This article must therefore be hereby marked     The abbreviations used are: MMP, matrix metalloproteinase; SCID,
     advertisement in accordance with 18 U.S.C. Section 1734 solely to       severe combined immunodeficient; IR, infrared; LC, liquid chromatog-
     indicate this fact.                                                     raphy; MS, mass spectrometry; ES, electron spin; MTT, 3-(4,5-dimeth-
     1
       To whom requests for reprints should be addressed, at Parker Hughes   ylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide; ECM, extracellular
     Institute, 2665 Long Lake Road, Suite 330, St. Paul, MN 55113. Phone:   matrix; AUC, area under the concentration-time curve; TdT, terminal
     (651) 697-9228; FAX: (651) 697-1042; E-mail: fatih_uckun@               deoxynucleotidyltransferase; TUNEL, TdT-mediated dUTP nick-end
     mercury.ih.org.                                                         labeling.

         Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                         Research.
In Vivo Antitumor Activity of Bis(4,7-dimethyl-1,10-phenanthroline) Sulfatooxovanadium(IV) METVAN VO(SO4)(Me2-Phen)2
Clinical Cancer Research 2125

acute myeloid leukemia, and chronic myeloid leukemia patients
(30). We now report that concentrations of METVAN, highly
cytotoxic to human glioblastoma and breast cancer cells in vitro,
can be achieved in vivo after i.p. administration of a single
nontoxic dose. METVAN showed favorable pharmacokinetics
in mice and exhibited significant in vivo antitumor activity in
SCID mouse xenograft models of human glioblastoma and
breast cancer.

MATERIALS AND METHODS
      Preparation and Characterization of METVAN. The
coordination compound METVAN, bis(4,7-dimethyl-1,10-phen-
anthroline) sulfatooxovanadium(IV) [VO(SO4)(Me2-Phen)2],
was synthesized as described previously in detail (26, 27).
METVAN was characterized by IR, LC-MS, and elemental
analysis. The results were as follows: (a) IR (KBr pellet),
IR (KBr pellet): ␯ (cm⫺1) 3421, 3066, 2924, 1624, 1608, 1578,
1524, 1423, 1383, 1297, 1237, 1157, 1030, 972, 930, 868, 729,
696, 663, 650, 590, 557, 545, 529, 485, 464; (b) LC-MS (ES),
LC conditions: eluant, 30:70 v/v mixture of acetonitrile and an
aqueous solution of 0.1% acetic acid; flow rate, 0.5 ml/min;
column, 250 ⫻ 4 mm LiChrospher 100 RP-18 (5 ␮m); injection
volume, 10 ␮l; detection, 300 nm; and LC-MS (ES), MS con-
ditions (positive ion mode): fragmentor voltage, 50 V; drying
gas flow rate, 10 liter/min; drying gas temperature, 350°C;
nebulizer pressure, 25 p.s.i.; retention time, 7.84 min; ES-MS,
Calculated for {M ⫹ H}⫹ 580.1 {M ⫽ [VO(SO4)(Me2-
Phen)2]}, C28H24N4O5SV}; found, 580.1; and (c) elemental            Fig. 1 Cytotoxic activity of METVAN against human glioblastoma
                                                                    and breast cancer cells. Human glioblastoma (A, U87, U118, U138,
analysis:    calculated    for     [VO(SO4)(Me2-Phen)2]䡠2H2O        U373, and T98) and breast cancer (B, BT-20, MDA-MB-231, MDA-MB-
(C28H28N4O7SV), C, 54.63; H, 4.59; and N, 9.10; found: C,           361, and MCF-7) cells were incubated with increasing concentrations
54.79; H, 4.50; N, 9.26.                                            (0.1–250 ␮M) of METVAN for 48 h (A and B) or 16 h (C) in 96-well
      Cell Lines. Human brain tumor cell lines U87 MG, U118         plates. Cell survival was determined by MTT assays. Data points, the
                                                                    mean (⫾ SE) values from three independent experiments. The mean (⫾
MG, U138, U373 MG, and T98G, and breast cancer cell lines
                                                                    SE) IC50 values for 48-h incubation were 2.1 ⫾ 0.6 ␮M (U87), 0.82 ⫾
BT-20, MDA-MB-231, MDA-MB-361, and MCF-7, were ob-                  0.1 ␮M (U118), 2.7 ⫾ 0.4 ␮M (U138), 2.0 ⫾ 0.4 ␮M (U373), 3.9 ⫾ 0.5
tained from American Type Culture Collection (Rockville, MD)        ␮M (T98), 1.6 ⫾ 0.7 ␮M (BT-20), 0.5 ⫾ 0.1 ␮M (MDA-MB-231), 1.9 ⫾
and were maintained as continuous cell lines in DMEM (U87,          0.5 ␮M (MDA-MB-361), and 2.3 ⫾ 0.4 ␮M (MCF-7). The mean (⫾ SE)
U118, U138, U373, T98, BT-20) or in Leibovitz’s L-15 medium         IC50 values for 16 h incubation were 16.3 ⫾ 1.8 ␮M (U87) and 11.1 ⫾
                                                                    1.8 ␮M (MDA-MB-231).
(MDA-MB-231 and MDA-MB-361). All of the media were
supplemented with 10% FCS, 4 mM glutamine, 100 units/ml
penicillin G, and 100 mg/ml streptomycin sulfate. All of the
tissue culture reagents were obtained from Life Technologies,       reader (Labsystems). The percentage survival and the IC50
Inc. Inc. (Gaithersburg, MD).                                       values were calculated using Graphpad Prism v2.0 (Graphpad
      Cytotoxicity MTT Assays. The cytotoxicity of                  Software, Inc., San Diego, CA).
METVAN against human cancer cell lines was analyzed                       Adhesion Assays. In vitro adhesion assays were used to
using the MTT assay (Boehringer Mannheim Corp., Indian-             evaluate the effects of METVAN on the adhesive properties of
apolis, IN) as described previously (27). Briefly, exponen-         U87 and MDA-MB-231 cells as described previously (31). The
tially growing tumor cells were seeded into a 96-well plate at      plates for the adhesion assays were precoated with the ECM
a density of 2.5 ⫻ 104 cells/well. METVAN was dissolved in          proteins laminin, fibronectin, vitronectin, and type IV collagen
0.1% DMSO in PBS and then added to each well to yield               (each at a final concentration of 1 ␮g/ml in PBS) overnight at
final concentrations ranging from 0.1 to 250 ␮M. After incu-        4°C and dried. Exponentially-growing cells were incubated with
bation at 37°C for 48 or 16 h, 10 ␮l of MTT (0.5 mg/ml final        METVAN at concentrations ranging from 0.5 ␮M to 25 ␮M for
concentration) were added to each well. The plates were then        16 h in a humidified 5% CO2 atmosphere. The cells were then
incubated at 37°C for an additional 4 h to allow MTT to form        detached from the flasks with 0.05% trypsin (Life Technologies,
formazan crystals by reacting with metabolically active cells.      Inc.), resuspended in medium, incubated at 37°C for 2 h to allow
The formazan crystals were solubilized overnight at 37°C in         them to recover from the trypsinization stress, and examined for
a solution containing 10% SDS in 0.01 M HCl. The absorb-            their ability to adhere to the plates precoated with ECM proteins.
ance at 540 nm (a reference wavelength of 690 nm was used)          Cells were centrifuged, washed twice with serum-free medium,
of the solution in each well was measured using a microplate        counted, and resuspended in serum-free medium at a final

    Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                    Research.
In Vivo Antitumor Activity of Bis(4,7-dimethyl-1,10-phenanthroline) Sulfatooxovanadium(IV) METVAN VO(SO4)(Me2-Phen)2
2126 In Vivo Antitumor Activity of METVAN

                                                                                                        Fig. 2 METVAN induces apoptosis
                                                                                                        in glioblastoma and breast cancer
                                                                                                        cells. U87 glioblastoma (A, A⬘) and
                                                                                                        MDA-MB-231 breast cancer (B, B⬘)
                                                                                                        cells were incubated with 10 ␮M of
                                                                                                        METVAN for 24 h, fixed, permeabi-
                                                                                                        lized and visualized for DNA degra-
                                                                                                        dation in a TUNEL assay using
                                                                                                        dUTP-labeling. Red fluorescence,
                                                                                                        nuclei stained with propidium iodide.
                                                                                                        Green or yellow (i.e., superimposed
                                                                                                        red and green) fluorescence, apop-
                                                                                                        totic nuclei containing fragmented
                                                                                                        DNA. When compared with controls,
                                                                                                        treated with 0.1% DMSO (A, B), sev-
                                                                                                        eral of the cells incubated with
                                                                                                        METVAN (A⬘, B⬘) exhibited apo-
                                                                                                        ptotic nuclei.

    concentration of 2.5 ⫻ 105 cells/ml. The cell suspension was         per filter were counted to determine the invasive fraction. The
    added to each well in aliquots of 100 ␮l, and the cells were         invasive fractions of cells treated with METVAN were com-
    allowed to adhere for 1 h at 37°C in a humidified 5% CO2             pared with those of DMSO-treated control cells and the percent-
    atmosphere. The nonadherent cells were removed by gently             age inhibition of invasiveness was determined.
    washing the cells with PBS. The adherent fraction was quanti-              Migration Assay. Migration of brain tumor cells was
    tated using MTT assays as described previously (27, 32).             monitored using U373 glioblastoma cell spheroids, as described
          In Vitro Invasion Assays. The in vitro invasiveness of         previously (31). Glioblastoma cell spheroids were cultured in
    cancer cells treated with METVAN was assayed using Matrigel-         100-mm2 tissue culture plates precoated with 0.75% agar pre-
    coated Costar 24-well transwell cell culture chambers (Boyden        pared in MEM supplemented with 10% fetal bovine serum. The
    chambers) with 8.0-␮m-pore polycarbonate filter inserts as de-       cells (5 ⫻ 106 cells) were suspended in the medium, seeded onto
    scribed previously (31, 33, 34). Exponentially-growing U87 and       agar-coated plates, and cultured for 5–7 days at 37°C. Spheroids
    MDA-MB-231 cells were incubated with METVAN at various               with a diameter of 200 – 400 ␮m were selected for use in
    concentrations ranging from 0.5 ␮M to 25 ␮M overnight. The           additional experiments. For the migration experiments, the se-
    cells were trypsinized, washed twice with serum-free medium          lected spheroids were incubated for 2 h at 37°C in serum-free
    containing BSA, counted and resuspended in a serum-free me-          medium containing METVAN in concentrations ranging from 1
    dium at 1 ⫻ 105 cells/ml. The cell suspension (0.5 ml aliquots)      ␮M to 25 ␮M. The METVAN-treated and vehicle-control
    was added to the Matrigel-coated and rehydrated filter inserts.      (DMSO, 0.1%) spheroids were transferred onto fibronectin-
    The inserts were then placed in 24-well plates containing 750 ␮l     coated coverslips (Becton Dickinson, Bedford, MA) and placed
    of NIH fibroblast-conditioned medium (31, 34) as a chemoat-          in 6-well plates containing the same concentrations of
    tractant and were incubated at 37°C for 48 h. The filter inserts     METVAN or DMSO in serum-free medium. Spheroids were
    were then removed, the medium was decanted, and the cells on         then kept in a humidified 5% CO2 incubator at 37°C for 48 h. A
    the top of the filter that did not migrate were scraped off with a   total of four to six spheroids were used for each concentration.
    cotton-tipped applicator. The invasive cells that migrated to the    After the 48-h incubation period, the spheroids were fixed and
    lower side of the filter were fixed, stained with Hema-3 solution,   stained with Hema-3 solutions and mounted onto glass slides.
    and counted under a light microscope. Five to 10 random fields       The distance of tumor cell migration from the spheroid was

         Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                         Research.
Clinical Cancer Research 2127

measured using an ocular micrometer and a transmitted light
microscope.
      Mice. The animal protocols used in this study were ap-
proved by the Parker Hughes Institute Institutional Animal Care
and Use Committee (IACUC), and all of the animal care pro-
cedures conformed to the Guide for the Care and Use of Lab-
oratory Animals (National Research Council, National Acad-
emy Press, Washington DC 1996). Female CB.17 SCID and
CD-1 mice were obtained from Taconic (Germantown, NY) and
housed in a specific-pathogen-free room located in a secure
indoor facility with controlled temperature, humidity, and noise
levels. Mice were housed in microisolater cages and fed with
autoclaved rodent chow. Water was also autoclaved and sup-
plemented with trimethoprim/sulfomethoxazol 3 days a week.
      Toxicity Studies in Mice. The toxicity profile of
METVAN in CD-1 mice was examined, as reported previously
for other new agents (35–38). Female CD-1 mice (7 weeks old;
average weight, 24.5 g) were used and monitored daily for
lethargy, cleanliness, and morbidity. At the time of death, nec-
ropsies were performed, and the toxic effects of METVAN
administration were assessed. For histopathological studies, tis-
sues were fixed in 10% neutral buffered formalin, dehydrated,
and embedded in paraffin by routine methods. Glass slides with
affixed 6-␮m tissue sections were prepared and stained with
H&E. Female CD-1 mice were given an i.p. bolus injection of
METVAN in 0.2 ml of PBS supplemented with 10% DMSO, or
                                                                     Fig. 3 Effect of METVAN on U87 and MDA-MB-231 cell adhesion to
0.2 ml of PBS supplemented with 10% DMSO alone (control              ECM proteins. The U87 glioblastoma (A) and MDA-MB-231 breast
mice). No sedation or anesthesia was used throughout the treat-      cancer (B) cells were preincubated with various concentrations of
ment period. Mice were monitored daily for mortality for de-         METVAN and processed for adhesion assays. METVAN inhibited
termination of the day-30 LD50 values. Blood samples were            adhesion of the U87 and MDA-MB-231 cells to ECM proteins. Data
                                                                     points, the mean (⫾SE) values from three independent experiments.
collected prior to and after METVAN administration for a
complete blood cell count (CBC) with differential and platelets,
serum samples for total protein, albumin, bilirubin/ALT, alka-
line phosphatase, creatinine, CPK, and electrolytes. Mice sur-             Pharmacokinetic Studies in Mice. Female CD-1 mice
viving until the end of the 30-days monitoring were killed. and      (6 – 8 weeks old) from Charles River (Wilmington, MA) were
the tissues were immediately collected from randomly selected        housed in a controlled environment (12-h light/12-h dark pho-
mice and preserved in 10% neutral buffered formalin. Standard        toperiod, 22 ⫾ 1°C, 60 ⫾ 10% relative humidity). The mice
tissues collected for histological evaluation included: bone, bone   were allowed free access to autoclaved pellet food and tap water
marrow, brain, cecum, heart, kidney, large intestine, liver, lung,   throughout the study. The animal studies are approved by the
lymph node, ovary, pancreas, skeletal muscle, skin, small intes-     Parker Hughes Institute Animal Care and Use Committee, and
tine, spleen, stomach, thymus, thyroid gland, urinary bladder,       all animal care procedures conformed to the Guide for the Care
and uterus (as available).                                           and Use of Laboratory Animals (National Research Council,
      SCID Mouse Xenograft Models of Human Glioblas-                 National Academy Press, Washington DC 1996). A solution
toma and Breast Cancer. The right and left hind legs of the          (100 ␮l) of METVAN (dissolved in 5% DMSO in PBS) was
SCID mice were inoculated s.c. with 1 ⫻ 106 U87 human                administered i.p. to mice at a dose of 10 mg/kg. This volume of
glioblastoma or 1 ⫻ 106 MDA-MB-231 breast cancer cells in            DMSO is well tolerated by mice when administrated by rapid
0.1 ml of PBS. Mice were treated with i.p.-administered injec-       i.v. or extravascular injection (40, 41). Blood samples (⬃500
tions of METVAN (10 mg/kg in 5% DMSO in PBS; n ⫽ 10) or              ␮l) were obtained from the ocular venous plexus by retro-orbital
vehicle alone (5% DMSO in PBS; n ⫽ 10). Injections were              venipuncture at 0, 2, 5, 10, 15, 30, 45 min, and 1, 2, 4, 6, and
given once daily, 5 days per week, for 4 consecutive weeks           24 h after the i.p. injection. All of the collected blood samples
beginning the day after inoculation of the tumor cells. Mice         were heparinized and centrifuged at 7000 ⫻ g for 5 min to
were monitored daily for health status as well as tumor growth       separate the plasma fraction from the whole blood. The plasma
and were killed if they became moribund or developed tumors          samples were then processed immediately, using the procedure
that impeded their ability to attain food or water, or at the end    described below for determining vanadium concentrations.
of the six-week observation period. Tumors were measured                   Determination of Plasma METVAN Levels by Induc-
using Vernier calipers three times per week. Tumor volumes           tively Coupled Plasma Atomic Emission Spectroscopy. All
were calculated according to the following formula, as described     of the glassware was kept in 10% nitric acid for at least 48 h and
previously (32, 39): tumor volume ⫽ (width)2 (length)/2.             was subsequently washed with distilled and Millipore water

    Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                    Research.
2128 In Vivo Antitumor Activity of METVAN

                                                                                                               Fig. 4 Effects of METVAN
                                                                                                               on glioblastoma cell migration
                                                                                                               from spheroids. U373 glioblas-
                                                                                                               toma spheroids with diameters
                                                                                                               of 200 –356 ␮m were treated
                                                                                                               for 2 h with 1 ␮M (B), 2 ␮M (C)
                                                                                                               or 5 ␮M (D) METVAN in 0.1%
                                                                                                               DMSO or with 0.1% DMSO
                                                                                                               alone (A). The control cells mi-
                                                                                                               grated 745 ⫾ 29 ␮m from the
                                                                                                               spheroids, whereas treatment of
                                                                                                               the spheroids with 1 ␮M (B), 2
                                                                                                               ␮M (C) and 5␮M (D) METVAN
                                                                                                               resulted in migration distances
                                                                                                               of only 311.1 ⫾ 54.4 ␮m,
                                                                                                               214.9 ⫾ 25.7 ␮m and 32.6 ⫾
                                                                                                               8.4 ␮m (corresponding to 58.3
                                                                                                               ⫾ 7.3%, 71.2 ⫾ 3.4%, and 95.6
                                                                                                               ⫾ 5.8% inhibition of tumor cell
                                                                                                               migration), respectively.

    before use. Standard solutions containing 0 –200 ␮g/liter of
    vanadium were prepared by diluting 100 ␮l of plasma samples
    containing various concentrations of METVAN with 3.7% HCl
    to 5 ml. The plasma samples from mice (100 ␮l) were diluted
    with 3.7% HCl to 5 ml and were analyzed by inductively
    coupled plasma atomic emission (ICP) spectroscopy carried out
    in the Research Analytical Laboratory at University of Minne-
    sota using a Perkin-Elmer Optima 3000DV spectrometer. The
    analytical wavelength was 292.396 nm with plasma gas flow of
    15 liters/min, auxiliary gas flow of 0.5 liter/min and nebulizer
    gas flow of 0.75 liter/min. The output power was 1350 W. The
    reporting limit for vanadium was 0.001 ␮g/ml. (Reporting limit
    is based on the concept of the lowest quantitatively determinable
    concentration (LQDC). Precision at the LQDC is approximately
    ⫾10% and analytical results are quantitative.).
          Pharmacokinetic Analysis. Pharmacokinetic modeling
    and parameter calculations were carried out using the WinNon-
    lin Professional Version 3.0 (Pharsight, Inc., Mountain, CA)
    pharmacokinetics software (35, 36, 42, 43). An appropriate
    model was chosen on the basis of the lowest sum of weighted
    squared residuals, the lowest Schwartz criterion (SC), the lowest
    Akaike’s information criterion (AIC) value, the lowest SEs of
    the fitted parameters, and the dispersion of the residuals. The
    elimination t1/2 life was estimated by linear regression analysis
    of the terminal phase of the plasma concentration-time profile.
    The AUC was calculated according to the linear trapezoidal rule     Fig. 5 Anti-invasive activity of METVAN against U87 glioblastoma
    between the first sampling time (0 h) and the last sampling time    and MDA-MB-231 breast cancer cells. Cells were incubated for 24 h
    plus C/k, where C is the concentration of the last sampling and     with METVAN in concentrations ranging from 0.5 to 10 ␮M. The cells
    k is the elimination rate constant. The systemic clearance (CL)     were then trypsinized and processed for invasion assays using Matrigel
                                                                        matrix-coated Boyden chambers. Data points, the mean (⫾SE) values
    was determined by dividing the dose by the AUC.                     from 3 independent experiments. The mean (⫾ SE) IC50 values were
          In Situ Detection of Apoptosis. Apoptosis was detected        0.78 ⫾ 0.4 ␮M and 0.61 ⫾ 0.3 ␮M for the U87 and MDA-MB-231 cells,
    using an in situ cell death detection kit (Boehringer Mannheim      respectively.

         Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                         Research.
Clinical Cancer Research 2129

                                            Table 1   Laboratory studies of METVAN toxicity in mice
                                                                                      METVAN dose level (mg/kg)
                                        Vehicle
        Laboratory test                (n ⫽ 20)            12.5 (n ⫽ 20)            25 (n ⫽ 20)           50 (n ⫽ 20)            100 (n ⫽ 20)
     Hematology
       WBCa (⫻ 109/liter)              8.7 ⫾ 0.9             9.6 ⫾ 0.7               9.2 ⫾ 0.8              9.9 ⫾ 0.9             10.6 ⫾ 0.9
       Hgb (g/dl)                     14.0 ⫾ 0.2            13.8 ⫾ 0.2              14.1 ⫾ 0.2             13.7 ⫾ 0.1             13.2 ⫾ 0.2
                     9
       Plt. ct. (⫻ 10 /liter)         488 ⫾ 65              400 ⫾ 59                452 ⫾ 65               392 ⫾ 71               530 ⫾ 101
     Electrolytes and kidney function
       K (mmol/liter)                  3.6 ⫾ 0.1              3.7 ⫾ 0.1               4.0 ⫾ 0.3             3.3 ⫾ 0.1              3.3 ⫾ 0.1
       Ca (mg/dl)                      8.7 ⫾ 0.1              8.7 ⫾ 0.1               8.8 ⫾ 0.2             8.9 ⫾ 0.1              9.4 ⫾ 0.1
       PO4 (mg/dl)                     6.6 ⫾ 0.1              6.1 ⫾ 0.2               6.3 ⫾ 0.2             6.9 ⫾ 0.2              6.6 ⫾ 0.2
       Creat. (mg/dl)                  0.4 ⫾ 0.0              0.3 ⫾ 0.0               0.4 ⫾ 0.0             0.3 ⫾ 0.0              0.4 ⫾ 0.0
     Metabolism and liver function
       Alb. (g/dl)                     1.6 ⫾ 0.0             1.6 ⫾ 0.0               1.5 ⫾ 0.0              1.5 ⫾ 0.0              1.5 ⫾ 0.0
       Total protein (g/dl)            4.9 ⫾ 0.1             5.0 ⫾ 0.1               4.8 ⫾ 0.1              4.6 ⫾ 0.1              4.6 ⫾ 0.1
       ALT (IU/liter)                   71 ⫾ 3.2              40 ⫾ 6                  65 ⫾ 15                49 ⫾ 5                 38 ⫾ 5
       Alk. Ptse. (IU/liter)            99 ⫾ 6                98 ⫾ 7                 123 ⫾ 11               106 ⫾ 6                106 ⫾ 5
       Bili. (mg/dl)                   0.4 ⫾ 0.0             0.4 ⫾ 0.0                0.5 ⫾ 0.1              0.4 ⫾ 0.0              0.5 ⫾ 0.0
       LD-L (IU/liter)                 536 ⫾ 77              709 ⫾ 101               666 ⫾ 83               554 ⫾ 41               598 ⫾ 74
     Cardiac enzymes
       CPK (IU/liter)                  581 ⫾ 188             682 ⫾ 222               363 ⫾ 65               396 ⫾ 85               482 ⫾ 163
    a
     WBC, white blood count; Hgb, hemoglobin; Plt. ct., platelet count; K, potassium; Ca, calcium, PO4, phosphorous; Creat., creatinine; Alb.,
albumin; Bili., bilirubin; LD-L, lactate dehydrogenase-liver derived; ALT, alanine aminotransferase; Alk. Ptse., alkaline phosphatase; CPK, creatine
phosphokinase.

Corp., Indianapolis, IN) as described earlier (27, 44). Cells were          mine whether METVAN is capable of impairing the adhesion of
incubated with either METVAN in 0.1% DMSO or 1:16-diluted                   cancer cells to ECM proteins, cells were incubated for 16 h with
plasma samples from METVAN-treated mice for 48 h at 37°C,                   noncytotoxic concentrations of METVAN (the cytotoxic IC50
and were fixed, permeabilized, incubated with the reaction mix-             values for 16 h incubation were 16.3 ⫾ 1.8 ␮M for U87 and
ture containing TdT- and FITC-conjugated dUTP, and counter-                 11.1 ⫾ 1.8 ␮M for MDA-MB-231 (see Fig. 1C) and the integrin-
stained with propidium iodide. Cells were transferred to slides             mediated cancer cell adhesion was examined. As shown in Fig.
and viewed with a confocal laser scanning microscope (Bio-Rad               3, pretreatment of cells with noncytotoxic concentrations of
MRC 1024) mounted on a Nikon Eclipse E800 series upright                    METVAN inhibited the adhesion of U87 glioblastoma and
microscope as reported previously (27, 44).                                 MDA-MB-231 breast cancer cells to laminin-, fibronectin-,
                                                                            vitronectin-, and collagen-coated plates. The inhibition of glio-
RESULTS AND DISCUSSION                                                      blastoma cell adhesion was concentration-dependent with aver-
      In Vitro Activity of METVAN against Glioblastoma and                  age IC50 values of 1.4 ⫾ 0.1 ␮M, 1.3 ⫾ 0.1 ␮M, 1.2 ⫾ 0.1 ␮M,
Breast Cancer Cells. The cytotoxic activity of METVAN                       and 1.2 ⫾ 0.2 ␮M for laminin, fibronectin, vitronectin and
against brain tumor and breast cancer cells was first confirmed             collagen, respectively. The corresponding IC50 values for the
using MTT assays. As shown in Fig. 1, METVAN exhibited                      MDA-MB-231 breast cancer cells were 1.0 ⫾ 0.1 ␮M, 1.0 ⫾ 0.0
potent cytotoxicity against each of the five brain tumor and four           ␮M, 1.1 ⫾ 0.1 ␮M, and 1.3 ⫾ 0.1 ␮M, respectively.
breast cancer cell lines with nanomolar or low micromolar IC50                    Dissociation and migration are the initial steps for infiltra-
values. The METVAN-induced cell death was confirmed to be                   tion of tumor cells into the surrounding tissue (53–55). To
apoptotic using the TUNEL of exposed 3⬘-OH termini of DNA                   determine whether METVAN is capable of preventing the mi-
with dUTP-FITC. As shown in the confocal laser scanning                     gration of tumor cells, U373 glioblastoma multicellular sphe-
microscopy images in Fig. 2, METVAN-treated U87 glioblas-                   roids were used. The spheroids were prepared by plating the
toma and MDA-MB-231 breast cancer cells, examined for                       cells over agar plates as described above. Spheroids measuring
dUTP-FITC incorporation (green fluorescence) and propidium                  259.2 ⫾ 41.9 ␮m in diameter were incubated with METVAN or
iodide counterstaining (red fluorescence), exhibited many ap-               with DMSO on fibronectin-coated coverslips. As shown in Fig.
optotic yellow nuclei (superimposed green and red fluores-                  4, U373 glioblastoma cells incubated with DMSO rapidly mi-
cence) at 24 h after treatment.                                             grated to a distance 745 ⫾ 29 ␮m away from the spheroid.
      During the multistep process of tissue invasion, tumor cells          Treatment of the spheroids with METVAN inhibited tumor-cell
initially adhere to the ECM proteins via cell surface integrin              migration in a concentration-dependent fashion with an IC50
receptors and then gain migratory capacity to enter the sur-                value of ⬍1 ␮M. Treatment with 1 ␮M, 2 ␮M, and 5 ␮M
rounding tissue. ECM proteins such as laminin, fibronectin,                 METVAN resulted in 58.3 ⫾ 7.3% (migration distance of
vitronectin and type IV collagen are thought to play an impor-              311.1 ⫾ 54.4 ␮m), 71.2 ⫾ 3.4% (migration distance of 214.9 ⫾
tant role in tumor cell attachment and migration because these              25.7 ␮m), and 95.6 ⫾ 5.8% (migration distance of 32.6 ⫾ 8.4
proteins have been found in the basal lamina that promote the               ␮m) inhibition of tumor cell migration from the spheroids,
adhesion and invasion of tumor cells in situ (45–52). To deter-             respectively (Fig. 4).

    Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                    Research.
2130 In Vivo Antitumor Activity of METVAN

                                                                                               Fig. 6 Pharmacokinetic features of
                                                                                               METVAN in mice. The measured plasma
                                                                                               concentration of vanadium (E) as a function
                                                                                               of time in mice after i.p. injection of 10
                                                                                               mg/kg METVAN (five mice per time point).
                                                                                               A, plasma concentration-time curve. B, com-
                                                                                               puter-fitted pharmacokinetic parameter val-
                                                                                               ues. The pharmacokinetic parameters in
                                                                                               CD-1 mice (n ⫽ 5 mice per time point) are
                                                                                               presented as the average values estimated
                                                                                               from composite plasma concentration-time
                                                                                               curves of pooled data; in parentheses,
                                                                                               mean ⫾ SE values; CL, systemic clearance
                                                                                               from plasma; Cmax, measured maximum
                                                                                               plasma concentration; t ⁄ , terminal elimina-
                                                                                                                        12

                                                                                               tion half-life; tmax, the time required to reach
                                                                                               the maximum plasma drug concentration af-
                                                                                               ter i.p. administration; Vc, central volume of
                                                                                               distribution. aApparent Vc and systemic CL
                                                                                               without correction for bioavailability (F).

          Tumor invasion of the basement membrane is a crucial step   injections of METVAN at dose levels of 12.5 mg/kg (n ⫽ 40),
    in the complex multistage process that leads to metastatic        25 mg/kg (n ⫽ 40), or 50 mg/kg (n ⫽ 40). Twenty mice from
    spread. Tumor cells cross the basement membrane as they           each group were electively killed on days 7 and 30 after the
    initially invade the lymphatic or vascular beds during dissemi-   administration of METVAN. METVAN was not toxic to mice at
    nation and as they penetrate their target tissues (56 –59). The   these dose levels. None of the 120 mice treated with i.p. bolus
    Matrigel matrix, an artificial basement membrane composed of      injections of METVAN in this dose range experienced side
    growth factors and several ECM components such as collagens,      effects or died of toxicity (data not shown). No evidence of
    laminin, and proteoglycans, was used to examine the effect of     acute toxicity was evident from laboratory tests performed on
    METVAN on tumor cell invasion. To determine whether               the blood samples from mice killed on day 7 (Table 1). In
    METVAN impairs cancer cell invasion through Matrigel, the         particular, there was (a) no neutropenia, anemia, or thrombocy-
    cells were incubated with various noncytotoxic concentrations     topenia suggestive of a hematological toxicity, (b) no increase in
    of the compound, and then the invasion was examined using         serum creatinine levels or an electrolyte imbalance suggestive of
    MDA-MB-231 and U87 cells (Fig. 5). METVAN inhibited the           renal toxicity, (c) no elevation of liver enzymes or total bilirubin
    invasion of tumor cells through Matrigel matrix in a concentra-   levels suggestive of hepatic toxicity, and (d) no CPK elevation
    tion-dependent fashion with mean IC50 values of 0.78 ⫾ 0.4 ␮M     suggestive of cardiac toxicity (Table 1). Similarly, no laboratory
    and 0.6 ⫾ 0.3 ␮M for U87 and MDA-MB-231 cells, respec-            abnormalities suggestive of subacute toxicity were evident from
    tively.                                                           analysis of blood samples obtained from mice killed on day 30.
          Taken together, these findings show that treatment with     No histopathological lesions were found in the organs of
    METVAN at concentrations ⬎1 ␮M is associated with a nearly        METVAN-treated mice that were electively killed at day 7 or
    complete loss of the adhesive, migratory, and invasive proper-    day 30.
    ties of the treated tumor cell populations.                            We next sought to determine whether cytotoxic concentra-
          In Vivo Pharmacokinetic Features of METVAN after            tions of METVAN are achievable in vivo at nontoxic dose
    i.p. Administration. We first evaluated the toxicity of           levels. To this end, a single nontoxic 10 mg/kg bolus dose of
    METVAN in mice to identify nontoxic dose levels for pharma-       METVAN was administered to mice i.p., and the plasma vana-
    codynamic studies. Mice were treated with single i.p. bolus       dium concentrations were determined at various time points

         Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                         Research.
Clinical Cancer Research 2131

Fig. 7 Anticancer activity of plasma
samples from METVAN-treated mice.
Plasma samples, obtained from vehicle-
treated (A, B) and METVAN-treated (A⬘,
B⬘) mice 30 min after administration of the
drug, were diluted with PBS 1:16 and then
tested for their cytotoxic activity against
MDA-MB-231 human breast cancer (A,
A⬘) and NALM-6 human leukemic (B, B⬘)
cells in vitro. After a 48-h incubation, cells
were examined for apoptotic changes us-
ing standard TUNEL/apoptosis assays, as
described in “Materials and Methods.”

after the injection using inductively coupled plasma atomic         MDA-MB-231 breast cancer cells. METVAN significantly de-
emission (ICP) spectroscopy. A two-compartment, first-order         layed the onset of tumor growth and inhibited visible tumor
pharmacokinetic model was fit to the pharmacokinetic data (Fig.     progression when administered i.p. in single daily injections (10
6, A and B). The computer-fitted pharmacokinetic parameter          mg/kg/dose) given 5 days per week for 4 weeks beginning the
values are shown in Fig. 6B. Therapeutic plasma concentrations      day after s.c. inoculation of the tumor cells (Fig. 8). All of the
ⱖ5␮M, which are highly cytotoxic against human cancer cells,        control mice developed measurable tumors within 7 days,
were rapidly achieved and maintained in mice for at least 24 h      whereas 40% of the mice with U87 xenografts treated with
after i.p. bolus injection of a single 10-mg/kg nontoxic dose of    METVAN remained alive and free of detectable tumors for ⬎16
METVAN. At this dose level, the maximum plasma METVAN               days. At 40 days after the inoculation of tumor cells, the average
concentration (Cmax) was 37.0 ␮M, which was achieved with a         size of the U87 tumor xenografts in vehicle-treated control
tmax of 21.4 min. Notably, 1:16-diluted plasma samples obtained     SCID mice was 4560 ⫾ 654 mm3, whereas the average size of
from METVAN-treated mice 30 min after the administration of         the U87 tumors in METVAN-treated SCID mice was only
the drug killed 85% of MDA-MB-231 human breast cancer cells         1688 ⫾ 571 mm3 (P ⫽ 0.003; Fig. 8A). Similarly, the average
and ⬎99% of NALM-6 human leukemia cells in vitro (Fig. 7).          size of MDA-MB-231 tumors in vehicle-treated control SCID
METVAN was slowly eliminated with an apparent plasma                mice at 40 days after tumor cell inoculation was 487 ⫾ 82 mm3,
half-life of 17.5 h and systemic clearance of 42.1 ml/h/kg, which   whereas the average size of MDA-MB-231 tumors in
is much slower than the blood flow to liver (5400 ml/h/kg) or       METVAN-treated SCID mice was 174 ⫾ 29 mm3 (P ⫽ 0.002;
kidney (3900 ml/h/kg). The apparent volume distribution of          Fig. 8B). Ninety % of the control mice developed measurable
METVAN at central compartment was 276.5 ml/kg [⬃5-fold              tumors within 7 days, whereas 50% of the mice treated with
greater than the plasma volume (50 ml/kg) but less than the total   METVAN remained alive and free of detectable tumors for ⬎19
body water (725 ml/kg); Ref. 60], indicating that METVAN            days.
might have limited extravascular distribution after i.p. adminis-        In summary, in accordance with its potent in vitro activity
tration.                                                            and favorable in vivo pharmacokinetics, METVAN exhibited
      In Vivo Antitumor Activity of METVAN in SCID                  significant antitumor activity and delayed tumor progression in
Mouse Xenograft Models of Human Glioblastoma and                    CB.17 SCID mouse xenograft models of human glioblastoma
Breast Cancer. CB.17 SCID mice developed rapidly growing            and breast cancer. The favorable in vivo pharmacodynamic
tumors after s.c. inoculation of 1 ⫻ 106 U87 glioblastoma or        features and antitumor activity of METVAN warrants further

     Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                     Research.
2132 In Vivo Antitumor Activity of METVAN

                                                                              7. Schwartz, M. K. Role of trace elements in cancer. Cancer Res., 35:
                                                                              3481–3487, 1975.
                                                                              8. Djordjevic, C. Antitumor activity of vanadium compounds. In: H.
                                                                              Sigel and A. Sigel (eds.), Metal Ions in Biological Systems, Vol. 31, pp.
                                                                              595– 616. New York: Marcel Dekker, 1995.
                                                                              9. Djordjevic, C., and Wampler, G. L. Antitumor activity and toxicity
                                                                              of peroxo heteroligand vanadates(V) in relation to biochemistry of
                                                                              vanadium. J. Inorg. Biochem., 25: 51–55, 1985.
                                                                              10. Schieven, G. L., Wahl, A. F., Myrdal, S., Grosmaire, L., and
                                                                              Ledbetter, J. A. Lineage-specific induction of B cell apoptosis and
                                                                              altered signal transduction by the phosphotyrosine phosphatase inhibitor
                                                                              bis(maltolato)oxovanadium(IV). J. Biol. Chem., 270: 20824 –20831,
                                                                              1995.
                                                                              11. Faure, R., Vincent, M., Dufour, M., Shaver, A., and Posner, B. I.
                                                                              Arrest at the G2/M transition of the cell cycle by protein-tyrosine
                                                                              phosphatase inhibition: studies on a neuronal and a glial cell line. J. Cell.
                                                                              Biochem., 59: 389 – 401, 1995.
                                                                              12. Kopf-Maier, P., Wagner, W., and Kopf, H. In vitro cell growth
                                                                              inhibition by metallocene dichlorides. Cancer Chemother. Pharmacol.,
                                                                              5: 237–241, 1981.
                                                                              13. Kopf-Maier, P., Wagner, W., and Liss, E. Induction of cell arrest at
                                                                              G1/S and in G2 after treatment of Ehrlich ascites tumor cells with
                                                                              metallocene dichlorides and cis-platinum in vitro. J. Cancer Res. Clin.
                                                                              Oncol., 106: 44 –52, 1983.
                                                                              14. Kopf-Maier, P., Wagner, W., and Liss, E. Cytokinetic behavior of
                                                                              Ehrlich ascites tumor after in vivo treatment with cis-diamminedichlo-
    Fig. 8 In vivo antitumor activity of METVAN against human glioblas-       roplatinum(II) and metallocene dichlorides. J. Cancer Res. Clin. Oncol.,
    toma and breast cancer in SCID mice. Shown are the effects of daily       102: 21–30, 1981.
    treatment with METVAN (10 mg/kg/day) or 5% DMSO in PBS (given             15. Barceloux, D. G. Vanadium. J. Toxicol. Clin. Toxicol. 37: 265–278,
    5 days per week for 4 weeks beginning the day after inoculation) on       1999.
    tumor growth and progression of glioblastoma (A) and breast cancer (B).   16. Golden, M. H., and Golden, B. E. Trace elements. Potential impor-
    Comparisons between groups were done using the log-rank test.             tance in human nutrition with particular reference to zinc and vanadium.
                                                                              Br. Med. Bull., 37: 31–36, 1981.
                                                                              17. Macara, I. G. Vanadium: an element in search of a role. Trends
                                                                              Biochem. Sci., 5: 92–94, 1980.
    development of this novel oxovanadium compound as a poten-                18. Rehder, D. The bioinorganic chemistry of vanadium. Angew.
    tial new anticancer agent. More preclinical work remains to be            Chem. Int. Ed. Engl., 130: 148 –167, 1991.
    done to determine the optimal dose and schedule for METVAN.               19. Krejsa, C. M., Nadler, S. G., Esselstyn, J. M., Kavanagh, T. J.,
    We are planning to evaluate the efficacy of METVAN in com-                Ledbetter, J. A., and Schieven, G. L. Role of oxidative stress in the
                                                                              action of vanadium phosphotyrosine phosphatase inhibitors. Redox in-
    parison with, as well as in combination with, standard anticancer         dependent activation of NF-␬B. J. Biol. Chem., 272: 11541–11549,
    drugs, including taxanes (e.g., Taxol, Taxotere), thymidylate             1997.
    synthase inhibitors anthracyclins (e.g., Adriamycin), topoi-              20. Nechay, B. R. Mechanisms of action of vanadium. Annu. Rev.
    somerase I inhibitors (e.g., irinotecan, topotecan), and topoi-           Pharmacol. Toxicol., 24: 501–524, 1984.
    somerase II inhibitors (etoposide), and alkylating agents (e.g.,          21. Abou-Seif, M. A. Oxidative stress of vanadium-mediated oxygen
    ifosfamid, cyclophosphamide).                                             free radical generation stimulated by aluminium on human erythrocytes.
                                                                              Ann. Clin. Biochem., 35: 254 –260, 1998.
                                                                              22. Shi, X., Jiang, H., Mao, Y., Ye, J., and Saffiotti, U. Vanadium(IV)-
    REFERENCES                                                                mediated free radical generation and related 2⬘-deoxyguanosine hy-
    1. Tolman, E. L., Barris, E., Burns, M., Pansini, A., and Partridge, R.   droxylation and DNA damage. Toxicology, 106: 27–38, 1996.
    Effects of vanadium on glucose metabolism in vitro. Life Sci., 25:        23. El-Naggar, M. M., El-Waseef, A. M., El-Halafawy, K. M., and
    1159 –1164, 1979.                                                         El-Sayed, I. H. Antitumor activities of vanadium(IV), manganese(IV),
    2. Dubyak, G. R., and Kleinzeller, A. The insulin-mimetic effects of      iron(III), cobalt(II) and copper(II) complexes of 2-methylaminopyri-
    vanadate in isolated rat adipocytes. Dissociation from effects of vana-   dine. Cancer Lett., 133: 71–76, 1998.
    date as a (Na⫹-K⫹)ATPase inhibitor. J. Biol. Chem., 255: 5306 –5312,      24. Byczkowski, J. Z., Wan, B., and Kulkarni, A. P. Vanadium-medi-
    1980.                                                                     ated lipid peroxidation in microsomes from human term placenta. Bull.
    3. Shechter, Y., and Karlish, S. J. Insulin-like stimulation of glucose   Environ. Contam. Toxicol., 41: 696 –703, 1988.
    oxidation in rat adipocytes by vanadyl (IV) ions. Nature (Lond.), 284:    25. Tsiani, E., and Fantus, I. G. Vanadium compounds. Biological
    556 –558, 1980.                                                           actions and potential as pharmacological agents. Trends Endocrinol.
    4. Heyliger, C. E., Tahiliani, A. G., and McNeill, J. H. Effect of        Metabol., 8: 51–58, 1997.
    vanadate on elevated blood glucose and depressed cardiac performance      26. Dong, Y., Narla, R. K., Sudbeck, E., and Uckun, F. M. Synthesis,
    of diabetic rats. Science (Wash. DC), 227: 1474 –1477, 1985.              X-ray structure, and anti-leukemic activity of oxovanadium(IV) com-
    5. Morinville, A., Maysinger, D., and Shaver, A. From Vanadis to          plexes. J. Inorg. Biochem., 78: 321–330, 2000.
    Atropos: vanadium compounds as pharmacological tools in cell death        27. Narla, R. K., Dong, Y., D’Cruz, O. J., Navara, C., and Uckun, F. M.
    signalling. Trends Pharmacol. Sci., 19: 452– 460, 1998.                   Bis(4,7-dimethyl-1,10-phenanthroline) sulfatooxovanadium(IV) as a
    6. Kopf-Maier, P. Complexes of metals other than platinum as antitu-      novel apoptosis-inducing anticancer agent. Clin. Cancer Res., 6: 1546 –
    mour agents. Eur. J. Clin. Pharmacol., 47: 1–16, 1994.                    1556, 2000.

         Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                         Research.
Clinical Cancer Research 2133

28. Narla, R. K., Dong, Y., Ghosh, P., Thoen, K., and Uckun, F. M.         43. Chen, C. L., Levine, A., Rao, A., O’Neill, K., Messinger, Y., Myers,
Apoptosis-inducing vanadium complexes. Drugs Future, 25: 1053–             D. E., Goldman, F., Hurvitz, C., Casper, J. T., and Uckun, F. M. Clinical
1068, 2000.                                                                pharmacokinetics of the CD19 receptor-directed tyrosine kinase inhib-
29. Narla, R. K., Dong, Y., and Uckun, F. M. Apoptosis-inducing novel      itor B43-Genistein in patients with B-lineage lymphoid malignancies.
antileukemic agent, bis(4,7-dimethyl-1,10 phenanthroline) sulfatoox-       J Clin. Pharmacol., 39: 1248 –1255, 1999.
ovanadium(IV) [VO(SO4)(Me2-Phen)2] depolarizes mitochondrial               44. Zhu, D. M., Narla, R. K., Fang, W. H., Chia, N. C., and Uckun,
membranes. Leuk. Lymphoma, 41: 625– 634, 2001.                             F. M. Calphostin C triggers calcium-dependent apoptosis in human
30. Narla, R. K., Dong, Y., Klis, D., and Uckun, F. M. Bis(4,7-            acute lymphoblastic leukemia cells. Clin. Cancer Res., 4: 2967–2976,
dimethyl-1,10-phenanthroline) sulfatooxovanadium(IV) (METVAN) as           1998.
a novel antileukemic agent with matrix metalloproteinase-inhibitory        45. Carbonetto, S. Facilitatory and inhibitory effects of glial cells and
activity. Clin. Cancer Res., 7: 1094 –1101, 2001.                          extracellular matrix in axonal regeneration. Curr. Opin. Neurobiol., 1:
31. Narla, R. K., Liu, X. P., Klis, D., and Uckun, F. M. Inhibition of     407– 413, 1991.
human glioblastoma cell adhesion and invasion by 4-(4⬘- hydroxylphe-       46. Carbonetto, S., and Cochard, P. In vitro studies on the control of
nyl)-amino-6,7-dimethoxyquinazoline (WHI-P131) and 4-(3⬘-bromo-            nerve fiber growth by the extracellular matrix of the nervous system.
4⬘-hydroxylphenyl)-amino-6,7-dimethoxyquinazoline           (WHI-P154).    J Physiol., 82: 258 –270, 1987.
Clin. Cancer Res., 4: 2463–2471, 1998.
                                                                           47. Giese, A., Loo, M. A., Rief, M. D., Tran, N., and Berens, M. E.
32. Narla, R. K., Liu, X. P., Myers, D. E., and Uckun, F. M. 4-(3⬘-        Substrates for astrocytoma invasion. Neurosurgery (Baltimore), 37:
Bromo-4⬘hydroxylphenyl)-amino-6,7-dimethoxyquinazoline: a novel
                                                                           294 –301; discussion, 301–302, 1995.
quinazoline derivative with potent cytotoxic activity against human
glioblastoma cells. Clin. Cancer Res., 4: 1405–1414, 1998.                 48. Giese, A., and Westphal, M. Glioma invasion in the central nervous
                                                                           system. Neurosurgery (Baltimore), 39: 235–250; discussion 250 –252,
33. Ghosh, S., Narla, R. K., Zheng, Y., Liu, X. P., Jun, X., Mao, C.,
                                                                           1996.
Sudbeck, E. A., and Uckun, F. M. Structure-based design of potent
inhibitors of EGF-receptor tyrosine kinase as anti-cancer agents. Anti-    49. Merzak, A., Koochekpour, S., and Pilkington, G. J. Adhesion of
cancer Drug Des., 14: 403– 410, 1999.                                      human glioma cell lines to fibronectin, laminin, vitronectin and collagen
34. Albini, A., Iwamoto, Y., Kleinman, H. K., Martin, G. R., Aaronson,     I is modulated by gangliosides in vitro. Cell Adhes. Commun., 3:
S. A., Kozlowski, J. M., and McEwan, R. N. A rapid in vitro assay for      27– 43, 1995.
quantitating the invasive potential of tumor cells. Cancer Res., 47:       50. Rooprai, H. K., Vanmeter, T., Panou, C., Schnull, S., Trillo-Pazos,
3239 –3245, 1987.                                                          G., Davies, D., and Pilkington, G. J. The role of integrin receptors in
35. Uckun, F. M., Ek, O., Liu, X. P., and Chen, C. L. In vivo toxicity     aspects of glioma invasion in vitro. Int. J. Dev. Neurosci., 17: 613– 623,
and pharmacokinetic features of the janus kinase 3 inhibitor WHI-P131      1999.
[4-(4⬘hydroxyphenyl)-amino-6,7-dimethoxyquinazoline. Clin. Cancer          51. Rutka, J. T., Apodaca, G., Stern, R., and Rosenblum, M. The
Res., 5: 2954 –2962, 1999.                                                 extracellular matrix of the central and peripheral nervous systems:
36. Uckun, F. M., Bellomy, K., O’Neill, K., Messinger, Y., Johnson, T.,    structure and function. J. Neurosurg., 69: 155–170, 1988.
and Chen, C. L. Toxicity, biological activity, and pharmacokinetics of     52. Venstrom, K. A., and Reichardt, L. F. Extracellular matrix. 2: role
TXU (anti-CD7)-pokeweed antiviral protein in chimpanzees and adult         of extracellular matrix molecules and their receptors in the nervous
patients infected with human immunodeficiency virus. J. Pharmacol.         system. FASEB J., 7: 996 –1003, 1993.
Exp. Ther., 291: 1301–1307, 1999.                                          53. Pilkington, G. J., Bjerkvig, R., De Ridder, L., and Kaaijk, P. In vitro
37. Waurzyniak, B., Schneider, E. A., Tumer, N., Yanishevski, Y.,          and in vivo models for the study of brain tumour invasion. Anticancer
Gunther, R., Chelstrom, L. M., Wendorf, H., Myers, D. E., Irvin, J. D.,    Res., 17: 4107– 4109, 1997.
Messinger, Y., Ek, O., Zeren, T., Langlie, M. C., Evans, W. E., and
                                                                           54. Giese, A., Kluwe, L., Laube, B., Meissner, H., Berens, M. E., and
Uckun, F. M. In vivo toxicity, pharmacokinetics, and antileukemic
                                                                           Westphal, M. Migration of human glioma cells on myelin. Neurosurgery
activity of TXU (anti-CD7)-pokeweed antiviral protein immunotoxin.
                                                                           (Baltimore), 38: 755–764, 1996.
Clin. Cancer Res., 3: 881– 890, 1997.
38. Uckun, F. M., Narla, R. K., Zeren, T., Yanishevski, Y., Myers,         55. Chintala, S. K., and Rao, J. K. Invasion of human glioma: role of
D. E., Waurzyniak, B., Ek, O., Schneider, E., Messinger, Y., Chelstrom,    extracellular matrix proteins. Front. Biosci., 1: d324 – d339, 1996.
L. M., Gunther, R., and Evans, W. In vivo toxicity, pharmacokinetics,      56. Nagano, N., Sasaki, H., Aoyagi, M., and Hirakawa, K. Invasion of
and anticancer activity of Genistein linked to recombinant human epi-      experimental rat brain tumor: early morphological changes following
dermal growth factor. Clin. Cancer Res., 4: 1125–1134, 1998.               microinjection of C6 glioma cells. Acta Neuropathol., 86: 117–125,
39. Friedman, H. S., Dolan, M. E., Pegg, A. E., Marcelli, S., Keir, S.,    1993.
Catino, J. J., Bigner, D. D., and Schold, S. C., Jr. Activity of temozo-   57. Pilkington, C. J. The role of the extracellular matrix in neoplastic
lomide in the treatment of central nervous system tumor xenografts.        glial invasion of the nervous system. Braz. J. Med. Biol. Res., 29:
Cancer Res., 55: 2853–2857, 1995.                                          1159 –1172, 1996.
40. Rosenkrantz, H., Hadidian, Z., Seay, H., and Masson, H. Dimethyl       58. Schiffer, D., Chio, A., Giordana, M. T., Mauro, A., Migheli, A., and
sulfoxide: its steroid solubility and endocrinologic and pharmacologic-    Vigliani, M. C. The vascular response to tumor infiltration in malignant
toxicologic characteristics. Cancer Chemother. Rep., 31: 7–24, 1963.       gliomas. Morphometric and reconstruction study. Acta Neuropathol.,
41. Wilson, J. E., Brown, D. E., and Timmens, E. K. A toxicological        77: 369 –378, 1989.
study of dimethyl sulfoxide. Toxicol. Appl. Pharmacol., 7: 104 –112,       59. Brown, L. F., Guidi, A. J., Schnitt, S. J., Van De Water, L.,
1965.                                                                      Iruela-Arispe, M. L., Yeo, T. K., Tognazzi, K., and Dvorak, H. F.
42. Chen, C. L., Malaviya, R., Navara, C., Chen, H., Bechard, B.,          Vascular stroma formation in carcinoma in situ, invasive carcinoma, and
Mitcheltree, G., Liu, X. P., and Uckun, F. M. Pharmacokinetics and         metastatic carcinoma of the breast. Clin. Cancer Res., 5: 1041–1056,
biologic activity of the novel mast cell inhibitor, 4-(3-hydroxyphenyl)-   1999.
amino-6,7-dimethoxyquinazoline in mice. Pharm Res. (NY), 16: 117–          60. Davies, B., and Morris, T. Physiological parameters in laboratory
122, 1999.                                                                 animals and humans [editorial]. Pharm Res., 10: 1093–1095, 1993.

    Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                    Research.
In Vivo Antitumor Activity of
Bis(4,7-dimethyl-1,10-phenanthroline) Sulfatooxovanadium(IV)
{METVAN [VO(SO4)(Me2-Phen)2]}
Rama Krishna Narla, Chun-Lin Chen, Yanhong Dong, et al.

Clin Cancer Res 2001;7:2124-2133.

 Updated version         Access the most recent version of this article at:
                         http://clincancerres.aacrjournals.org/content/7/7/2124

     Cited articles      This article cites 55 articles, 17 of which you can access for free at:
                         http://clincancerres.aacrjournals.org/content/7/7/2124.full#ref-list-1

     E-mail alerts       Sign up to receive free email-alerts related to this article or journal.

     Reprints and        To order reprints of this article or to subscribe to the journal, contact the AACR Publications
    Subscriptions        Department at pubs@aacr.org.

     Permissions         To request permission to re-use all or part of this article, use this link
                         http://clincancerres.aacrjournals.org/content/7/7/2124.
                         Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
                         Rightslink site.

          Downloaded from clincancerres.aacrjournals.org on January 15, 2021. © 2001 American Association for Cancer
                                                          Research.
You can also read