throbber
Antibody-Maytansinoid Conjugates Are Activated in
`Targeted Cancer Cells by Lysosomal Degradation and
`Linker-Dependent Intracellular Processing
`(cid:160)
`Hans K. Erickson, Peter U. Park, Wayne C. Widdison, et al.
`Cancer Res(cid:160)(cid:160)
`
`2006;66:4426-4433.
`
`(cid:160)
`(cid:160)
`(cid:160)
`(cid:160)
`
`(cid:160)
`(cid:160)
`
`Updated version
`(cid:160)
`Supplementary
`Material
`(cid:160)
`
`Access the most recent version of this article at:
`http://cancerres.aacrjournals.org/content/66/8/4426
`(cid:160)
`Access the most recent supplemental material at:
`http://cancerres.aacrjournals.org/content/suppl/2006/04/14/66.8.4426.DC1.html
`(cid:160)
`
`
`
`
`
`Cited Articles
`(cid:160)
`Citing articles
`(cid:160)
`
`This article cites by 22 articles, 11 of which you can access for free at:
`
`http://cancerres.aacrjournals.org/content/66/8/4426.full.html#ref-list-1
`(cid:160)
`This article has been cited by 33 HighWire-hosted articles. Access the articles at:
`
` http://cancerres.aacrjournals.org/content/66/8/4426.full.html#related-urls
`(cid:160)
`
`E-mail alerts
`(cid:160)
`Reprints and
`Subscriptions
`(cid:160)
`Permissions
`(cid:160)
`
`Sign up to receive free email-alerts
`
` related to this article or journal.
`
`To order reprints of this article or to subscribe to the journal, contact the AACR Publications
`.
`pubs@aacr.org
`Department at
`(cid:160)
`To request permission to re-use all or part of this article, contact the AACR Publications
`.
`permissions@aacr.org
`Department at
`(cid:160)
`
`
`
`
`
`Downloaded from Downloaded from cancerres.aacrjournals.org on June 10, 2014. © 2006 American Association for Cancercancerres.aacrjournals.org on June 10, 2014. © 2006 American Association for Cancer
`
`
`
`
`
`Research. Research.
`
`IMMUNOGEN 2175, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Research Article
`
`Antibody-Maytansinoid Conjugates Are Activated in Targeted Cancer
`Cells by Lysosomal Degradation and Linker-Dependent
`Intracellular Processing
`
`Hans K. Erickson, Peter U. Park, Wayne C. Widdison, Yelena V. Kovtun, Lisa M. Garrett,
`Karen Hoffman, Robert J. Lutz, Victor S. Goldmacher, and Walter A. Bla¨ttler
`
`ImmunoGen, Inc., Cambridge, Massachusetts
`
`Abstract
`Antibody-drug conjugates are targeted anticancer agents
`consisting of a cytotoxic drug covalently linked to a mono-
`clonal antibody for tumor antigen–specific activity. Once
`bound to the target cell-surface antigen, the conjugate must
`be processed to release an active form of the drug, which can
`reach its intracellular target. Here, we used both biological
`and biochemical methods to better define this process for
`antibody-maytansinoid conjugates. In particular, we exam-
`ined the metabolic fate in cells of huC242-maytansinoid
`conjugates containing either a disulfide linker (huC242-
`SPDB-DM4) or a thioether linker (huC242-SMCC-DM1). Using
`cell cycle analysis combined with lysosomal inhibitors, we
`showed that lysosomal processing is required for the activity
`of antibody-maytansinoid conjugates,
`irrespective of the
`linker. We also identified and characterized the released
`maytansinoid molecules from these conjugates, and measured
`their rate of release compared with the kinetics of cell cycle
`arrest. Both conjugates are efficiently degraded in lysosomes
`to yield metabolites consisting of the intact maytansinoid drug
`and linker attached to lysine. The lysine adduct is the sole
`metabolite from the thioether-linked conjugate. However, the
`lysine metabolite generated from the disulfide-linked conju-
`gate is reduced and S-methylated to yield the lipophilic and
`potently cytotoxic metabolite, S-methyl-DM4. These findings
`provide insight into the mechanism of action of antibody-
`maytansinoid conjugates in general, and more specifically,
`identify a biochemical mechanism that may account for the
`significantly enhanced antitumor efficacy observed with
`disulfide-linked conjugates. (Cancer Res 2006; 66(8): 4426-33)
`
`Introduction
`
`Antibody-drug conjugates are designed to selectively eliminate
`cancer cells that express the target cell surface antigen ( for
`recent reviews, see refs. 1–3). Several conjugates are currently in
`clinical evaluation (3) and one conjugate, gemtuzumab ozoga-
`micin, is licensed for the treatment of refractory acute myeloid
`leukemia (4).
`Antibody-drug conjugates are structurally composed of a
`monoclonal antibody that binds specifically to a target antigen, a
`
`Note: Supplementary data for this article are available at Cancer Research Online
`(http://cancerres.aacrjournals.org/).
`H.K. Erickson and P.U. Park contributed equally to this work.
`Requests for reprints: Hans K. Erickson, ImmunoGen, Inc., 128 Sidney Street,
`Cambridge, MA 02139. Phone: 617-995-2500; Fax: 617-995-2510; E-mail: hans.erickson@
`immunogen.com.
`I2006 American Association for Cancer Research.
`doi:10.1158/0008-5472.CAN-05-4489
`
`small chemical cytotoxic drug, and a linker that stably connects the
`two. We have developed conjugates that contain maytansinoid
`drugs, which are derivatives of the natural cytotoxic polyketide,
`maytansine (5). In particular, we have developed maytansinoid
`conjugates of the humanized monoclonal antibody, huC242, which
`binds to the CanAg antigen expressed on colorectal, pancreatic,
`and certain non–small cell lung cancers (6). During the preclinical
`development, we compared several conjugates of huC242 that
`differed in the chemical nature of the linker between huC242 and
`the maytansinoid and established that one conjugate, huC242-
`SPDB-DM4, had the highest activity in several human xenograft
`tumor models in mice and the widest therapeutic window as
`defined by the largest difference between the minimal effective
`dose and the maximal tolerated dose. Based on these results,
`huC242-SPDB-DM4 was selected for clinical development and is
`currently being tested in a phase I trial.
`Here, we report
`that
`the huC242-maytansinoid conjugate,
`huC242-SMCC-DM1, with a ‘‘noncleavable’’
`linker containing a
`thioether bond was at least as potent in vitro as the selected
`conjugate, huC242-SPDB-DM4, which has a ‘‘cleavable’’
`linker
`containing a disulfide bond. This was surprising because
`huC242-SMCC-DM1 displayed significantly lower in vivo activity
`in multiple xenograft tumor models. To investigate this conun-
`drum, we undertook a series of experiments to elucidate the
`mechanism of cell killing by the conjugates. The results delineate
`an activation process, for both conjugates, that requires lysosomal
`degradation of the antibody component of the conjugate. However,
`distinct maytansinoid metabolites produced by intracellular
`processing of huC242-SPDB-DM4 were identified and character-
`ized, providing a likely mechanism for its superior antitumor
`efficacy.
`
`Materials and Methods
`
`Materials. RPMI 1640 and glutamine were from Cambrex Bioscience
`(Walkersville, MD). Ultima Flo M scintillation fluid was from Perkin-Elmer
`Life and Analytical Sciences (Wellesley, MA) Gentamicin sulfate was from
`Life Technologies (Gaithersburg, MD). N-Ethylmaleimide and all other
`chemicals were obtained from Sigma (St. Louis, MO). All antibodies used,
`huC242, Tras, and huB4, are humanized IgG1 antibodies. The maytansi-
`q
`noids [3H]DM4, D-[3H]DM4, S-methyl-DM4,
`lysine-N
`-SMCC-DM1, and
`q
`-SPDB-DM4, and the conjugates, huC242-SPDB-[3H]DM4,
`lysine-N
`huC242-SMCC-[3H]DM4, Tras-SMCC-[3H]DM4, and huB4-SPDB-[3H]DM4,
`were prepared at ImmunoGen following published procedures (7, 8).
`Treatment of COLO 205 cells with antibody-drug conjugates. COLO
`
`205 cells (6  106) suspended in 3 mL culture medium containing an
`antibody-drug conjugate at a concentration of 10-7 mol/L of conjugated
`antibody were incubated at 37jC for 3 to 30 hours. The cells and the
`medium were then separated by centrifugation (2,000  g, 5 minutes). The
`supernatant (3 mL) was chilled on ice, mixed with 4 mL ice-cold acetone,
`and kept at 80jC for at least l hour or until further processing. The cells
`
`Cancer Res 2006; 66: (8). April 15, 2006
`
`4426
`
`www.aacrjournals.org
`
`Downloaded from
`
`on June 10, 2014. © 2006 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`IMMUNOGEN 2175, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`were suspended in 3 mL HBBS buffer and sedimented by centrifugation,
`then resuspended in 0.3 mL TBS containing 0.5% bovine serum albumin.
`At
`this point,
`if alkylation was desired for capping of
`free thiols,
`N-ethylmaleimide was added to 7.5 mmol/L, and the solution was mixed
`and kept at room temperature for 30 minutes. The cell suspension was then
`mixed with 0.6 mL ice-cold acetone. The samples were placed at 80jC for
`at least 1 hour or until further processing. Precipitated protein was removed
`by centrifugation at 2,500  g and the supernatants were acidified with
`
`5% acetic acid and evaporated to dryness. The samples were dissolved in
`0.12 mL of 20% aqueous CH3CN containing 0.025% trifluoroacetic acid
`(TFA), aliquots of 0.1 mL were submitted to high-performance liquid
`chromatography (HPLC).
`In vivo studies. The antitumor activity of the conjugates was assessed in
`female CB-17 severe combined immunodeficient mice (SCID mice; Taconic
`Labs, Germantown, NY) bearing HT-29 tumors as described previously (9).
`Conjugates were administered i.v. to groups of six mice.
`Cell cycle studies. Exponentially growing COLO 205 cells were
`
`resuspended at 1  105/mL to 2  105/mL before drug or conjugate
`
`treatment. The nuclei of the cells were stained with propidium iodide as
`previously described (10). DNA content analysis was done using
`FACSCalibur (Becton Dickinson, San Jose, CA). For each sample, 10,000
`events were collected and FL2-A histograms were generated. The cell cycle
`analysis software, ModFit LT 3.1 (Verity Software House, Topsham, ME)
`was used to determine the percentage of cells in different phases of the cell
`cycle.
`Analytic methods. All maytansinoids were separated on an analytic
`
`C-18 column (0.46  25 cm) equilibrated with 20% aqueous CH3CN
`containing 0.025% TFA and developed with a linear gradient of 2% CH3CN/
`min and a flow rate of 1 mL/min. The effluent was directed to a diode array
`detector followed either by a Radiomatic 150 h-counter (Perkin-Elmer Life
`and Analytic Sciences) where the effluent was mixed continuously with
`3 mL scintillation cocktail before it was directed to a 0.5 mL flow cell or
`a Bruker Daltonics Esquire 3000 electrospray mass spectrometer depending
`on the application. The data sampling interval was 6 seconds. The peak
`areas of the chromatograms were converted to picomoles as follows:
`A standard curve to relate the peak of radioactivity (mV2) on the
`chromatograms to cpm of tritium was prepared by injecting 1,000 to
`50,000 cpm of a stock solution of [3H]DM4 (250 mCi/mmol) diluted in 20%
`aqueous CH3CN containing 0.025% TFA. Duplicates for each sample were
`submitted to HPLC as described above.
`
`Results
`In vitro potency and in vivo activity of huC242-maytansinoid
`conjugates. The disulfide-linked antibody-maytansinoid conju-
`gate, huC242-SPDB-DM4, and the thioether-linked conjugate,
`huC242-SMCC-DM1, were first assayed for their cytotoxic potency
`against antigen-positive COLO 205 cells and antigen-negative
`Namalwa cells (both sensitive to maytansine with IC50 values of
`f30 to 60 pmol/L for both cell
`lines) using an 3-(4,5-
`dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)–
`based assay. The conjugates displayed similar potencies with
`IC50 values of 40 pmol/L against COLO 205 cells and 20 to 80
`nmol/L against Namalwa cells upon a 4-day exposure of the cells
`to the conjugates (Fig. 1A). The antitumor activity of the two
`conjugates was assessed in SCID mice bearing s.c. HT-29 or COLO
`205 tumors (both CanAg-positive human colon adenocarcinoma).
`The activity of huC242-SPDB-DM4 at a single dose of 50 Ag/kg
`(concentration based on conjugated DM4) in eradicating HT-29
`tumors was greater
`than that of huC242-SMCC-DM1 when
`administered as five daily injections at a dose of 150 Ag/kg/d
`(Fig. 1B). HuC242-SPDB-DM4 also displayed greater activity than
`huC242-SMCC-DM1 when both were administered as five daily
`injections at a dose of 150 Ag/kg/d in SCID mice bearing COLO
`205 tumors ( five of five tumor free mice at 122 days compared
`
`Intracellular Activation of Antibody-Maytansinoid
`
`Figure 1. Cytotoxicity of huC242-SPDB-DM4 and huC242-SMCC-DM1 and
`their effects on the cell cycle progression in the presence of the lysosomal
`inhibitor BafA1. A, surviving fractions of antigen-positive COLO 205 (closed
`symbols ) and antigen-negative Nalmawa cells (open symbols ) were measured
`using an MTT assay after exposure to different concentrations of huC242-SPDB-
`DM4 (diamonds ) or huC242-SMCC-DM1 (squares ) for 4 days, and plotted
`versus conjugate concentrations. B, antitumor activities of huC242-SPDB-DM4
`and huC242-SMCC-DM1 in SCID mice bearing HT-29 human colon tumor
`xenografts. Doses are based on the amount of conjugated DM4 or DM1.
`Tumor-bearing mice were treated with PBS (closed circles, left and right ), a
`single dose of 50 Ag/kg huC242-SPDB-DM4 (open diamonds), 150 Ag/kg
`huC242-SPDB-DM4 (closed diamonds ), or five daily injections of huC242-
`SMCC-DM1 at a dose of 150 Ag/kg (open squares ). C, FACS histograms
`(FL2-A) display DNA contents of asynchronous, exponentially growing COLO
`205 cells treated for 20 hours with 0.66 Amol/L nocodazole, 10-8 mol/L
`DM1-SMe, 3  10-9 mol/L huC242-SMCC-DM1, or 3  10
`9 mol/L huC242-
`SPDB-DM4 in the presence of 30 nmol/L BafA1 or no treatment.
`
`with two of five tumor free mice at 122 days; data not shown).
`These results show that although both conjugates display similar
`cytotoxicity in vitro, their efficacy against tumors in vivo differs
`significantly.
`Effect of lysosomal inhibitors on cell cycle arrest induced by
`huC242-maytansinoid conjugates. Maytansinoids inhibit tubulin
`polymerization, which leads to cell cycle arrest in the G2-M phase
`(11); therefore, we checked if this activity was also present in both
`conjugates. Samples of asynchronously growing cells were exposed
`to conjugate (3 nmol/L), DM1-SMe (10 nmol/L; see Fig. 5 for
`structure), or nocodazole (0.66 Amol/L), another inhibitor of
`microtubule polymerization, at 37jC for 20 hours. The DNA
`content of the cells was then analyzed by flow cytometry. In
`cultures treated with nocodazole or DM1-SMe, >50% of the cells
`were arrested in the G2-M phase (Fig. 1C, first row), compared with
`only 10% of untreated COLO 205 cells (typically f60% in G1 and
`30% in S). In samples treated with either of the two conjugates,
`f70% to 80% of the cells were arrested at the G2-M phase,
`indicating that the cell cycle effects of maytansinoid conjugates are
`similar to those of the free maytansinoids (Fig. 1C). No cell cycle
`arrest was induced by the conjugates in cells lacking the target
`antigen (data not shown).
`To test whether uptake and processing of conjugates through
`lysosomes was necessary for their activity against cancer cells, we
`examined G2-M arrest induced by conjugates in the presence of
`bafilomycin A1 (BafA1), a lysosomal inhibitor. BafA1 selectively
`
`www.aacrjournals.org
`
`4427
`
`Cancer Res 2006; 66: (8). April 15, 2006
`
`Downloaded from
`
`on June 10, 2014. © 2006 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`IMMUNOGEN 2175, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Cancer Research
`
`inhibits V-ATPase, a proton pump present in endosomes and
`lysosomes, which leads to neutralization of the pH in these vesicles
`(12, 13). The pH neutralization blocks trafficking from late
`endosomes to lysosomes and lysosomal processing, yet modestly
`affects the rate of internalization and recycling and does not
`inhibit trafficking between endosomes and trans-Golgi (14–16). We
`also found that the rate of
`internalization of a fluorescently
`modified huC242 was not affected by BafA1 (data not shown). The
`treatment of COLO 205 cells with BafA1 alone did not significantly
`alter the distribution of cells between the phases of the cell cycle
`(Fig. 1C). However, in the presence of BafA1, G2-M arrest induced
`by either huC242-SMCC-DM1 or huC242-SPDB-DM4 was almost
`completely abolished (6-9% in G2-M; Fig. 1C). In contrast, BafA1
`treatment had a modest effect on the extent of G2-M arrest caused
`by the free maytansinoid, DM1-SMe or by nocodazole (37-39% in
`G2-M; Fig. 1C). Similar results were also found when chloroquine,
`another lysosomal
`inhibitor that neutralizes pH by a different
`mechanism (16, 17), was used (Supplementary Fig. S1). These
`findings were the first indication of the importance of lysosomal
`processing in the activation of both huC242-SMCC-DM1 and
`huC242-SPDB-DM4.
`Isolation of maytansinoid metabolites from huC242-may-
`tansinoid conjugates. To examine the fate of the maytansinoid
`drug upon incubation of target cells with an antibody-maytansi-
`noid conjugate, we prepared conjugates with maytansinoids that
`were 3H-labeled at the C-20 methoxy group (see Fig. 5). Radio-
`labeled maytansinoid conjugates, huC242-SPDB-[3H]DM4 (250
`mCi/mmol) and huC242-SMCC-[3H]DM1 (214 mCi/mmol), exhibit
`in vitro cytotoxicities similar to nonradiolabeled conjugate samples
`(data not shown). Cultures of 2  106 COLO 205 cells were exposed
`7 mol/L 3H-labeled conjugates for periods of 5, 9, and 26
`to 10
`hours, separated into cell and conditioned medium fractions, and
`each sample was extracted with acetone. We determined that
`cells treated under these conditions remained viable with intact
`
`plasma membranes for at least the first 26 hours of exposure as
`determined by trypan blue staining (data not shown). The acetone
`extracts were analyzed for 3H-labeled metabolites by reversed-
`phase HPLC. The chromatograms in Fig. 2A display signals for
`the amount of radioactivity in the acetone-extracted samples from
`the thioether-linked huC242-SMCC-[3H]DM1-treated cells. A con-
`trol was prepared from an acetone extract of the conjugate used
`in the experiment to identify free maytansinoid species present in
`the huC242-SMCC-[3H]DM1 sample before the exposure to cells
`(Fig. 2A, g ). Two new, partially separated peaks of radioactivity
`with retention times of f18.7 and 19.2 minutes, respectively,
`were identified in extracts from cell pellets. These metabolites are
`readily detectable after a 5-hour exposure, and increase by 9 and
`26 hours (Fig. 2A, a, c, e). These same metabolites were also detect-
`able in the culture medium but not until the 9-hour time point, and
`then were readily detectable in the 26-hour sample. In separate
`experiments, the two metabolites were isolated and found by mass
`q
`spectrometry (MS) to be the two isomers of
`lysine-N
`-SMCC-
`[3H]DM1 (R or S configuration at the carbon of the thioether bond
`formed in the conjugation reaction; both peaks M+ = 1,103.5; M +
`Na = 1,125.5).
`The results from an analogous experiment with the disulfide-
`linked huC242-SPDB-[3H]DM4 conjugate are shown in Fig. 2B. Three
`distinct peaks of radioactivity with retention times of 20.5, 26.5, and
`27 minutes were observed in the chromatograms derived from the
`cell pellets treated with huC242-SPDB-[3H]DM4 for 5 and 9 hours
`(Fig. 2B, a and c). Cells treated for 26 hours yielded largely one
`peak with a retention time of 27 minutes (Fig. 2B, e), indicating that
`the metabolites eluting at 20.5 and 26.5 minutes had either been
`converted to the metabolite eluting at 27 minutes or were no longer
`present in the cells. The peak at 26.5 minutes has the same retention
`time as DM4. To further study this metabolite and to assess whether
`any of the other metabolites contained a free sulfhydryl group, an
`acetone extract from a 9-hour exposure was treated with
`
`Figure 2. Maytansinoid metabolites formed upon treatment of COLO 205 cells with huC242-SMCC-[3H]DM1 (A) or huC242-SPDB-[3H]DM4 (B ). Metabolites
`were extracted with acetone from the cell pellet and the spent medium, respectively, and then analyzed by HPLC. The effluent from the column was monitored for tritium
`using an in-line flow scintillation analyzer with an output in mV2; thus, the chromatograms show retention time on the abscissa and mV2 as a measure of [3H]
`on the ordinate. a to f, chromatograms associated with the medium and cell pellets from COLO 205 cells that were treated for 5, 9, and 26 hours with huC242-SMCC-
`[3H]DM1 (A) or huC242-SPDB-[3H]DM4 (B). g (A ) and h (B ), chromatograms obtained from the acetone extract of the huC242-SMCC-[3H]DM1 and huC242-
`SPDB-[3H]DM4 conjugate sample, respectively, used in the experiment. g (B), chromatogram derived from cells that were treated equally to the cell pellet from the
`9-hour incubation (as in c ), except that the sample was additionally exposed to N-ethylmaleimide (NEM ) before chromatography to alkylate any sulfhydryl groups.
`
`Cancer Res 2006; 66: (8). April 15, 2006
`
`4428
`
`www.aacrjournals.org
`
`Downloaded from
`
`on June 10, 2014. © 2006 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`IMMUNOGEN 2175, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Intracellular Activation of Antibody-Maytansinoid
`
`Figure 3. Accumulation of maytansinoid
`metabolites inside and outside of COLO 205 cells
`treated with 3H-maytansinoid conjugates and
`their correlation with mitotic arrest. A, areas
`associated with the peaks of radioactivity for the
`metabolites in Fig. 2 were quantified and converted
`to picomoles as described in Materials and
`Methods. a to c, changes in the amount of various
`metabolites in the samples over a 26-hour
`q
`incubation period. a, accumulation of lysine-N
`-
`SMCC-DM1 in the medium (open circles ) and cells
`(closed circles ) following treatment of COLO 205
`cells with huC242-SMCC-[3H]DM1. b, changes
`in the amounts of maytansinoid metabolites in the
`cell pellet (closed symbols ) and in the medium
`(open symbols ) following treatment of COLO 205
`with huC242-SPDB-[3H]DM4: S -methyl-DM4 +
`q
`DM4 (squares ), lysine-N
`-SPDB-DM4 (circles ),
`S-cysteinyl-DM4 (diamonds ). c, change of the
`sum of all metabolites present for both conjugates,
`huC242-SMCC-[3H]DM1 (circles) and huC242-
`SPDB-[3H]DM4 (squares ), either in the cell pellet
`(solid symbols ) or in the cell pellet and medium
`together (open symbols ). B, COLO 205 cells were
`synchronized in S phase with a 24 hours of
`treatment of 2 Ag/mL aphidicolin. The cells were
`released from S phase by the removal of
`8 mol/L
`aphidicolin and incubated with 10
`DM1-SMe, 3  10
`9 mol/L huC242-SMCC-DM1,
`3  10
`9 mol/L huC242-SPDB-DM4, or left
`untreated. FACS analysis was done at 5, 10, and
`18 hours after aphidicolin release.
`
`N-ethylmaleimide and then subjected to HPLC (Fig. 2B, g ). Following
`alkylation, the peak at 26.5 minutes, observed in the 5- and 9-hour
`samples, was replaced with a new peak at 25 minutes. This new peak
`coelutes with a standard of the purified DM4-N-ethylmaleimide
`reaction product (data not shown), suggesting that the metabolite at
`26.5 minutes is indeed DM4.
`The corresponding chromatograms associated with the
`conditioned medium from treated cells are displayed in
`Fig. 2B (b, d , and f ). After 5 hours of exposure, no metabolites
`can be detected; the peaks present are also visible in the control
`chromatogram of the acetone extract of the conjugate sample
`(Fig. 2B, h). After 9 hours of exposure, however, the unknown
`metabolite with a retention time of 20.5 minutes that was
`observed in cells after 5 hours can be detected in the medium,
`and after 26 hours, two additional metabolites with retention
`times of 18.5 and 27 minutes were present. The unique
`metabolite in the medium that elutes at 18.5 minutes was
`found to have the same retention time as the mixed disulfide
`between DM4 and cysteine (S-cysteinyl-DM4; data not shown).
`RPMI 1640 contains 0.21 mmol/L cystine, and separate experi-
`ments have shown that DM4 under these conditions reacts
`rapidly (t 1/2 < 1 hour) to form S-cysteinyl-DM4 through a thiol-
`disulfide interchange reaction (data not
`shown). Thus,
`the
`S-cysteinyl-DM4 disulfide compound would be expected to form
`in the medium if the DM4 observed in the cell pellet leaves the
`cells. To further test whether any of the metabolites in the
`medium were disulfide compounds, a cell supernatant sample
`from a 26-hour exposure was divided into two equal portions of
`which one was subjected directly to chromatography, and the
`other portion was
`treated with DTT and selenol before
`chromatography to reduce all disulfide bonds (18). Following
`reduction, all of the peaks of radioactivity in the chromatograms
`disappeared except the peak eluting at 27 minutes and a single
`
`new peak appeared with a retention time of 26.5 minutes, which
`is that of DM4 (Supplementary Fig. S2). Thus, the metabolites
`eluting at 18.5 and 20.5 minutes, but not the metabolite at 27
`minutes, are DM4 species containing a disulfide-linked substit-
`uent. In separate experiments, the unknown metabolites eluting
`at 20.5 and 27 minutes were found to have a mass consistent
`q
`with lysine-N
`-SPDB-DM4 (M + Na = 1048.4) and S-methyl-DM4
`(M + Na = 816.4/M + K = 832.5), respectively.
`In the above experiments, 2  106 COLO 205 cells were incubated
`7 mol/L conjugate. From this large
`in 3 mL medium containing 10
`amount of conjugate, <20% of the conjugate-bound maytansinoid
`was recovered in the identified maytansinoid metabolites after
`incubation with the cells for 26 hours (Fig. 3A, c; dotted line
`represents 20% level). In a separate experiment following the fate
`of huC242-SPDB-[3H]DM4 that was bound to cells at 4jC, we
`determined that within a 22-hour period at 37jC, 73% of the
`conjugated DM4 bound to COLO 205 cells was converted into
`the four maytansinoid metabolites—DM4, S-cysteinyl-DM4, lysine-
`q
`N
`-SPDB-DM4, and S-methyl-DM4 (Supplementary Fig. S3). We
`conclude that these metabolites represent the majority (if not all)
`of the metabolites formed in the cellular activation process.
`Moreover, production of these maytansinoid metabolites was
`not observed when COLO 205 cells were incubated with disulfide-
`linked or SMCC-linked maytansinoid conjugates, which do not bind
`target antigens on these cells (Supplementary Fig. S4).
`The kinetics for the accumulation of metabolites and cell
`cycle arrest. The peak areas of radioactivity associated with the
`metabolites generated from huC242-SMCC-[3H]DM1 and from
`huC242-SPDB-[3H]DM4 (Fig. 2) were determined and converted
`to picomoles of maytansinoid as described in Materials and
`Methods. The results are shown in Fig. 3A as accumulation of
`the maytansinoid metabolites over time from the addition of
`the conjugates to the cells (T = 0). Figure 3A (a) shows the
`
`www.aacrjournals.org
`
`4429
`
`Cancer Res 2006; 66: (8). April 15, 2006
`
`Downloaded from
`
`on June 10, 2014. © 2006 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`IMMUNOGEN 2175, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Cancer Research
`
`q
`-SMCC-DM1, the single metabolite from
`accumulation of lysine-N
`huC242-SMCC-[3H]DM1, in the cell pellet and in the medium. The
`concentration of the metabolite in the cells reaches a steady state
`after 9 hours of incubation, which may be due to the saturation of
`an intracellular binding site. This maytansinoid metabolite begins
`to appear in the medium at 9 hours, suggesting efficient efflux of
`q
`the charged lysine-N
`-SMCC-DM1 species across the plasma
`membrane. Figure 3A (b) shows the accumulation of the three
`stable metabolites generated from huC242-SPDB-[3H]DM4, in the
`cell pellet and in the medium. The intracellular amount of
`q
`the disulfide compound, lysine-N
`-SPDB-DM4, declines over the
`observed period of 26 hours, presumably through disulfide cleavage
`to DM4 in the intracellular reducing environment, followed by
`subsequent conversion of the resulting free thiol, DM4, to the
`stable S-methyl-DM4 derivative. Very little, if any, DM4 was observed
`inside the cells after 9 hours of
`incubation,
`indicating rapid
`methylation probably catalyzed by a methyl transferase. The total
`intracellular accumulation of metabolites reaches a steady state
`after 9 hours of exposure to either conjugate and the levels of
`metabolites at the steady state are about the same for both
`conjugates (Fig. 3A, c), suggesting that the conjugates share the
`same rate-limiting step with respect to production of
`the
`metabolites and that these metabolites likely bind to the same
`intracellular target.
`Because only f9 hours is required for intracellular accumu-
`lation of metabolites to reach the steady-state level, we
`investigated whether G2-M arrest induced by either conjugate
`can occur within that time frame. COLO 205 cells were first
`synchronized with a 24-hour treatment of aphidicolin, a reversible
`DNA replication inhibitor that blocks cells in S phase (19).
`Because the majority of the asynchronous population of COLO
`205 cells were in G1 phase, most of the aphidicolin-treated cells
`arrested in the beginning of S phase, which cannot be
`distinguished from G1 phase in the fluorescence-activated cell
`sorting (FACS) analysis (Fig. 3B, top picture). The synchronized
`cells were then released from the arrest by the removal of
`aphidicolin, and either left untreated, or treated with DM1-SMe
`or with either conjugate. Untreated cells immediately continue
`with DNA synthesis, and the majority is in G2-M phase (59%)
`after 5 hours (Fig. 3B), and has undergone mitosis at 10 hours
`and are in G1 phase (76%). In contrast, the majority of cells
`treated with DM1-SMe or either conjugate are in G2-M phase
`(75% to 85%) after 10 and 18 hours,
`indicating that sufficient
`amounts of active drug species have accumulated intracellularly
`
`to induce mitotic arrest when the aphidicolin-released cells
`reached mitosis. Therefore, COLO 205 cells require as little as 5
`hours, but not more than 10 hours to produce enough of active
`drug metabolites to cause cell cycle arrest. Thus, the kinetics of
`G2-M phase arrest induced by these conjugates correlate with the
`time required to reach the steady-state level of
`intracellular
`accumulation of the drug metabolites.
`Activity of maytansinoid metabolites. The two major
`q
`metabolites, S-methyl-DM4 and lysine-N
`-SMCC-DM1, were syn-
`thesized and tested for their in vitro cytotoxicity against COLO 205
`cells and Namalwa cells. S-methyl-DM4 was highly cytotoxic with
`q
`an IC50 value of 2 pmol/L against both cell lines, whereas lysine-N
`-
`SMCC-DM1 was f105-fold less potent against both cell lines with
`an IC50 value of 0.1 Amol/L (data not shown). This difference may
`be explained by the different charge status of the compounds. The
`charged lysine derivatives are expected to penetrate cell mem-
`branes very inefficiently such that high external concentrations are
`needed to reach the required toxic intracellular concentrations,
`whereas penetration of cell membranes is efficient for the neutral
`lipophilic S-methyl-DM4 compound.
`q
`Accumulation of the lysine-N
`-SMCC-DM1 metabolite from the
`noncleavable conjugate is coincident with the observed forma-
`q
`-SPDB-DM4
`tion of the potent S-methyl-DM4, DM4, and lysine-N
`q
`from the cleavable conjugate. This suggests that the lysine-N
`-
`SMCC-DM1 metabolite is as potent as the metabolites from the
`cleavable conjugate when delivered intracellularly and that all of
`the maytansinoid metabolites are active when produced in the
`cell. The cell cycle arrest of both the cleavable and noncleavable
`conjugates was abrogated in the presence of
`the lysosomal
`q
`inhibitor BafA1 (Fig. 1C). If the lysine-N
`-SMCC-DM1 metabolite
`and the other metabolites observed from the cleavable conjugate
`arrest cells, BafA1 should prevent their formation. To investi-
`gate this possibility, COLO 205 cells were incubated at 37jC for
`7 mol/L huC242-SMCC-[3H]DM1 or huC242-
`22 hours with 10
`SPDB-[3H]DM4 conjugate in the presence or in the absence of
`300 nmol/L BafA1. We found that BafA1 abolished the formation
`q
`of both the lysine-N
`-SMCC-DM1 from the thioether-linked
`conjugate, and the S-methyl-DM4 from the disulfide linked
`conjugate (Fig. 4A). In addition, no metabolites were detected in
`the medium of cells treated with either conjugate in the
`presence of BafA1 (data not shown). These results suggest that
`the lysine metabolites are toxic when delivered intracellularly
`and that lysosomal processing is necessary for the production of
`all the observed metabolites.
`
`Figure 4. The role of the major
`metabolites in cellular toxicity. A, influence
`of BafA1 on the cellular metabolism of
`huC242-SMCC-[3H]DM1 and huC242-
`SPDB-[3H]DM4 in the presence or absence
`of BafA1. The chromatograms are from
`acetone extracts of COLO 205 cells treated
`with huC242-SMCC-[3H]DM1 or huC242-
`SPDB-[3H]DM4 for 22 hours in the
`presence of BafA1 or no treatment.
`B, accumulation of maytansinoid
`metabolites formed upon treatment of
`COLO 205 cells with huC242-SPDB-D-
`[3H]DM4 versus huC242-SPDB-[3H]DM4.
`Total amounts of maytansinoid metabolites
`generated from huC242-SPDB-D-[3H]DM4
`(closed squares ) and from huC242-SPDB-
`[3H]DM4 (closed circles ). Th

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket