throbber
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/17/20/6448/1999115/6448.pdf by guest on 10 February 2023
`
`CCR FOCUS
`
`SAR3419: An Anti-CD19-Maytansinoid Immunoconjugate for
`the Treatment of B-Cell Malignancies
`
`Veronique Blanc1, Anne Bousseau1, Anne Caron1, Chantal Carrez1, Robert J. Lutz2, and John M. Lambert2
`
`Abstract
`
`SAR3419 is a novel anti-CD19 humanized monoclonal antibody conjugated to a maytansine derivate
`through a cleavable linker for the treatment of B-cell malignancies. SAR3419 combines the strengths of a
`high-potency tubulin inhibitor and the exquisite B-cell selectivity of an anti-CD19 antibody. The inter-
`nalization and processing of SAR3419, following its binding at the surface of CD19-positive human
`lymphoma cell lines and xenograft models, release active metabolites that trigger cell-cycle arrest and
`apoptosis, leading to cell death and tumor regression. SAR3419 has also been shown to be active in different
`lymphoma xenograft models, including aggressive diffuse large B-cell lymphoma, resulting in complete
`regressions and tumor-free survival. In these models, the activity of SAR3419 compared favorably with
`rituximab and lymphoma standard of care chemotherapy. Two phase I trials with 2 different schedules of
`SAR3419 as a single agent were conducted in refractory/relapsed B-cell non-Hodgkin lymphoma. Activity
`was reported in both schedules, in heavily pretreated patients of both follicular and diffuse large B-cell
`lymphoma subtypes, with a notable lack of significant hematological toxicity, validating SAR3419 as an
`effective antibody-drug conjugate and opening opportunities in the future. Numerous B-cell–specific anti-
`CD19 biologics are available to treat B-cell non-Hodgkin lymphoma, and early phase I results obtained with
`SAR3419 suggest that it is a promising candidate for further development in this disease. In addition, thanks
`to the broad expression of CD19, SAR3419 may provide treatment options for B-cell leukemias that are often
`CD20-negative. Clin Cancer Res; 17(20); 6448–58. Ó2011 AACR.
`
`Introduction
`
`Non-Hodgkin lymphoma (NHL) is the most common
`lymphoma, with an estimated 65,540 new cases and 20,210
`deaths registered in the United States in 2010 (1). NHL
`originates from the malignant development of B or T
`lymphocytes and comprises a heterogeneous group of
`malignancies (2), among which B-cell lymphomas account
`for 85% of all cases (3). Despite the significant improve-
`ment in overall survival of patients with both aggressive and
`indolent B-cell NHL that has been achieved following the
`addition of rituximab to conventional chemotherapy (4),
`not all patients who have CD20-positive tumors respond to
`the treatment (5, 6), and most patients who have disease
`response will relapse (7). More than 50% of patients with
`NHL will die of their disease (4–8). Since 2005, 5 new drugs
`have been approved for the treatment of lymphoma (6, 9),
`including 2 for B-cell NHL, namely, bortezomib, a protea-
`some inhibitor approved for the treatment of mantle cell
`
`Authors' Affiliations: 1Oncology Business Division, Sanofi, Vitry sur
`Seine, France; 2ImmunoGen, Inc., Waltham, Massachusetts
`Corresponding Author: Veronique Blanc, Sanofi, 13 quai Jules Guesde,
`Vitry sur Seine, 94403 France. E-mail:
`Veronique.Blanc2@sanofi-aventis.com
`
`doi: 10.1158/1078-0432.CCR-11-0485
`Ó2011 American Association for Cancer Research.
`
`lymphoma in 2006, and bendamustine, a DNA alkylating
`and antimetabolite agent approved for the treatment of
`indolent B-cell NHL in 2008. However, despite the large
`number of trials of novel agents, including monoclonal
`antibodies and small-molecule inhibitors (3, 10), there is
`still an important unmet medical need, particularly in
`second-line and subsequent lines of therapy. Concerning
`monoclonal antibodies, different modalities of conjugation
`have been tested in the clinic and have shown responses in
`hematological malignancies and NHL in particular, as dis-
`cussed in this CCR Focus section, dedicated to antibody
`conjugates (11). These modalities include radionuclide
`conjugation (12), toxin conjugation (13), and small-mol-
`ecule drug conjugation, also called antibody-drug conju-
`gates (ADC; refs. 14–16). ADCs represent the most active
`field today, with at least 20 such ADCs in development (11).
`Very promising clinical responses have been achieved in
`both solid tumors, as shown by T-DM1 for the treatment of
`HER2-positive breast
`tumors (14), and hematological
`malignancies, as shown by SGN35 (15) and CMC-544
`(16) for Hodgkin lymphoma and NHL, respectively.
`
`Monoclonal Antibodies beyond Rituximab for
`Treating B-Cell Malignancies
`
`Since the introduction of rituximab in 1997, only 2 more
`"naked" (nonconjugated) antibodies have been approved
`for the treatment of lymphoma: (i) alemtuzumab, an
`
`6448
`
`Clin Cancer Res; 17(20) October 15, 2011
`
`UPenn Ex. 2073
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Anti-CD19-Maytansinoid Conjugate
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/17/20/6448/1999115/6448.pdf by guest on 10 February 2023
`
`anti-CD52 antibody approved in 2001 for the treatment of
`CLL (17); and (ii) ofatumumab, a second-generation anti-
`CD20 antibody displaying increased complement-depen-
`dent cytotoxicity compared with rituximab,
`that was
`approved in 2009 for relapse/refractory CLL patients who
`failed fludarabine and alemtuzumab (18). In addition to
`these 2 naked antibodies, 2 murine anti-CD20 radionuclide
`conjugates were approved for consolidation treatment in
`2002 (90Y-ibritumomab) and 2003 (131I-tositumomab;
`refs. 12 and 19).
`In recent years, investigators have made a considerable
`effort to further improve NHL treatment, and several sec-
`ond- and third-generation anti-CD20 monoclonal antibo-
`dies have been examined in clinical trials (20). In addition,
`exploration of other B-cell–specific antigens,
`including
`CD19, CD22, CD37, and CD40, as potential targets for
`the development of new antibody-based treatments is grow-
`ing. Antibodies are being investigated as naked molecules,
`such as the anti-CD22 epratuzumab (21), the anti-CD40
`dacetuzumab (22), and the different anti-CD19 antibodies
`(Table 1), or in an engineered format, such as the anti-
`CD19/anti-CD3–bispecific blinatumomab (23) and the
`anti-CD37–specific small modular immunopharmaceuti-
`cal TRU-016 (24). The advent of ADC technologies has also
`led to clinical evaluation of conjugated antibodies, such as
`
`the anti-CD22-calicheamicin conjugate CMC544 (16) and
`the subject of this review, the anti-CD19-maytansinoid
`conjugate SAR3419 (25–28).
`
`CD19 Antigen
`
`CD19 is a type I transmembrane glycoprotein of the
`immunoglobulin Ig superfamily, with expression restricted
`to B cells (29). CD19 is involved in B-cell fate and differ-
`entiation through the modulation of B-cell receptor signal-
`ing at multiple stages of B-cell development (29, 30). CD19
`is ubiquitously expressed on B cells (25), as it is found
`expressed from the early pre-B stage throughout B-cell
`differentiation up to mature B cells, before it is downmo-
`dulated at the plasma cell stage (Fig. 1). Thus, CD19 has
`broader expression than CD20. The pattern of CD19 expres-
`sion is maintained in B-cell malignancies, covering all
`subtypes of B-cell lymphoma, from indolent to aggressive
`forms, as well as B-cell chronic lymphocytic leukemia and
`non-T acute lymphoblastic leukemia (31–33), and allows
`the targeting of tumor indications of early B cells, such as
`acute lymphoblastic leukemia (ALL), which cannot be
`targeted by rituximab. The quantification of the number of
`CD19 molecules at the surface of tumor cells and cell lines
`derived from malignant B cells resulted in a wide range of
`
`Table 1. Clinical trials and discovery projects targeting CD19
`
`Biologic compound
`
`Type
`
`Mechanism of action
`
`Phase (initiation date)
`
`MT-103 (blinatumomab)
`Micromet, Inc.
`SAR3419
`Sanofi-Aventis/ImmunoGen,
`Inc.
`MEDI-551 MedImmune/
`Astra-Zeneca
`MOR-208/XmAb5574
`Xencor/Morphosys
`MDX-1342 Medarex/
`Bristol-Myers Squibb
`Combotox
`University of Texas
`Southwestern/Abiogen
`
`DI-B4
`Merck KGaA/Cancer
`Research UK
`SGN-19A
`Seattle Genetics
`MDX-1206 Medarex/
`Bristol-Myers Squibb
`
`AFM-11
`Affimed Therapeutics AG
`AFM-12
`Affimed Therapeutics AG
`
`Bispecific scFv anti-CD19/anti-CD3
`BiTE
`Humanized anti-CD19 mAb
`conjugated to maytansinoid DM4
`
`Glycoengineered humanized anti-CD19
`mAb (BioWa's Potelligent)
`Fc engineered humanized
`anti-CD19 mAb
`Glycoengineered fully human
`anti-CD19 mAb (BioWa's Potelligent)
`Mixture of chimeric anti-CD19 mAb
`HD37 and anti-CD22 mAb RFB4,
`both conjugated to deglycosylated
`ricin A-chain (HD37-dgA þ RFB4-dgA)
`Chimeric anti-CD19 mAb monoclonal
`antibody
`
`Fully human anti-CD19 mAb (hBU12)
`conjugated to auristatin (vc-MMAE)
`Fully human anti-CD19 mAb (MDX1435)
`conjugated to duocarmycin
`(vc-MGBAA)
`Tetravalent tandem antibody (TandAb)
`anti-CD19/anti-CD3
`Tetravalent tandem antibody (TandAb)
`anti-CD19/anti-CD16
`
`T-cell recruitment and
`activation
`ADC (tubulin binder)
`
`I/II/III (2007) Pivotal

`ALL
`trial for MDR
`I/II (2007)
`
`Naked antibody high-affinity
`FcgRIII-enhanced ADCC
`Naked antibody high-affinity
`FcgRIII-enhanced ADCC
`Naked antibody high-affinity
`FcgRIII-enhanced ADCC
`Immunotoxin conjugate with
`deglycosylated ricin
`A-chain
`
`I (2010)
`
`I (2010)
`
`I (2008, on hold)
`
`I (2005)
`
`Naked antibody ADCC
`
`I (2010)
`
`ADC (tubulin binder)
`
`Discovery
`
`ADC (DNA alkylating agent)
`
`Discovery
`
`T-cell recruitment
`
`Discovery
`
`NK cell recruitment
`
`Discovery
`
`www.aacrjournals.org
`
`Clin Cancer Res; 17(20) October 15, 2011
`
`6449
`
`UPenn Ex. 2073
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`CCR FOCUS
`
`Figure 1. Pattern of expression of
`CD19 and CD20 antigens during B-
`cell development and associated
`malignancies. A simplified cartoon
`of B-cell lineage, B-cell
`malignancies, and antigen
`expression (59, 60). The positioning
`of the different B-cell malignancies
`associated with different stages of
`B-cell development is abridged and
`illustrative only; a detailed
`description is beyond the scope of
`this review.
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/17/20/6448/1999115/6448.pdf by guest on 10 February 2023
`
`published values due to differences in cell line clones, the
`antibodies studied, and the methodologies used. For exam-
`ple, CD19 expression was reported to be both >100,000
`sites per cell (34) and 10,000 sites per cell (35) for the
`same cellular models. In our hands, using a calibration
`curve of beads loaded with different amounts of human
`IgG1, the quantification of CD19 antigen at the surface of
`Ramos, Raji, and Daudi lymphoma cell lines gave average
`values of 42,166, 18,315, and 14,077 antibody-binding
`sites per cell, respectively. With the same assay, the quan-
`tification of CD19 on the surface of normal B lymphocytes
`yielded values in the range of 11,000 to 16,000 antibody-
`binding sites per cell, an order of magnitude similar to that
`of the lymphoma cell lines tested.
`CD19 was shown to be internalized efficiently in lym-
`phoma tumor models with the use of different antibodies,
`such as huB4 (35) and hBU12 (36). In our laboratories, we
`quantified the internalization and processing of the anti-
`CD19 antibody huB4 using an Alexa488 antibody conju-
`
`gate and different tumor cell lines upon incubation at 37
`C.
`The kinetics of appearance of antibody-free fluorescence,
`produced by lysosomal degradation of the antibody moiety,
`showed internalization and processing of
`the huB4-
`Alexa488 in all models, with some variability between
`models (35) that may reflect differences in the capacity of
`CD19 to internalize in different cellular contexts. Indeed,
`the level of CD21 coreceptor, expressed in a subset of
`lymphomas, was described to affect the internalization and
`efficacy of an anti-CD19-maytansinoid conjugate through a
`noncleavable linker (37). However, this observation was
`not confirmed in a second study using auristatin derivatives
`conjugated to a different anti-CD19 antibody through a
`cleavable linker (36).
`
`Anti-CD19 Intervention
`
`CD19’s B-cell lineage–restricted expression and moderate
`to high homogeneous expression in most cases of lympho-
`
`ma make it an attractive target for therapies for B-cell
`malignancies, as shown by the many previous and ongoing
`therapeutic interventions that focus on CD19 (Table 1).
`Different types of molecules targeting CD19 are being
`developed that can broadly fit into 3 main classes: (i) naked
`antibodies, most of which undergo modifications of their Fc
`portion to enhance binding to FcRgIII and subsequently
`enhance antibody-dependent cell-mediated cytotoxicity
`(ADCC) activity, such as MOR-208 and MEDI-551; (ii)
`bispecific antibodies with one arm binding to CD19 and
`one arm binding to either the T-cell receptor (as exemplified
`by blinatumomab, the most advanced anti-CD19 therapeu-
`tic molecule) or NK-cell receptors (as in the molecule
`AFM12, a tetravalent tandem antibody that is in the dis-
`covery stage of development); and (iii) antibody conju-
`gates, for which anti-CD19 antibodies are conjugated to
`either a toxin (as exemplified by the ricin-based immuno-
`toxin Combotox) or a potent low-molecular-weight cyto-
`toxic molecule (as exemplified by SAR3419). As of today,
`Combotox has shown anticancer activity in NHL (38) and
`ALL (39) that may warrant further clinical testing. Studies of
`blinatumomab (23) and SAR3419 (27) have provided a
`clinical proof of concept, showing a notable rate of objective
`response in pretreated patients in phase I clinical trials.
`
`SAR3419 Structure and Mechanism of Action
`
`SAR3419 is an ADC that consists of a humanized mono-
`clonal IgG1 antibody (huB4) attached to a highly potent
`tubulin inhibitor, the maytansinoid DM4 (40), through
`reaction with an optimized cleavable linker, N-succini-
`midyl-4-(2-pyridyldithio)butyrate (SPDB linker; Fig. 2).
`The succinimidyl group of the linker reacts with amino
`groups of lysine residues of the antibody to form stable
`amide bonds, and the pyridyldithio moiety reacts with
`the sulfhydryl group of DM4 to form a hindered disulfide
`bond between the linker and DM4. The disulfide bond
`can be cleaved inside target cells by thiol-disulfide
`
`6450
`
`Clin Cancer Res; 17(20) October 15, 2011
`
`Clinical Cancer Research
`
`UPenn Ex. 2073
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Anti-CD19-Maytansinoid Conjugate
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/17/20/6448/1999115/6448.pdf by guest on 10 February 2023
`
`Figure 2. Structure of SAR3419. Figure is adapted from Al-Katib et al. (43).
`
`exchange reactions to release fully active DM4 (28).
`SAR3419 contains an average of 3.5 DM4 molecules
`per molecule of antibody.
`The murine B4 antibody, one of the earliest antibodies to
`define the CD19 antigen (31), was humanized via a vari-
`able-domain resurfacing method (41) to yield huB4. The
`huB4 antibody displays a subnanomolar affinity for CD19
`on the surface of B cells, and this affinity is conserved
`following conjugation to create SAR3419 (35). The huB4
`antibody was shown to induce ADCC (data not shown) but
`not complement-dependent cytotoxicity as described for
`other anti-CD19 antibodies (42). The ADCC activity was
`conserved following conjugation to the maytansinoid. The
`naked antibody has no direct antiproliferative or proapop-
`totic activity. Therefore, huB4 was not found to be active in
`vivo as a single agent in severe combined immunodeficient
`(SCID) mice bearing several different lymphomas (43). The
`huB4 antibody binds only to the human CD19 antigen
`(29); it does not recognize CD19 in rodent or cynomolgus
`monkey toxicology models, and thus no B-cell depletion
`studies have been done with either the huB4 antibody or
`SAR3419 in animal models.
`Maytansine and other maytansinoids are antimitotic
`agents that bind to tubulin, inhibiting microtubule assem-
`bly and inducing G2/M arrest in the cell cycle, which
`subsequently leads to cell death (44). Maytansinoids are
`very potent, displaying cytotoxic activity in the 10 to 90
`pmol/L range across several tumor cell lines, including
`lymphoma lines (45). The SPDB linker was selected based
`on its superior activity in vivo compared with SPP (another
`cleavable linker with a more labile disulfide bond) and the
`noncleavable SMCC linker in studies analogous to those
`described by Kellogg and colleagues (46) for another anti-
`body-maytansinoid conjugate.
`SAR3419 was shown to display potent in vitro cytotoxicity
`after 5 days of exposure to different CD19-positive lym-
`phoma cell lines, with EC50 values ranging from subnano-
`molar to a few nanomolars (35). SAR3419 induced a rapid
`
`cell-cycle arrest at the G2/M phase (within the first 24 hours
`of exposure) followed by an increase in apoptotic cells from
`24 to 48 hours (35), as expected for delivery of the may-
`tansinoid payload into CD19þ cells. As has been shown in
`the case of other maytansinoid conjugates (47) and in
`experiments with SAR3419 in which the DM4 moiety was
`radiolabeled (48), the mechanism of action of SAR3419
`involves binding to the cell-surface antigen, followed by
`internalization via endocytosis and subsequent intracellu-
`lar routing to lysosomes where the huB4 antibody moiety of
`the ADC is degraded to yield the lysine-SPDB-DM4 metab-
`olite. Once it is in the reducing environment of the cyto-
`plasm, this metabolite is further subjected to cleavage by
`thiol-disulfide exchange reactions to release the free may-
`tansinoid thiol compound DM4 (47, 48). DM4 is then
`S-methylated to form S-methyl-DM4 by an endogenous
`S-methyl-transferase (47). The final metabolite of cancer
`cell metabolism of SAR3419, S-methyl-DM4, has a high
`potency as a microtubule poison that was shown to be
`similar to the potency of maytansine itself when compared
`in cytotoxicity assays in vitro on different tumor cell lines
`(40, 44, 49). The early metabolite formed upon lysosomal
`processing of the ADC, lysine-SPDB-DM4, shows a 1,000-
`fold lower activity on cell lines in vitro resulting from the
`negative effect of the charged lysine residue on its ability to
`diffuse across the plasma membrane into cells (47). How-
`ever, such lysine-linker-maytansinoid species are potent
`microtubule poisons when they are released inside cells
`following uptake and lysosomal processing of an ADC
`(28, 47).
`The mechanism of action of SAR3419 was also shown in
`vivo in immunodeficient SCID mice bearing human Ramos
`Burkitt’s lymphoma, a model that was shown to be highly
`sensitive to SAR3419 (35). In recent studies done in our
`laboratories, SAR3419 was administered at a single dose of
`20 mg/kg 14 days after tumor subcutaneous implantation.
`Tumor sampling was done at different times, and immu-
`nohistochemical
`evaluation of phospho-histone H3
`
`www.aacrjournals.org
`
`Clin Cancer Res; 17(20) October 15, 2011
`
`6451
`
`UPenn Ex. 2073
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/17/20/6448/1999115/6448.pdf by guest on 10 February 2023
`
`CCR FOCUS
`
`(p-histone H3, a mitosis biomarker) and cleaved caspase 3
`(a proapoptotic marker) was done. The single-dose treat-
`ment with SAR3419 induced a significant increase in stain-
`ing for p-histone H3, revealing a mitosis blockade that was
`apparent 24 hours after administration (Fig. 3). This mitosis
`blockade was then followed by tumor cell apoptosis as
`evidenced 48 hours after dosing with the ADC by a marked
`increase in cells that stained positively for cleaved caspase 3
`(Fig. 3).
`Studies were also done in the same Ramos xenograft
`model to assess whether the pattern of metabolic processing
`observed in cell lines was the same as that seen in xenograft
`models. Mice bearing the tumor xenografts were injected
`with a single administration of radiolabeled [3H]-SAR3419
`(conjugate made with tritiated DM4) at 15 mg/kg in experi-
`ments similar to previous experiments involving another
`ADC with the same linker-maytansinoid design (49).
`Tumor sampling was done at different time points, and all
`metabolites were quantified. The total amount of low-
`molecular-weight (i.e., not protein-linked) metabolites
`increased rapidly from 2 to 8 hours, with a subsequent
`linear progression from 8 to 48 hours, and an overall 6-fold
`accumulation from 2 to 48 hours (Fig. 4). At the latter time
`point, 71% of the total radioactivity in the tumor xenograft
`corresponded to low-molecular-weight metabolites, in con-
`trast to just 30% at 2 hours (data not shown). The proces-
`sing of radiolabeled SAR3419 in vivo was found to be similar
`to the in vitro findings (Fig. 4), with the sequential formation
`of lysine-SPDB-DM4 (the major species at 2 hours) fol-
`lowed by DM4 (about 10% of total nonproteic metabolites
`at 2 hours, rising to 24% at 8 hours) and then by the
`appearance of S-methyl-DM4 (first detected at 8 hours). As
`observed in vitro, similar quantities of lysine-SPDB-DM4
`and S-methyl-DM4 were found in the tumors after 48 hours
`
`(Fig. 4; ref. 49). The time course of the formation of the
`active maytansinoid metabolites in the tumor is consistent
`with the observed high induction of p-histone H3 and
`cleaved caspase 3 at 24 and 48 hours, respectively (Fig. 3).
`
`SAR3419 Preclinical Activities in Different
`Lymphoma Models
`
`SAR3419 has shown in vivo efficacy in different lympho-
`ma models, including Burkitt’s lymphomas (Namalwa,
`Ramos, and Raji) and diffuse large B-cell
`lymphoma
`[DLBCL (RL, WSU-DLCL2, and WSU-FSCCL)] implanted
`in SCID mice (35, 43). In all models, SAR3419 showed a
`high level of activity leading to complete responses, with
`mice being tumor-free at study termination at the highest
`doses and with significant tumor growth delay at the lower
`doses (both when injected as a single dose and in a multiple-
`dose schedule).
`As an illustration, in the Ramos lymphoma xenograft
`model (35), when treated by the intravenous route (2 doses
`with 4-day interval), SAR3419 was highly active at the
`dosage of 15 mg/kg (a well-tolerated dosage as evidenced
`by no loss in body weight), and 100% of the mice were
`tumor-free at the end of the study (day 124). The lower
`doses of 7.5 and 3.3 mg/kg were also highly active, with
`complete tumor regressions in 100% of the animals and
`with 5 out of 7 mice being tumor-free at the dose of 7.5 mg/
`kg on day 124. In comparison, treatment with unconjugat-
`ed DM4 at a dose equivalent to the 15 mg/kg dose of
`SAR3419 (0.35 mg/kg of DM4, 2 doses with 4-day interval)
`showed no significant antitumor activity, indicating that
`conjugation of the DM4 to the antibody was critical for
`delivering sufficient maytansinoid to the cancer cells in vivo
`to induce tumor shrinkage.
`
`pHistone H3
`C-Caspase 3
`
`48
`Time (hours)
`© 2011 American Association for Cancer Research
`
`72
`
`96
`
`6
`
`24
`
`45
`
`40
`
`35
`
`30
`
`25
`
`20
`
`15
`
`10
`
`05
`
`Percentage of positive tumor cells area
`
`Figure 3. Kinetics of
`pharmacodynamic markers
`p-histone H3 and cleaved
`caspase 3 (C-Caspase 3) in
`Ramos xenograft model.
`Quantitative evaluation
`of p-Histone H3 and C-Caspase 3
`by image analysis quantification
`at 6, 24, 48, 72, and 96 hours after
`administration of a single
`intravenous dose of 20 mg/kg of
`SAR3419 in SCID mice bearing
`established Ramos
`subcutaneous xenograft tumors.
`
`6452
`
`Clin Cancer Res; 17(20) October 15, 2011
`
`Clinical Cancer Research
`
`UPenn Ex. 2073
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Anti-CD19-Maytansinoid Conjugate
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/17/20/6448/1999115/6448.pdf by guest on 10 February 2023
`
`300
`
`Total nonproteic metabolites
`
`DM4-SPDB-lys
`
`DM4
`
`200
`
`S-methyl-DM4
`
`
`
`100
`
`0
`
`0
`
`8
`
`16
`
`24
`Time (hours)
`
`32
`
`40
`
`48
`
`© 2011 American Association for Cancer Research
`
`Tumor concentration (ng Eq/g)
`
`Figure 4. Kinetics of the
`production of nonproteic
`maytansinoid metabolites in the
`Ramos xenograft model.
`Quantitative evaluation of
`nonproteic maytansinoid
`metabolites (metabolites not linked
`to proteins) in nanograms per gram
`of tumor (ng Eq/g) at 2, 6, 8, 24, and
`48 hours after administration of a
`single intravenous dose of 15
`mg/kg of [3H]-SAR3419 in SCID
`mice bearing established Ramos
`xenograft tumors.
`
`The specificity of tumor targeting by SAR3419 was further
`shown by comparing the efficacy of a 10 mg/kg single dose
`of SAR3419 with the efficacy of the same dose of SAR3419
`administered together with 50 mg/kg of naked huB4 anti-
`body. The coadministration of SAR3419 with excess huB4
`antibody resulted in complete abrogation of the antitumor
`activity of SAR3419, because binding of the naked huB4
`antibody to the CD19 antigen blocks binding of the con-
`jugate to its target (Fig. 5). Coadministration of SAR3419
`with an excess of a nonspecific humanized IgG1 control
`antibody (huMy9-6) had no effect on blocking the antitu-
`mor activity of SAR3419 (Fig. 5).
`SAR3419 was tested in a systemic model of DLBCL in
`which animals that were injected intravenously with the
`WSU-FSCCL cell line developed disseminated disease with
`bone marrow, liver, spleen, lymph node, and central ner-
`vous system involvement (43). In this model, SAR3419 also
`showed a dose-response relationship (2 doses with 4-day
`interval), with all animals surviving at the end of the
`experiment (150 days postdose) at 30 mg/kg, the highest
`dose tested. In the control group, all mice were euthanized
`between day 60 and day 73 because they showed a large
`burden of disseminated tumor. At the lower dose groups of
`15 mg/kg and 7.5 mg/kg, 4 out of 7 mice and 2 out of 7 mice,
`respectively, survived until day 150 (43). In contrast, there
`was no activity of the naked huB4 injected at 30 mg/kg
`(2 doses with 4-day interval) or 0.6 mg/kg of DM4 (2 doses
`with 4-day interval), corresponding to the amount of DM4
`in the highest SAR3419 dose of 30 mg/kg.
`Taken together, the preclinical results show a high and
`specific activity of SAR3419 in a variety of different lym-
`phoma models. The results highlight the role of conjugation
`
`in directing the cytotoxic molecule to the tumor, allowing
`delivery of enough DM4 in vivo to induce tumor regression.
`It confers a large therapeutic window to a cytotoxic mole-
`cule that otherwise would be too toxic to be used alone (50),
`with free maytansine having little or no activity in most
`tumor models at
`its maximum tolerated dose (MTD;
`refs. 35, 43, and 45). In addition, the activity of SAR3419
`compared favorably with rituximab or CHOP treatments in
`the DLBCL model tested, suggesting opportunities for
`future improvements in lymphoma treatment (43).
`
`Imaging SAR3419 Activity in a Mouse Model
`
`Metabolic imaging with fluorodeoxyglucose positron
`emission tomography (FDG-PET) combined with comput-
`ed tomography is widely accepted for staging of lymphoma
`patients and assessment of response after completion of
`therapy. It is now being used to monitor response during
`treatment in lymphoma patients (51). We therefore inves-
`tigated whether FDG uptake in tumor is an appropriate
`noninvasive readout for monitoring response and predict-
`ing outcome of SAR3419 therapy in a disseminated model
`of lymphoma. Mice inoculated with human Daudi lym-
`phoma cells were followed longitudinally by FDG-PET. At
`an advanced stage of disease, involvement of kidney, ovary,
`and spinal cord sites was evident in all mice 23 days after cell
`inoculation (Fig. 6), and the PET signal was used to ran-
`domize the mice at baseline before treatment with 12.5 mg/
`kg SAR3419. In comparison with the control group, the
`metabolic activity in tumor-invaded regions was reduced by
`50% after treatment (Fig. 6). The response to SAR3419
`seemed to be homogeneous, with a decrease of signal in
`
`www.aacrjournals.org
`
`Clin Cancer Res; 17(20) October 15, 2011
`
`6453
`
`UPenn Ex. 2073
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/17/20/6448/1999115/6448.pdf by guest on 10 February 2023
`
`Figure 5.
`Impact of naked huB4
`antibody on the efficacy of a single
`administration of SAR3419 against
`a bulky Ramos subcutaneous
`tumor xenograft model. SCID mice
`were inoculated with Ramos tumor
`cells, and once the xenografts had
`reached about 295 mm3 on day 11,
`mice (6 per group) were treated with
`a test article given by intravenous
`injection (tail vein). The huMy9-6
`(anti-CD33 antibody) is an isotype-
`matched, nonspecific control
`humanized IgG1 antibody; huB4 is
`the anti-CD19 humanized IgG1
`antibody moiety of SAR3419;
`huB4-DM4 is a synonym for
`SAR3419.
`
`PBS
`10 mg/kg huB4-DM4
`
`10 mg/kg huB4-DM4 + 50 mg/kg huB4
`10 mg/kg huB4-DM4 + 50 mg/kg huMy9-6
`
`CCR FOCUS
`
`2500
`
`2000
`
`1500
`
`1000
`
`500
`
`Measured tumor volume (mm3)
`
`0
`
`0
`
`5
`
`10
`
`15
`
`30
`
`25
`20
`Days (post inoculation)
`© 2011 American Association for Cancer Research
`
`35
`
`40
`
`45
`
`all invaded sites, including the spinal cord. This decrease in
`PET signal translated into prolonged survival for the mice in
`the treated group, with a 40% median increase in lifespan.
`In a separate experiment, modulation of the metabolic
`signal was used to adapt the treatment schedule. The mice
`were injected twice with 15 mg/kg SAR3419 with a 5-day
`interval between both injections. Within 5 days after the
`start of therapy, the metabolic activity had decreased to
`background values. The PET signal then recurred and
`increased to reach the initial values 15 days after the end
`of the first cycle, showing relapse of the disease. A second
`cycle of treatment was then administered and was shown to
`control the disease, as evidenced by the stabilization of the
`PET signal for 15 additional days. Again, the modulation of
`the PET signal by SAR3419 therapy was predictive of out-
`come, with a 175% increase in lifespan for treated mice.
`Serial imaging of tumor uptake of FDG can therefore serve
`as a translational efficacy readout for SAR3419 therapy.
`
`SAR3419 Clinical Development
`
`Two phase I dose escalation studies exploring 2 different
`schedules of administration [every 3 weeks (Q3W; ref. 27)
`and weekly (Q1W; ref. 52)] have been conducted with
`SAR3419 in patients with refractory/relapsed B-cell NHL
`expressing CD19.
`The preliminary results from the Q3W trial, in which
`patients were eligible for up to 6 cycles of treatment, showed
`that the MTD was 160 mg/m2 (4.3 mg/kg), a dose level
`that was subsequently used to treat an expanded cohort of
`20 patients (27). The dose-limiting toxicity (DLT) at doses >
`200 mg/m2 was reversible toxicity to the cornea that did not
`preclude continued dosing (at 208 mg/m2) in patients
`
`receiving clinical benefit, albeit with dose delays of 1 to 2
`weeks (27). This ocular toxicity, which consisted mainly of
`blurred vision, was associated with microcystic epithelial
`corneal changes and was typically seen after cycle 2 or later
`cycles. The DLT observed in this Q3W phase I study of
`SAR3419 is similar to that observed in phase I studies of
`albumin-bound paclitaxel (53), as well as one other ADC
`compound from among the several ADCs in clinical eval-
`uation that have the same SPDB-DM4 linker-maytansinoid
`format (28, 54). No other clinically significant grade 3 or 4
`toxicities exceed an incidence of 10%, including peripheral
`neuropathy and gastrointestinal toxicities that are classical-
`ly seen with agents targeting tubulin (27). As with other
`antibody-maytansinoid conjugates (28), there were no
`clinically significant hematologic toxicities, and no gastro-
`intestinal toxicities were reported at the MTD, showing the
`efficacy of the linker between the huB4 antibody and the
`DM4 in the blood stream [the toxicities of free maytansine
`included myelosuppression and severe gastrointestinal
`toxicities (50)]. The half-life of SAR3419 in these patients
`was 4 to 6 days and was approximately linear across all doses
`from 20 mg/m2 to 270 mg/m2 in the phase I trial (27). This
`suggests that there was a negligible antigen sink from
`normal tissue expression, as might be anticipated for a
`population of heavily pretreated patients with NHL, all of
`whom had received prior therapy with rituximab, which
`effectively depletes normal B cells.
`Tumor shrinkage was observed in more than half of the
`35 response-evaluable patients (74%) at the time of initial
`reporting of the study, with 6 objective responses (27). Of
`note, 7 of 15 patients with rituximab-refractory disease
`showed tumor shrinkage, with 1 objective response.
`Although these are early results from a phase I trial that
`
`6454
`
`Clin Cancer Res; 17(20) October 15, 2011
`
`Clinical Cancer Research
`
`UPenn Ex. 2073
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/17/20/6448/1999115/6448.pdf by guest on 10 February 2023
`
`Anti-CD19-Maytansinoid Conjugate
`
`Day23
`
`Day27
`
`Day30
`
`Urinary
`bladder
`
`Adipose
`tissues
`
`Day24
`
`
`
`© 2011 American Association for Cancer Research© 2011 American Association for Cancer Research
`
`Control
`
`Tail
`
`Hind
`limb
`
`Front
`limb
`
`Head
`
`SAR3419
`12.5mg/kg
`
`Figure 6. Longitudinal monitoring of SAR3419 activity using FDG-PET in mice inoculated intravenously with Daudi lymphoma. Samples of serial FDG-PET
`images obtained in mice bearing disseminated

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