throbber

`International Journal of
`
`Molecular Sciences
`
`Communication
`High Affinity Promotes Internalization of Engineered
`Antibodies Targeting FGFR1
`
`Łukasz Opali ´nski 1,*, Jakub Szymczyk 1, Martyna Szczepara 1, Marika Kuci ´nska 1,
`Daniel Krowarsch 2, Małgorzata Zakrzewska 1 and Jacek Otlewski 1,*
`
`1 Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a,
`50-383 Wroclaw, Poland; jakub.szymczyk@uwr.edu.pl (J.S.); martyna.szczepara@uwr.edu.pl (M.S.);
`kucinska.marika@gmail.com (M.K.); malgorzata.zakrzewska@uwr.edu.pl (M.Z.)
`2 Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a,
`50-383 Wroclaw, Poland; daniel.krowarsch@uwr.edu.pl
`* Correspondence: lukasz.opalinski@uwr.edu.pl (Ł.O.); jacek.otlewski@uwr.edu.pl (J.O.);
`Tel.: +48-71-375-2631 (Ł.O.); +48-71-375-2824 (J.O.)
`
`Received: 15 April 2018; Accepted: 8 May 2018; Published: 10 May 2018
`
`# & !*-
`0+/ .
`
`Abstract: Fibroblast growth factor receptor 1 (FGFR1) is a plasma membrane protein that transmits
`signals from the extracellular environment, regulating cell homeostasis and function. Dysregulation
`of FGFR1 leads to the development of human cancers and noncancerous diseases. Numerous tumors
`overproduce FGFR1, making this receptor a perspective target for cancer therapies. Antibody-drug
`conjugates (ADCs) are highly potent and selective anticancer agents. ADCs are composed of
`antibodies (targeting factors) fused to highly cytotoxic drugs (warheads). The efficiency of ADC
`strategy largely depends on the internalization of cytotoxic conjugate into cancer cells. Here, we have
`studied an interplay between affinity of anti-FGFR1 antibodies and efficiency of their cellular uptake.
`We have developed a unique set of engineered anti-FGFR1 antibodies that bind the same epitope
`in the extracellular part of FGFR1, but with different affinities. We have demonstrated that these
`antibodies are effectively taken up by cancer cells in the FGFR1-dependent manner. Interestingly,
`we have found that efficiency, defined as rate and level of antibody internalization, largely depends
`on the affinity of engineered antibodies towards FGFR1, as high affinity antibody displays fastest
`internalization kinetics. Our data may facilitate design of therapeutically relevant targeting molecules
`for selective treatment of FGFR1 overproducing cancers.
`
`Keywords: affinity; cancer therapy; engineered antibodies; FGFR1; internalization
`
`1. Introduction
`
`Cancer is one of the top causes of mortality worldwide. Currently, nearly one in six deaths is due
`to cancer and it is expected that the number of new cases will rise by 70% in the coming two decades [1].
`Traditional anti-cancer therapies usually aim for inhibition of high proliferative capacity of cancer cells.
`However, most of the targeted pathways are also critical for maintenance of normal cells, thus giving
`rise to numerous side effects of conventional anti-cancer drugs. In recent years, engineered monoclonal
`antibodies and antibody fragments have attracted attention as molecules that may ensure specificity
`of the cancer treatment [2]. Such antibodies inactivate the specific receptor on cancer cells, resulting
`in induction of apoptosis, or lead to cancer cell death by stimulation of the immune system of the
`patient [2]. Alternatively, engineered antibodies may be physically linked to highly potent cytotoxic
`drugs in the antibody-drug conjugates (ADCs). In the ADC approach, antigen-positive cancer cells
`are recognized by the antibody part of the ADCs. Next, ADCs bound to the cell surface antigen are
`internalized, utilizing one of cellular endocytic routes. Subsequently, ADCs traffic via cellular vesicular
`
`Int. J. Mol. Sci. 2018, 19, 1435; doi:10.3390/ijms19051435
`
`Lassen - Exhibit 1013, p. 1
`
`www.mdpi.com/journal/ijms
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`2 of 14
`
`compartments to their final lysosomal destination, where proteolytic degradation releases cytotoxic
`drugs from the ADCs. Drug moiety diffuses out from lysosomes and binds its intracellular target
`causing cell death [3,4]. Therefore, the effectiveness of ADC therapy depends on the selectivity and
`strength of antigen binding, tumor penetration and on the efficiency of ADCs internalization from the
`cell surface [5–9].
`The fibroblast growth factor receptors comprise a group of four conserved receptor tyrosine
`kinases (FGFR1-FGFR4) that, in conjunction with extracellular fibroblast growth factors (FGFs),
`transmit signals across the plasma membrane. Binding of FGFs to FGFRs (fibroblast growth factor
`receptors) leads to the activation of the receptor cytoplasmic tyrosine kinase domain that recruits
`numerous signaling molecules further propagating the signal [10,11]. The FGFR-dependent signaling
`cascades govern cell metabolism, proliferation, and apoptosis and are critical for angiogenesis,
`organogenesis, and wound healing [12]. The aberrations in the FGFRs such as gene amplification,
`rearrangements, and somatic mutations are often observed in cancer and can be found in over 7% of
`all tumors [13].
`FGFR1 is an attractive target for selective chemotherapy in ADC, as it is localized on the cell
`surface, thus being easily accessible to extracellular targeting molecules [10,11]. In numerous cancer
`cell types, the level of FGFR1 is elevated in comparison to the normal cells that may ensure selectivity of
`drug targeting [14]. Moreover, FGFR1 is rapidly internalized, mainly via clathrin-mediated endocytosis,
`providing the intracellular release of the drug after lysosomal degradation of ADC inside cancer
`cells [15]. The requirements for the design of highly internalizing antibodies against FGFR1 suitable as
`an ADC’s carrier are still largely undefined. We have recently developed novel antibody fragment
`scFvD2-Fc and have demonstrated that bivalency of scFvD2-Fc promotes internalization of this
`anti-FGFR1 engineered antibody by inducing receptor dimerization [16,17]. Here, we have assessed
`the importance of engineered antibodies affinities towards FGFR1 for their internalization. Using the
`phage display approach, we have selected two novel FGFR1-specific antibodies that bind to the
`same epitope within extracellular part of FGFR1 as scFvD2-Fc, but with different affinities. We have
`demonstrated that all these engineered antibodies are efficiently internalized via receptor-mediated
`endocytosis and are delivered through endosomes to lysosomes. Interestingly, our data show that an
`antibody with the highest affinity to FGFR1 displays the fastest internalization rate. Taken together,
`our data may facilitate the effective design of highly internalizing engineered antibodies suitable for
`ADC strategy of cancer treatment.
`
`2. Results
`
`2.1. Engineered Antibody Fragments Recognize the Same Epitope within D1 Domain of the FGFR1
`
`To select the panel of antibody fragments that specifically recognize FGFR1, we employed the
`phage display technique using Tomlinson I and Tomlinson J libraries and the extracellular part of
`FGFR1 (composed of D1, D2, and D3 domains) fused to the Fc fragment (FGFR1.D1-D2-D2-Fc) as an
`antigen [18]. For each library, we performed three consecutive rounds of selection. We obtained four
`novel scFv proteins that interact with the extracellular region of FGFR1: scFvK10, scFvL8, scFvL12
`and scFvP4.
`Our group has recently selected and characterized scFvD2 as a high affinity FGFR1 interactor [16].
`To obtain panel of scFv fragments that bind to the same region of the extracellular part of FGFR1
`as scFvD2 we employed epitope binning using surface plasmon resonance (SPR). We intended to
`select scFv proteins whose interaction with FGFR1-D1-D2-D3-Fc was blocked by the saturating
`concentrations of scFvD2. To this end, sensors with immobilized FGFR1-D1-D2-D3-Fc were incubated
`first with high concentration (1 µM) of scFvD2 and then with novel anti-FGFR1 scFv proteins at
`the same concentration. We observed that binding of scFvD2 to the extracellular part of FGFR1
`inhibited receptor interaction with scFvK10 and scFvL12 (Figure 1a). Moreover, the saturation of
`FGFR1-D1-D2-D3-Fc with scFvL12 blocked subsequent binding of scFvK10, suggesting that these three
`
`Lassen - Exhibit 1013, p. 2
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`3 of 14
`
`antibody fragments compete for the same binding site on FGFR1 (Figure 1a). To identify the epitope
`recognized by these three scFv proteins, we performed SPR binding studies with sensor-immobilized
`full length extracellular part of FGFR1 (FGFR1.D1-D2-D3-Fc) and truncated receptor variant lacking
`D1 domain (FGFR1.D2-D3-Fc). The lack of the D1 domain abolished the interaction of scFvD2,
`scFvK10, and scFvL12 with immobilized FGFR1 fragment, indicating that these proteins recognize
`epitope within the D1 domain of the receptor (Figure 1b). We confirmed this finding, showing
`that all three engineered antibodies bind to sensor-immobilized purified D1 domain fused to GST
`(GST-FGFR1.D125–124) (Figure 1b).
`To validate our experiments, we used FGF1, a natural FGFR1 ligand that recognizes binding site
`formed by D2 and D3 domains of the receptor. As expected, FGF1 interacted with FGFR1.D1-D2-D3-Fc
`and FGFR1.D2-D3-Fc, but did not bind to GST-FGFR1.D125–124 (Figure 1b). To identify the precise
`epitope within the D1 domain recognized by scFv proteins, we prepared a set of C-terminal truncations
`of D1 domain fused to glutathione S-transferase GST (Figure 1c). Next, we performed pull down
`experiments with purified GST-tagged D1 domain truncations and GST as a control. We found that
`scFvD2, scFvK10, and scFvL12 bound to the full length D1 domain (GST-FGFR1.D125–124) and to the D1
`truncation composed of residues 25–76 (GST-FGFR1.D125–76). The interaction was not observed when
`only N-terminal peptide, containing residues 25–40, was applied (GST-FGFR1.D125–40) (Figure 1c).
`These data demonstrate that engineered anti-FGFR1 antibodies recognize epitope that is located within
`residues 41–76 of the D1 domain of FGFR1.
`
`Figure 1. Cont.
`
`Lassen - Exhibit 1013, p. 3
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`4 of 14
`
`Figure 1. Engineered antibodies recognize the same epitope within the extracellular region of FGFR1.
`(a) SPR-based epitope binning. FGFR1.D1-D2-D3-Fc was immobilized on sensors and incubated with
`high concentrations of saturating antibodies. Next, competing antibodies were injected to assess
`if their binding to the extracellular region of FGFR1 is blocked by saturating antibodies. (b) SPR
`analysis of the interaction of scFv proteins with FGFR1 truncations. CM5 sensors were coated with
`FGFR1.D1-D2-D3-Fc, FGFR1.D2-D3-Fc, or GST-FGFR1-D125–124, and the association and dissociation
`were monitored for 240 s after injection of scFv proteins (1 µM), or FGF1 (1 µM). (c) Pull down
`experiments with scFv proteins and truncated variants of the D1 domain of FGFR1. GST and
`GST-tagged truncated versions of the D1 domain were bound to Glutathione Sepharose and incubated
`with scFv proteins. After extensive washing, bound proteins were eluted and analyzed by Western
`blotting. Membranes were first stained with CBB to visualize eluted proteins and then detected with
`anti-c-myc antibodies (for visualization of scFv proteins containing C-teminal c-myc).
`
`Next, we assessed specificity of scFvD2, scFvK10, and scFvL12 towards FGFR1 using SPR. Using
`extracellular regions of all four FGF receptors (FGFR1-4) immobilized on CM5 sensors, we observed
`binding of scFvD2, scFvK10, and scFvL12 only to FGFR1 (Figure 2). The high selectivity of scFv-Fc
`proteins towards FGFR1 can be attributed to the very low homology of FGF receptors within residues
`41–76 (13.9% identity, while sequence identity in the full extracellular region of FGF receptor is 36.7%)
`(Figure S1). To confirm that immobilized fragments of FGF receptors were functional, we employed
`developed in our group scFvF7 that recognizes FGFR2 [19] as well as commercial anti-FGFR3 and
`anti-FGFR4 antibodies as controls (Figure 2 and Figure S2).
`These results demonstrate that three independently selected scFv proteins display high specificity
`towards FGFR1 and recognize the same epitope (residues 40–76) within the D1 domain of FGFR1.
`
`Lassen - Exhibit 1013, p. 4
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`5 of 14
`
`Extracellular regions of FGFR1
`Specificity of scFv proteins towards FGFR1.
`Figure 2.
`(FGFR1.D1-D2-D3-Fc (a); FGFR2 (FGFR2.D1-D2-D3-Fc (b); FGFR3 (FGFR3.D1-D2-D3-Fc (c); and FGFR4
`(FGFR4.D1-D2-D3-Fc (d); were immobilized on CM5 sensors and incubated with scFvD2, scFvL10,
`scFvK12, scFvF7 (specific towards FGFR2), and commercial anti-FGFR3 and anti-FGFR4 antibodies.
`
`Lassen - Exhibit 1013, p. 5
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`6 of 14
`
`2.2. Engineered Antibodies Differ in the Affinity towards FGFR1
`
`We have recently demonstrated that reformatting of the scFvD2 into bivalent scFvD2-Fc largely
`improved internalization of this engineered antibody by inducing FGFR1 dimerization [16,17]. To study
`the impact of antibody affinity to FGFR1 on the internalization of antibody-receptor complexes,
`we have also prepared Fc-fusions of scFvK10 and scFvL12 (scFvK10-Fc and scFvL14-Fc). Proteins were
`expressed in chinese hamster ovary (CHO) cells and purified using affinity chromatography. Next,
`the binding of scFvK10-Fc and scFvL12-Fc to the extracellular region of FGFR1 (FGFR1.D1-D2-D3-Fc)
`were analyzed by SPR technique. The calculated kinetic parameters (kon, koff and KD) clearly
`demonstrate that scFvD2-Fc displays the highest affinity towards FGFR1 (0.59 nM) [16], while
`scFvK10-Fc and scFvL12-Fc bind to the FGFR1 with over tenfold weaker affinities than scFvD2-Fc
`(9.41 nM and 13.4 nM, respectively) (Table 1, Figure S3).
`
`Table 1. Kinetic parameters of scFv-FGFR1 interaction measured with SPR. Sensor-immobilized
`FGFR1.D1-D2-D3-Fc was incubated with various concentrations of scFv-Fc proteins and kinetic
`parameters (ka, kd, and KD) were determined using BIAevaluation 4.1 software. * values from
`[16]. KD errors represent standard deviation.
`
`scFvD2-Fc *
`
`scFvK10-Fc
`
`scFvL12-Fc
`
`KD (nmol/L)
`kon (M−1s−1)
`koff (s−1)
`
`0.59
`6.47 × 105
`3.84 × 10−4
`
`9.41 ± 1.47
`6.82 × 105
`6.42 × 10−3
`
`11.6 ± 1.25
`4.08 × 105
`5.45 × 10−3
`
`2.3. Antibody Fragments Are Internalized via Receptor Mediated Endocytosis
`
`Next, we studied the internalization of engineered antibody fragments. For this purpose,
`we labeled scFvD2-Fc, scFvK10-Fc, and scFvL12-Fc with pH sensitive fluorophore pHrodo Red.
`The fluorescence intensity of pHrodo Red is very low at neutral or basic pH (outside the cells or on the
`cell surface), but largely increases in the acidic milieu of the endosomal and lysosomal lumen (when
`labeled antibody is internalized) [20]. Fluorescently labeled engineered antibodies were incubated
`with model U2OSR1 cells (overproducing FGFR1) and control U2OS cells (with very low level of FGF
`receptors) and analyzed by fluorescence microscopy. Internalization of scFv-Fc proteins was minimal
`for U2OS cells, while all three studied engineered antibodies were efficiently taken up by U2OSR1 cells
`(Figure 3a). Quantitative analysis of fluorescence intensities confirmed that all three scFv-Fc proteins
`required FGFR1 for their internalization (Figure 3b).
`Upon internalization, engineered antibodies can be directed to recycling endosomes, or to
`endosomes that fuse with lysosomes for degradation [15]. As epidermal growth factor (EGF) is
`a marker of endosomes following the degradation pathway, we simultaneously applied fluorescently
`labeled EGF (EGF-AlexaFluor-488) to visualize these compartments [21]. We observed a high
`degree of colocalization of pHrodo Red-labeled scFvD2-Fc, scFvK10-Fc, and scFvL12-Fc with
`EGF-AlexaFluor-488, which suggests that engineered antibodies are sequestered into endosomal
`compartments destined for fusion with lysosomes (Figure 4a). As expected, at longer time
`points (120 min) we observed colocalization of pHrodo Red-labeled antibody fragments with the
`lysosome-specific dye–Lysotracker Green DND-20 (Figure 4b). All these data demonstrate that
`engineered antibodies are internalized via FGF receptor-mediated endocytosis and are directed to
`lysosomes for degradation.
`
`Lassen - Exhibit 1013, p. 6
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`7 of 14
`
`Figure 3. FGFR1-dependence of scFv proteins internalization. (a) U2OS (control) and U2OSR1 cells
`(overproducing FGFR1) were incubated for 2 h with pHrodo Red labeled scFv proteins (red). Nuclei
`were stained with NucBlue Live (blue) and cells were analyzed by fluorescence microscopy. Scale
`bars represent 20 µm. (b) Quantification of internalization of pHrodo Red-labeled scFvs into U2OS
`and U2OSR1 cells. pHrodo Red fluorescence intensity inside single cells was measured using Zen
`2.3 software (Zeiss, Oberkochen, Germany). At least 30 cells were measured from five fields of view
`for U2OS and U2OSR1 cells. Each square on the graph represents fluorescence intensity of single cell.
`Black squares represent average fluorescence intensity for particular cell line.
`
`Lassen - Exhibit 1013, p. 7
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`8 of 14
`
`Figure 4. Intracellular trafficking of engineered antibodies. (a) U2OSR1 cells were incubated for 1 h
`with pHrodo Red labeled scFv proteins (red) and EGF-AlexaFluor-488 (green) (marker of internalization
`pathway directing cargo for lysosomal degradation). Nuclei were stained with NucBlue Live (blue)
`and cells were analyzed by fluorescence microscopy. Scale bars represent 20 µm. (b) U2OSR1 cells
`were incubated for 2 h with pHrodo Red labeled scFv proteins (red). Lysosomes were subsequently
`stained with Lysotracker Green DND-20 (green) and nuclei were labeled with NucBlue Live (blue).
`Cells were analyzed by fluorescence microscopy. Arrowheads mark selected regions of high degree of
`signal colocalization. Scale bars represent 20 µm.
`
`2.4. High Affinity Promotes Internalization of Engineered Antibodies
`
`As differences in the efficiency of chemical labeling made the quantitative comparison of the
`internalization rates of fluorescently labeled antibodies impossible, we developed a fluorescence
`
`Lassen - Exhibit 1013, p. 8
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`9 of 14
`
`microscopy assay where we utilized non-labeled scFv-Fc proteins. Engineered antibodies were
`incubated with the U2OSR1 cells and the internalization was blocked at various time points by placing
`the cells on ice. Surface-bound antibodies were subsequently removed by washing the cells with
`buffer containing high salt concentration and low pH (HSLP). Cells were then fixed, permeabilized,
`and incubated with Zenon-AF-488, fluorescently labeled Fab fragment specifically recognizing the Fc
`fragment of the human origin, to visualize internalized scFv-Fc proteins (Figure 5a).
`
`(a) Scheme of the experimental
`Figure 5. Kinetics of engineered antibodies internalization.
`setup. Non-labeled scFv-Fc proteins were incubated with U2OSR1 cells to allow for binding and
`internalization. Next, cell surface-bound engineered antibodies were removed by washing, cells
`were permeabilized and incubated with Zenon-AlexaFluor-488, a fluorescently labeled Fab fragment
`specifically recognizing Fc fragment of human origin that detects non-labeled, internalized scFv-Fc
`proteins.
`(b) U2OSR1 cells were incubated with scFv-Fc proteins for various time points and
`internalized engineered antibodies were detected with Zenon-AlexaFluor-488 (green), as described
`above. Nuclei were labeled with NucBlue Live (blue). Scale bars represent 20 µm. (c) Kinetics of scFv-Fc
`proteins internalization. Fluorescence intensity of Zenon-AlexaFluor-488 (visualizing internalized
`scFv-Fc proteins) was measured within single U2OSR1 cells at various time points. At least 20 cells
`were measured from three fields of view for each time point. The values represent average fluorescence
`intensity from three experiments per cell. Error bars represent +/− SEM.
`
`Lassen - Exhibit 1013, p. 9
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`10 of 14
`
`We observed that scFvD2-Fc displayed the fastest kinetics of internalization and the highest
`accumulation within U2OSR1 cells (Figure 5b,c). The internalization of the other two engineered
`antibodies (scFvK10-Fc and scFvL12-Fc) that bind to the same antigen within FGFR1, but with about
`15 times lower affinity, was much slower and lower amounts of antibodies were taken up by the
`cells (Figure 5b,c). These data demonstrate that efficiency of the receptor-mediated endocytosis of
`anti-FGFR1 antibodies largely depends on the affinity of antibodies towards the receptor.
`
`3. Discussion
`
`Success in selective targeting of tumors with engineered antibodies and their conjugates largely
`depends on the properties of the cancer antigen and applied antibody. The characteristics of an “ideal”
`antigen for ADC strategy are still under the debate. Nevertheless, the “ideal” antigen is believed to
`be present at an elevated level on the surface of cancer cells, efficiently endocytosed and targeted to
`lysosomes [3–5]. On the other hand, the perfect targeting molecule (antibody), especially in the ADC
`approach, should display high affinity towards specific cancer marker (ensuring selectivity) and high
`internalization rate, facilitating the efficient delivery of its cargo (cytotoxic drug) [3–5]. The relationship
`between affinity of antibodies to cell surface antigens and their internalization kinetics is still not
`established. It was demonstrated that high affinity to antigen promotes internalization of antibodies
`directed against CD44 and HER2 receptors [22,23]. In contrast, differences in binding to target had
`no influence of the internalization rates of anti-CEA scFv proteins [24]. Moreover, the significance
`of antibodies affinities for tumor targeting in vivo is controversial. Some high affinity antibodies
`display poor tumor penetration, while in the other cases high affinity supports tumor targeting by
`antibodies [6–9]. Therefore, in case of each antigen–antibody pair it is important to assess the antigen
`properties and the significance of interaction strength for the efficiency of antibody internalization and
`for tumor targeting in vivo.
`Here, we have analyzed the interplay between the affinity of anti-FGFR1 antibody fragments to
`the receptor and their cellular uptake. We have developed a unique set of engineered antibodies that
`bind to the same epitope of extracellular part of FGFR1, but with different strength. All these antibodies
`are internalized by FGFR1-dependent endocytosis, however high affinity antibody is taken up by
`the cells more rapidly than antibodies displaying lower affinities towards FGFR1. We have recently
`demonstrated that bivalent anti-FGFR1 antibodies are rapidly internalized by promoting receptor
`dimerization that constitutes the trigger for receptor endocytosis [17]. In our experiments, we used
`excessive concentrations of scFv-Fc proteins (over 20 times higher than equilibrium dissociation
`constants), thus a vast majority of the cell surface FGFR1 was occupied by antibody fragments.
`Therefore, we hypothesize that rapid internalization of the high affinity bivalent antibody-FGFR1
`complexes is caused by enhanced FGFR1 dimerization on the cell surface.
`Taken together, our data imply that bivalency and high affinity ensure efficient internalization of
`engineered anti-FGFR1 antibodies. These results may facilitate design of targeting molecules suitable
`for ADCs directed against FGFR1-overproducing cancers.
`
`4. Materials and Methods
`
`4.1. Antibodies and Recombinant Proteins
`
`The primary anti-c-myc antibodies (sc-40), anti-FGFR3 (sc-73994), and anti-FGFR4 (sc-73995)
`were from Santa Cruz Biotechnology (Dallas, TX, USA). The primary antibody against FGFR1
`was generated by Davids Biotechnologie GmbH (Regensburg, Germany) [17]. Recombinant FGF1
`(Met-Ala-FGF122-155) was produced in E. coli, as described previously [25]. The Fc fragment and
`the full length extracellular region of FGFR1 and the extracellular part of FGFR1 lacking N-terminal
`D1 domain fused to the Fc were produced in CHO cells and purified using Protein A Sepharose
`(GE Healthcare, Piscataway, NJ, USA) [18].
`
`Lassen - Exhibit 1013, p. 10
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`11 of 14
`
`Antibody fragments in the scFv format (scFvD2, scFvI4, scFvK10, scFvL8, scFvL12, scFvP4,
`scFvF7 containing c-myc) were expressed in E. coli HB2151 and purified using Protein A Sepharose.
`scFv proteins in the bivalent Fc format (scFvD2-Fc, scFvK10-Fc and scFvL12-Fc) were produced in
`CHO cells and purified with Protein A Sepharose [16].
`The DNA sequence encoding the D1 domain of FGFR1 (residues 25–124) was cloned into
`pDEST15 using Gateway Cloning (Thermo Fisher Scientific; Waltham, MA, USA), resulting in
`plasmid pEXP15-D125–124 (allowing for production of GST-FGFR1-D125–124). Plasmids for production
`of truncated variants of D1 domain: pEXP-D125–76 (for production of GST-FGFR1-D125–76) and
`pEXP-D125–40 (for production of GST-FGFR1-D125–40) were generated by introducing stop codon
`in the vector pEXP15-D125–124 by site directed mutagenesis. GST-tagged truncated variants of the D1
`domain were produced in E. coli BL21 CodonPlus(DE3)-RIL (Agilent Technologies; Santa Clara, CA,
`USA) and purified with Glutathione Sepharose (GE Healthcare, Piscataway, NJ, USA), according to the
`standard procedure.
`
`4.2. Cells
`
`U2OS (human osteosarcoma, ATCC #HTB-96) and U2OSR1 cells (U2OS cells stably expressing
`FGFR1) were a kind gift of Dr. E.M. Haugsten from the Department of Molecular Cell Biology, Institute
`for Cancer Research, Oslo University Hospital, Norway. Cells were cultured in 5% CO2 atmosphere at
`37 ◦C in Dulbecco’s Modified Eagle’s Medium (Biowest, Nuaille, France) supplemented with 10% fetal
`bovine serum (Thermo Fisher Scientific, Waltham, MA, USA), antibiotics mix (100 U/mL penicillin,
`100 µg/mL streptomycin (Thermo Fisher Scientific; Waltham, MA, USA), and 1 mg/mL geneticin
`(in the case of U2OSR1 cells) (Thermo Fisher Scientific; Waltham, MA, USA). Cells were seeded into
`tissue culture plates one day prior start of the experiments.
`
`4.3. Selection of Antibody Fragments against FGFR1 by Phage Display Technology
`
`The selection of scFv proteins recognizing an extracellular region of FGFR1 was carried out using
`two commercial phage libraries of human scFv (Tomlinson I and J) as described previously [16].
`Three cycles of panning were performed on MaxiSorp 96-well plates (Thermo Fisher Scientific;
`Waltham, MA, USA) coated with FGFR1.D1-D2-D3-Fc with counter selection using purified Fc
`protein. To confirm the interaction with FGFR1 bacterial supernatants containing selected scFv proteins
`were subjected to the monoclonal ELISA and the SPR assay with immobilized FGFR1.D1-D2-D3-Fc,
`as described in [16].
`
`4.4. SPR-Based Interaction Studies and Affinity Measurements
`
`SPR experiments were performed on the Biacore 3000 instrument (GE Healthcare, Piscataway,
`NJ, USA) at 25 ◦C. For epitope binning FGFR1.D1-D2-D3-Fc was immobilized on a CM5 sensor (GE
`Healthcre) at 825 RU and scFv proteins (1 µM) were sequentially injected at flow rate of 30 µL/min.
`The association and dissociation were monitored for 240 s.
`For analysis of the binding site of scFv proteins within FGFR1, CM5 sensors were coated with
`FGFR1.D1-D2-D3-Fc (at 825 RU), FGFR1.D2-D3-Fc (at 900 RU), or GST-FGFR1-D125–124 (at 300 RU).
`Next, scFv proteins (1 µM) and FGF1 (1 µM) were injected and the association and dissociation were
`monitored for 240 s.
`For specificity analysis of generated scFv proteins, FGFR1.D1-D2-D3-Fc (at 825 RU),
`FGFR2.D1-D2-D3-Fc (at 1000 RU), FGFR3.D1-D2-D3-Fc (at 1000 RU), and FGFR4.D1-D2-D3-Fc (at
`1000 RU) were immobilized on CM5 sensors. Next, scFv proteins (1 µM), anti-FGFR1 antibody (as a
`positive control for interaction with FGFR1.D1-D2-D3-Fc) [17], scFvF7 (1 µM) (as a positive control
`for interaction with FGFR2.D1-D2-D3-Fc) [15] and two commercial antibodies against FGFR3 and
`FGFR4 (0.1 µM; as positive controls for interaction with FGFR3.D1-D2-D3-Fc and FGFR4.D1-D2-D3-Fc,
`respectively) were injected independently over all sensors at 30 µL/min flow rate. The association
`
`Lassen - Exhibit 1013, p. 11
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`12 of 14
`
`and dissociation were monitored for 240 s. All sensograms were analyzed using BIAevaluation 4.1
`software (GE Healthcare, Piscataway, NJ, USA).
`interaction,
`antibodies–FGFR1
`engineered
`of
`To
`determine
`kinetic
`parameters
`FGFR1.D1-D2-D3-Fc was immobilized at 825 RU on a CM5 sensor. Various concentrations of
`scFv-Fc proteins (2.5–320 nM) were measured for 300 s at 30 µL/min flow rate. Kinetic constants (ka,
`kd, and KD) were calculated using BIAevaluation 4.1 software using 1:1 Langmuir binding model with
`drifting baseline.
`
`4.5. Pull-Down
`
`To study the interaction of scFv proteins with purified GST-tagged full length D1 domain of
`FGFR1 and the D1 domain truncations, purified GST (control, 10 µg) and GST-tagged D1 domain
`variants (10 µg) were diluted in washing buffer (WB): 50 mM Tris, 150 mM NaCl, pH 7.5, and bound
`to Glutathione Sepharose. Next, scFv proteins (10 µg) in WB were incubated with resin-bound scFv
`proteins. Resins were extensively washed with WB and bound proteins were eluted with elution
`buffer: 50 mM Tris, 150 mM NaCl, 20 mM glutathione, pH 7.5. Eluates were analyzed by Western
`blotting. Before detection membranes were stained with CBB to visualize the amount of eluted
`GST-tagged truncated variants of the D1 domain and GST. After destaining, membranes were probed
`with anti-c-myc antibodies.
`
`4.6. Fluorescence Microscopy
`
`Purified scFvD2-Fc, scFvK10-Fc, and scFvD12-Fc were chemically labeled with pHrodo Red
`(Thermo Fisher Scientific; Waltham, MA, USA) according to manufacturer’s protocol. For analysis of
`the FGFR1-dependence engineered antibodies internalization, U2OS and U2OSR1 cells were incubated
`with pHrodo Red-labeled engineered antibodies (10 µg/mL) for 2 h at 37 ◦C. Cells were extensively
`washed with PBS, fixed with 4% PFA, nuclei were stained with NucBlue Live (Thermo Fisher Scientific;
`Waltham, MA, USA) and cells were analyzed by fluorescence microscopy. For quantitation of
`engineered antibodies internalization, pHrodo Red fluorescence intensity was measured within single
`cells using ZEN 2.3 software (Zeiss, Oberkochen, Germany). At least 30 cells were measured from five
`fields of view for U2OS and U2OSR1 cells.
`For analysis of the intracellular trafficking of engineered antibodies, serum-starved U2OSR1
`cells were incubated for 1 h at 37 ◦C with pHrodo Red-labeled scFv-Fc proteins (10 µg/mL) and
`EGF-AlexaFluor-488 (15 µg/mL) (Thermo Fisher Scientific; Waltham, MA, USA). Alternatively,
`U2OSR1 cells were incubated for 2 h with pHrodo Red labeled scFv-Fc proteins (10 µg/mL) and
`stained with lysosome-specific dye Lysotracker Green DND-20 (Thermo Fisher Scientific; Waltham,
`MA, USA) according to manufacturer’s protocol. Nuclei were stained with NucBlue Live and cells
`were analyzed by fluorescence microscopy.
`For analysis of the kinetics of engineered antibodies internalization, non-labeled scFv-Fc proteins
`and U2OSR1 cells were used. Cells were incubated with scFv-Fc proteins (15 µg/mL) at 37 ◦C and
`internalization was stopped at various time points by placing the cells on ice. Cells were then washed
`extensively with PBS and with HSLP buffer (high salt low pH) (2 M NaCl, 40 mM NaAc, pH 4.0) to
`remove cell surface-bound antibodies. Next, cells were fixed with 4% PFA and permeabilized with 0.1%
`Triton X-100 in PBS. Cells were subsequently blocked with 2% BSA in PBS and internalized scFv-Fc
`proteins were visualized by addition of Zenon-AlexaFluor-488 (Thermo Fisher Scientific; Waltham,
`MA, USA) that recognizes the Fc fragment of human origin, accordingly to supplier’s protocol.
`The excessive Zenon-AlexaFluor-488 was blocked with Blocking Reagent and after extensive washing
`cells were fixed again to preserve scFv-Fc- Zenon-AlexaFluor-488 complexes. Next, nuclei were stained
`with NucBlue Live and cells were analyzed with fluorescence microscopy. Zenon-AlexaFluor-488
`fluorescence intensity within single U2OSR1 cells was measured using ZEN 2.3 software. At least
`20 cells were measured from three fields of view for three experiments. Average intensity +/− SEM
`were plotted.
`
`Lassen - Exhibit 1013, p. 12
`
`

`

`Int. J. Mol. Sci. 2018, 19, 1435
`
`13 of 14
`
`Wide-field fluorescence microscopy was carried out using a Zeiss Axio Observer Z1 fluorescence
`microscope (Zeiss, Oberkochen, Germany). Images were taken using LD-Plan-Neofluar 40×/0.6
`Korr M27 objective and Axiocam 503 camera. pHrodo Red signal was visualized with a 540/552 nm
`bandpass excitation filter and a 575/640 nm bandpass emission filter, EGF-AlexaFluor-488, Lysotracker
`Green DND-20 and Zenon-AlexaFluor-488 signal was visualized with a 450/490 nm bandpass
`excitation filter and a 550/590 nm bandpass emission filter. NucBlue Live signal was visualized
`with a 335/383-nm bandpass excitation filter and

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