RESEARCH ARTICLE IMMUNOTHERAPY

Anti-CD20/CD3 T cell–dependent bispecific antibody for the treatment of B cell malignancies

Bispecific antibodies and antibody fragments in various formats have been explored as a means to recruit cytolytic T cells to kill tumor cells. Encouraging clinical data have been reported with molecules such as the anti-CD19/CD3 bispecific T cell engager (BiTE) blinatumomab. However, the clinical use of many reported T cell–recruiting bispecific modalities is limited by liabilities including unfavorable pharmacokinetics, potential immunogenicity, and manufacturing challenges. We describe a B cell–targeting anti-CD20/CD3 T cell–dependent bispecific antibody (CD20-TDB), which is a full-length, humanized immunoglobulin G1 molecule with near-native antibody architecture constructed using “knobs-into-holes” technology. CD20-TDB is highly active in killing CD20-expressing B cells, including primary patient leukemia and lymphoma cells both in vitro and in vivo. In cynomolgus monkeys, CD20-TDB potently depletes B cells in peripheral blood and lymphoid tissues at a single dose of 1 mg/kg while demonstrating pharmacokinetic properties similar to those of conventional monoclonal antibodies. CD20-TDB also exhibits activity in vitro and in vivo in the presence of competing CD20-targeting antibodies. These data provide rationale for the clinical testing of CD20-TDB for the treatment of CD20-expressing B cell malignancies.

INTRODUCTION The addition of the CD20-targeting antibody rituximab to chemotherapy has greatly improved the outcome for patients with B cell lymphoma and chronic lymphocytic leukemia (CLL). Nevertheless, disease relapse or recurrence will occur in virtually all patients with follicular lymphoma and CLL, and about half of patients with aggressive B cell lymphoma, for example, diffuse large B cell lymphoma. Despite repeated rounds of treatment, most will ultimately die of disease-related sequelae (1). Monoclonal antibodies targeting B cell antigens, including CD19, CD20, CD22, CD30, and CD52, have been explored for treatment of various B cell malignancies, but none has yet definitively demonstrated a safety and efficacy profile superior to rituximab (2). There therefore remains a need to develop B cell–targeting therapeutics with a distinct mechanism of action (MOA) to increase cure rates in B cell malignancies. Recent clinical reports illustrate the effectiveness of therapies that involve redirection of T cell cytolytic activity against cancer cells. One approach involves the ex vivo manipulation of autologous or allogeneic T cells to express chimeric antigen receptors (CARs) that target lineage-specific surface molecules such as CD19. CAR-expressing T cells (CAR-T cells) have produced deep and durable responses in patients with relapsed/refractory acute leukemia (3, 4). However, toxicities related to severe cytokine release syndrome, the scalability of production to the broader cancer population, and the clinical benefit in disease beyond the acute leukemia remain significant challenges for the development of CAR-T therapies (5). An alternate approach to T cell therapy involves the use of bispecific molecules that redirect endogenous T cells to recognize tumor cells. Such therapeutics concomitantly bind to CD3 on T cells and a target anti1

Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA. 2The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA. *Corresponding author. E-mail: [email protected]

gen such as CD19 on target cancer cells. The therapeutic potential of this approach is exemplified by of the recent approval of blinatumomab, a bispecific T cell engager (BiTE) targeting CD19, for treatment of relapsed/ refractory B cell acute lymphoblastic leukemia (ALL). In adult patients with relapsed/refractory ALL, treatment with blinatumomab resulted in complete responses in 26 of 36 (72%) patients, with 88% of these responders achieving minimal residual disease (MRD)–negative status, and a median overall survival of 9.0 months (6). Blinatumomab treatment was also effective in achieving MRD negativity in patients who were MRD-positive after induction and consolidation chemotherapy (7). The clinical activity of blinatumomab has demonstrated that T cell responses against tumors were achievable without the need for ex vivo immune cell manipulation, which affords advantages over cell-based therapies, especially with respect to manufacturing and access. Cytokine release syndrome and central nervous system toxicity similar to that observed with CAR-T therapy were observed with blinatumomab (8, 9), suggesting a possible class effect of redirecting T cells to target B cell malignancies. In addition to blinatumomab, other B cell–targeting bispecific proteins with a variety of structural formats have been characterized preclinically and tested in small human clinical trials (8, 10–13). Following on the development of these first-generation T cell–recruiting agents, there remain opportunities to optimize T cell–directed therapies based on the generation, characterization, and use of preclinical models. Here, we describe CD20 T cell–dependent bispecific antibody (CD20TDB), a full-length, fully humanized immunoglobulin G1 (IgG1) antibody as T cell–recruiting therapeutic for CD20+ malignancies. CD20-TDB is cross-reactive to cynomolgus monkey CD3e and CD20 antigens, allowing for appropriate preclinical testing. We provide evidence of potent in vitro and in vivo B cell killing activity and characterization of MOArelated pharmacologic activities of CD20-TDB in normal and leukemic cells, three murine models, and cynomolgus monkey. These studies highlight the use of several new preclinical approaches for the characterization

www.ScienceTranslationalMedicine.org

13 May 2015

Vol 7 Issue 287 287ra70

1

Downloaded from stm.sciencemag.org on May 14, 2015

Liping L. Sun,1* Diego Ellerman,1 Mary Mathieu,1 Maria Hristopoulos,1 Xiaocheng Chen,1 Yijin Li,1 Xiaojie Yan,2 Robyn Clark,1 Arthur Reyes,1 Eric Stefanich,1 Elaine Mai,1 Judy Young,1 Clarissa Johnson,1 Mahrukh Huseni,1 Xinhua Wang,1 Yvonne Chen,1 Peiyin Wang,1 Hong Wang,1 Noel Dybdal,1 Yu-Waye Chu,1 Nicholas Chiorazzi,2 Justin M. Scheer,1 Teemu Junttila,1 Klara Totpal,1 Mark S. Dennis,1 Allen J. Ebens1

RESEARCH ARTICLE of such B cell–targeted therapies and provide the rationale for clinical testing of CD20-TDB.

forin (Fig. 1F) (fig. S5; P = 0046, paired t test; n = 3) released into cell culture medium (16).

RESULTS

CD20-TDB is a conditional T cell agonist dependent on the presence of both T cells and target for activity We next characterized the MOA for CD20-TDB. Although the molecule was produced in Escherichia coli and therefore expected to be devoid of antibodydependent cell-mediated cytotoxicity (ADCC) activity, we first confirmed that functional activity was not dependent on the presence of antibody constant domain regions. In vitro potency was assessed against the B cell line BJAB using healthy human donor peripheral blood mononuclear cells (PBMCs) and an F(ab′)2 fragment of CD20-TDB lacking the Fc region. The fragment was found on a molar basis to be equipotent to intact CD20-TDB (Fig. 1A), suggesting that Fc receptor–mediated ADCC does not contribute substantially to CD20-TDB activity. Conversely, depletion of CD3-expressing cells from PBMCs abolished the activity of CD20-TDB (Fig. 1B), indicating that T cells are required for activity. Purified CD4+ or CD8+ T cells used as effector cells were activated by CD20-TDB with comparable potency as measured by the induction of both CD69 and CD25 expression (Fig. 1C and fig. S3). T cell activation by CD20-TDB was strictly dependent on the presence of CD20+ target cells (Fig. 1C). T cell proliferation was also observed in vitro only in the presence of target B cells and CD20-TDB (fig. S4). CD8+ T cells appeared to be more potent in BJAB cell killing because CD8+ T cells resulted in a greater extent of cell killing compared to an equal number of CD4+ T cells (Fig. 1D) (fig. S5; P = 0074, paired t test; n = 3). Consistent with these results, we observed significant intracellular granzyme B up-regulation that was more prominent in activated CD8+ T cells compared to CD4+ T cells (Fig. 1E) (fig. S5; P = 0068, paired t test; n = 3), as well as higher levels of per-

Downloaded from stm.sciencemag.org on May 14, 2015

CD20-TDB is a full-length humanized IgG with conventional antibody pharmacokinetic properties We have recently described modular production of full-length IgG1 bispecific antibodies with near-native architecture using “knobs-into-holes” technology (14). Anti-CD20/CD3 TDB (CD20-TDB) was produced without detectable level of homodimers and aggregates (fig. S1). As predicted from our previous experience with this molecular format (15), the pharmacokinetic (PK) properties of CD20-TDB in rats are typical of a nonbinding human IgG1 antibody, suggesting that intermittent dosing on a weekly to monthly schedule will be clinically feasible (fig. S2).

Fig. 1. Anti-CD20/CD3 TDB activates T cells in the presence of target and mediates B cell killing in a T cell–dependent manner. (A) BJAB cells and PBMCs isolated from healthy donor (1:10 cell ratio) were incubated with various concentrations of full-length CD20-TDB or F(ab′)2 CD20-TDB for 24 hours (data shown as means ± SD, n = 3). (B) BJAB cells and PBMCs isolated from healthy donor, or PBMCs depleted of CD3+T cells (1:10 cell ratio), were incubated with various concentrations of CD20-TDB for 24 hours. (C to F) BJAB cells and purified CD8+T cells or CD4+ T cells, or CD8+T cells only, were incubated with various concentrations of CD20-TDB for 24 hours. [T cell–to–BJAB cell count ratio was 5:1 in (C), (E), and (F), or 3:1 in (D) as duplicate]. Cell killing and T cell activation marked as CD69+CD25+ were measured and calculated as described in Materials and Methods. Granzyme B (GrB) induction was also detected by fluorescence-activated cell sorting (FACS), and perforin concentration in medium was measured by enzyme-linked immunosorbent assay (ELISA). Assays were done either in triplicate with average and SD value plotted (A) or as single-dose response curve representative of multiple assays of different donors.

www.ScienceTranslationalMedicine.org

13 May 2015

Vol 7 Issue 287 287ra70

2

CD20-TDB is broadly active across healthy donor B cells and lymphoma cell lines CD20-TDB was tested in dose-response assays across a panel of seven B lymphoma cell lines with various levels of surface CD20 expression and half-maximal effective concentration (EC50) values ranging from 0.22 to 11 ng/ml in vitro (Fig. 2A). CD20-TDB had no activity against SU-DHL1 cells that are devoid of CD20 expression (Fig. 2A). CD20TDB was also highly active for killing of endogenous B cells by endogenous T cells in PBMCs prepared from healthy donors. Shown in Fig. 2B are representative dose-response curves from 8 healthy donors (of 30), and the summary of EC50 values and the extent of killing from 30 healthy donors demonstrating a mean EC50 for killing of less than 3 ng/ml and a mean extent of killing about 85% after 24 hours in culture. Of the 30 healthy donors tested, 57 to 96% of B cells were killed with antibody (1000 ng/ml) within 24 hours, with EC50s ranging from 0.4 to 140 ng/ml. CD20-TDB is highly effective in transgenic murine models To date, the lack of antibody cross-reactivity to nonhuman models has greatly limited preclinical characterization of T cell–recruiting bispecific antibodies (17). We developed a double-transgenic mouse strain expressing both human CD20 and human CD3e (huCD20-huCD3) to enable detailed studies in a mouse model with an intact and fully functional immune system (18, 19). In huCD20-huCD3 double-transgenic mice, we found the expression level for both human antigens on mouse B and T cells to be appropriately lineage-restricted but lower than that on healthy human donor B and T cells, suggesting that the model may be challenging with respect to antigen copy numbers (Fig. 3A). In a dose-ranging study to

identify a minimal efficacious dose that would cause robust depletion of B cells, we assessed numbers of circulating and splenic B cells after a single administration of CD20-TDB ranging from 0.00005 to 0.5 mg/kg. Whereas a dose of 0.05 mg/kg caused transient depletion of peripheral B cells 1 day after CD20-TDB administration, a dose of 0.5 mg/kg was required to maintain B cell depletion through day 7 in peripheral blood (Fig. 3B and fig. S6). In spleen samples at day 7, the 0.05 mg/kg dose approximated an ED50 (half-maximal effective) dose level, whereas the 0.5 mg/kg dose was sufficient for complete B cell clearance (Fig. 3C). To confirm the CD3 and CD20 specificity of these effects, we dosed human CD20 single-transgenic mice with CD20-TDB, HER2 (human epidermal growth factor receptor 2)–TDB, or rituximab (15), and assessed spleen B cell numbers 7 days after administration. In this experiment, CD20-TDB and rituximab bind mouse B cells but not mouse T cells, whereas HER2-TDB binds neither mouse B or T cells nor mouse HER2. As expected, both TDB test agents were inactive as compared to rituximab (Fig. 3D). We next conducted a similar experiment comparing the activity of CD20-TDB, HER2-TDB, and rituximab in huCD20-huCD3 double-transgenic mice. In this experiment, rituximab binds only to B cells, CD20-TDB binds to both B and T cells, and HER2-TDB binds only to T cells. In contrast to the single-transgenic mouse experiment, CD20TDB was highly active in the double-transgenic mice and showed significantly greater clearance of splenic B cells than rituximab despite a 20-fold lower dose. As expected, the HER2-TDB was inactive in B cell depletion in both single- and double-transgenic mice (Fig. 3E). We further characterized immune cell dynamics in spleens of the double-transgenic mice over a 2-week period after administration of a single dose of CD20-TDB (0.5 mg/kg). We observed that splenic B cells declined ~50% from baseline by day 1 (24 hours post-dose) and continued to decline to a nadir on day 3, with slight recovery detected at day 14 (Fig. 4A). Coincident with the large decline in B cells on day 1, we observed that most splenic T cells had an activated phenotype on day 1. By day 2, most T cells were no longer CD69positive, although levels of CD69+CD8+ cells continued to range from 10 to 30% for the remainder of the 2-week observation period (Fig. 4B). Total splenic CD8+ T cell counts were markedly increased by day 2 of the study, and the timing and size of the increase were suggestive of a proliferative expansion (Fig. 4C). We also noted that, on day 3 when splenic B cells were mostly cleared, CD8+ T cell counts started to decrease significantly after the expansion and eventually came down to below the baseline levels.

Fig. 2. Anti-CD20/CD3 TDB is potent in killing human lymphoma B cell lines and healthy donor primary autologous B cells in vitro. (A) B cells and PBMCs isolated from a single healthy donor (1:10 cell ratio) were incubated 24 hours with various concentrations of CD20-TDB. SU-DHL1 cells are CD20-negative. CD20 expressions for the B cell lines were shown, with an isotype control shown in gray. (B) PBMCs isolated from healthy donors were incubated for 24 hours with CD20-TDB at the indicated concentrations. Killing curves in each plot shown are from representative assays [of 4 different donors for (A) and more than 30 different donors for (B)]. For dot plots, extent of cell killing is reported for CD20-TDB (1000 ng/ml), and each dot represents a unique donor with a horizontal bar indicating the mean. www.ScienceTranslationalMedicine.org

CD20-TDB is highly effective in murine models reconstituted with healthy human donor immune cells To provide a second independent murine model, we investigated the use of immunocompromised NSG [NOD (nonobese diabetic) SCID (severe combined immunodeficient) gamma] mice engrafted 13 May 2015

Vol 7 Issue 287 287ra70

3

Downloaded from stm.sciencemag.org on May 14, 2015

RESEARCH ARTICLE

RESEARCH ARTICLE

CD20-TDB is potent in killing primary patient CLL B cells in vitro and in vivo To characterize the CD20-TDB in the setting of B cell malignancies, we next tested the potency against primary cells from CLL patients in vitro and in vivo. CLL was selected as a “high-bar” in vitro experimental system to assess the CD20-TDB because (i) CD20 exhibits particularly low surface copy number on CLL cells (21, 22); (ii) the effector-to-target ratio is skewed because, in relative terms, T cell numbers are decreased and B cell number are increased by disease; and (iii) the T cells that are present are functionally deficient (23–26). PBMCs prepared from peripheral blood of nine CLL patients were treated with CD20-TDB to determine the in vitro potency in the setting of CLL. We first characterized activity of a single high dose of CD20-TDB (1000 ng/ml) and measured Fig. 3. Double-transgenic mouse model. (A) Human CD3e and CD20 expression levels on mouse B B and T cell content after 48 hours of culand T cells from huCD20-huCD3 double-transgenic mice were compared to healthy human donor B ture. As expected, we found that T cell conand T cells by FACS. MFI, mean fluorescence intensity. (B and C) huCD20-huCD3 double-transgenic tent varied substantially (between 0.4 and mice were treated once intravenously on day 0 with various dose levels of CD20-TDB. Mouse blood 8% of mononuclear cells). Strikingly, we [days (D) 1 and 7] and spleens (day 7) were collected, and B cells were quantitated by FACS. (D and E) observed that the extent of tumor cell killhuCD20 single-transgenic (sTG) mice or huCD20-huCD3 double-transgenic (dTG) mice were treated ing was highly correlated to T cell content once intravenously on day 0 with antibodies as indicated (10 mg/kg for rituximab and 0.5 mg/kg for (R2 value of 0.9, Fig. 6A). From four doCD20-TDB and HER2-TDB). Mouse spleens were collected at day 7 for B cell quantitation by FACS. Bars in nors of low T cell content and low B cell the plots indicate mean values, with P values calculated by unpaired t test (n = 4 or 7 mice per group). killing with available sample material, we tested whether addition of healthy donor with human CD34+ hematopoietic stem cells (20) and qualified as in- T cells could enhance CD20-TDB activity. This was indeed the case, dividual mouse reconstituted with >25% human CD45+ cells in demonstrating that T cell content of the cultures was the primary deterperipheral blood. Mice used in experiments shown in Fig. 5 had 35 to minant of TDB activity in these cultures (Fig. 6B). For two samples, 80% human CD45+ cells in peripheral blood with percentages of viable dose-response curves based on B cell killing and CD8+T cell activation leukocyte gate of CD4+, CD8+, and CD20+ cells ranging from 12 to based on CD69 and CD25 expression were generated. CD20-TDB ac25%, 2 to 9%, and 32 to 60%, respectively (fig. S7A). Levels of CD3 tivated CD8+T cells from CLL patients with a B cell killing EC50 of 10 to and CD20 expression from these mice were compared also to those of 20 ng/ml, a value similar to that in healthy donor PBMC assays despite healthy human donors. CD20 levels on B cells were slightly higher, whereas the fact that the B cell leukemia tumor burden was 70% of mononuclear CD3 levels on T cells were lower (CD8+T) or comparable (CD4+T) to those cells with only 8% CD8+T cells in patient sample A1837, and 80% observed on healthy human donors (fig. S7B). tumor burden with 4% CD8+T cells in patient sample A183D. These mice were randomized to two groups according to human CD20-TDB can therefore achieve efficient B cell killing even at low CD45+ cell counts 5 days before dosing (day −5), and treated with vehicle effector-to-target ratios (1:8 and 1:18, respectively; Fig. 6C). www.ScienceTranslationalMedicine.org

13 May 2015

Vol 7 Issue 287 287ra70

4

Downloaded from stm.sciencemag.org on May 14, 2015

or with three weekly doses of CD20-TDB at 0.5 mg/kg starting on day 0. We observed a strong reduction of B cells in peripheral blood at day 7, with almost no B cells detected at day 21 (Fig. 5A). In peripheral blood, we observed up to a 10-fold increase in CD8+ T cells by day 7 and a return to baseline or lower at days 14 and 21 (Fig. 5B). A similar trend was also observed for CD4+ T cells (fig. S8). Robust B cell depletion was also observed in spleens of the CD20TDB–treated mice at day 21 (Fig. 5C).

To further characterize the potential of CD20-TDB against CLL leukemic cells, we used a patient leukemia cell–derived adoptive transfer mice model that is a modification of a previously described approach (27). Representative examples are shown in Fig. 6D, with vehicle-treated animals demonstrating successful engraftment of B leukemic cells and autologous T cells from CLL patients. CD20-TDB treatment was initiated after confirmation of the leukemic graft. Very few B cells could be detected after a single dose of CD20-TDB treatment at 0.1 or 0.5 mg/kg. B cell depletion was also observed with rituximab treatment, whereas no B cell depletion was detected with HER2-TDB as a nontarget cell binding control.

Fig. 4. Anti-CD20/CD3 TDB is active in dTG mice. (A) Time course of tissue B cell depletion. (B and C) CD8+T cell activation and cell numbers. After a single dose of CD20-TDB in huCD20-huCD3 double-transgenic mice. huCD20-huCD3 double-transgenic (dTG) mice were treated once intravenously on day 0 with CD20-TDB (0.5 mg/kg). Spleens were collected on the indicated days, and B cell or CD8+T cell counts and T cell activation were measured by FACS. Bars in the plots indicate mean values, with P values calculated by unpaired t test (n = 3 mice per group).

CD20-TDB potency requires minimal CD20 expression and is active in the presence of rituximab To explore the impact of CD20 expression on CD20-TDB activity, we tested the potency of CD20-TDB on lymphoma cell lines NALM-6, SC-1, and OCI-Ly19, which express CD20 at very low levels (Fig. 7A). The estimated CD20 copy number for these cell lines were less than 500 per cell, based on FACS binding data in comparison to BJAB cells (28). In contrast to comparable potency of CD20-TDB and rituximab against BJAB cells (fig. S9A), CD20-low cell lines were killed by CD20TDB with an EC50 99% of the signal) in gel filtration with less than 0.2% aggregates. No homodimers were detected by mass spectrometry. The CD20 arm in the bispecific is anti-CD20 clone 2H7, whereas the CD3 arm is either clone UCHT1 or a cynomolgus cross-reactive clone GMX3c (for Fig. 8 only). In vitro B cell killing and T cell activation assays B tumor cell lines were obtained from American Type Culture Collection, and PBMCs were isolated from whole blood of healthy donors by

www.ScienceTranslationalMedicine.org

13 May 2015

Vol 7 Issue 287 287ra70

8

Downloaded from stm.sciencemag.org on May 14, 2015

RESEARCH ARTICLE

RESEARCH ARTICLE

In vivo efficacy studies in murine models All in vivo experimental procedures conformed to the guiding principles of the American Physiology Society and were approved by Genentech’s Institutional Animal Care and Use Committee. Humanized NSG mice were purchased from The Jackson Laboratory. Human CD20 transgenic mice and human CD3 transgenic mice were generated as previously described, and huCD20-huCD3 double-transgenic mice were produced by crossing mice containing each of the two single transgenes (18, 19). Whole blood was collected by puncture of the retro-orbital sinus using heparinized pipets and immediately transferred into heparinized tubes, while the animals were under anesthesia, or by terminal cardiac puncture with a heparinized syringe after CO2 euthanasia. Spleens were collected after CO2 euthanasia. For all studies, clinical observations were performed twice per week to monitor the health of the animals. Animal body weights were taken at least once a week. PBMCs were isolated after red blood cell lysis and analyzed by FACS for B cells [murine CD45+CD19+ (muCD45+CD19+)] and T cells (muCD90.2+CD4+ or muCD90.2+CD8+). All antibodies used were purchased from BD Biosciences and eBioscience. Pharmacokinetic/pharmacodynamic study in cynomolgus monkeys All cynomolgus monkey studies were conducted at Charles River Laboratories (Reno, NV) using purpose-bred, naïve, cynomolgus monkeys of Chinese origin. For the single-dose study, three male cynomolgus monkeys were administered a single slow bolus intravenous dose of CD20TDB (1 mg/kg). For the repeat dose study, four cynomolgus monkeys were administered a slow bolus intravenous dose of CD20-TDB (1 mg/kg) once weekly for a total of four doses. Whole blood or tissues were collected at selected time points for B and T cell counts by FACS. Serum was collected and stored at −70°C until assayed using an ELISA to determine the amount of test article in each serum sample. Serum concentration– time profiles from each animal were used to estimate PK parameters using WinNonlin software (Pharsight). Statistical analyses Data are presented as means ± SD or means only as stated in the figure legends. Statistically significant differences were tested by specific tests as indicated in the figure legends. P values are calculated by unpaired t test with Prism software version 5.0 (GraphPad). Sample sizes for all studies are summarized in fig. S12.

SUPPLEMENTARY MATERIALS www.sciencetranslationalmedicine.org/cgi/content/full/7/287/287ra70/DC1 Fig. S1. Quality control analysis for anti-CD20/CD3 TDB. Fig. S2. Summary of PK analysis for anti-CD20/CD3 TDB in rats.

Fig. S3. Representative FACS data for Fig. 1C. Fig. S4. In vitro CD8+ T cell proliferation in the presence of CD20-TDB and BJAB. Fig. S5. Data with three healthy human donors for statistical analysis for Fig. 1 (D to F). Fig. S6. FACS gating strategy for B and T cells with blood and spleen samples of huCD20huCD3 double-transgenic mice. Fig. S7. Presence of human B and T cells in humanized NSG mice and antigen expression level for human CD20 and CD3. Fig. S8. CD4+ T cell counts in humanized NSG mice upon CD20-TDB treatment. Fig. S9. Control studies with rituximab and rituximab-DANA antibodies. Fig. S10. FACS gating strategy for B and T cells with blood and tissue samples of cynomolgus monkeys. Fig. S11. (A and B) Cytokine production in huCD20-huCD3 double-transgenic mice (A) and in cynomolgus monkeys (B) upon CD20-TDB treatment. Fig. S12. Summary of sample sizes for all presented studies.

REFERENCES AND NOTES 1. A. Jemal, F. Bray, M. M. Center, J. Ferlay, E. Ward, D. Forman, Global cancer statistics. CA Cancer J. Clin. 61, 69–90 (2011). 2. K. Bauer, M. Rancea, V. Roloff, T. Elter, M. Hallek, A. Engert, N. Skoetz, Rituximab, ofatumumab and other monoclonal anti-CD20 antibodies for chronic lymphocytic leukaemia. Cochrane Database Syst. Rev. 11, CD008079 (2012). 3. S. A. Grupp, M. Kalos, D. Barrett, R. Aplenc, D. L. Porter, S. R. Rheingold, D. T. Teachey, A. Chew, B. Hauck, J. F. Wright, M. C. Milone, B. L. Levine, C. H. June, Chimeric antigen receptor–modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 368, 1509–1518 (2013). 4. J. N. Kochenderfer, M. E. Dudley, S. A. Feldman, W. H. Wilson, D. E. Spaner, I. Maric, M. Stetler-Stevenson, G. Q. Phan, M. S. Hughes, R. M. Sherry, J. C. Yang, U. S. Kammula, L. Devillier, R. Carpenter, D. A. Nathan, R. A. Morgan, C. Laurencot, S. A. Rosenberg, B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor–transduced T cells. Blood 119, 2709–2720 (2012). 5. M. V. Maus, S. A. Grupp, D. L. Porter, C. H. June, Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood 123, 2625–2635 (2014). 6. M. S. Topp, N. Gökbuget, G. Zugmaier, E. Degenhard, M. E. Goebeler, M. Klinger, S. A. Neumann, H. A. Horst, T. Raff, A. Viardot, M. Stelljes, M. Schaich, R. Köhne-Volland, M. Brüggemann, O. G. Ottmann, T. Burmeister, P. A. Baeuerle, D. Nagorsen, M. Schmidt, H. Einsele, G. Riethmüller, M. Kneba, D. Hoelzer, P. Kufer, R. C. Bargou, Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood 120, 5185–5187 (2012). 7. M. S. Topp, P. Kufer, N. Gökbuget, M. Goebeler, M. Klinger, S. Neumann, H. A. Horst, T. Raff, A. Viardot, M. Schmid, M. Stelljes, M. Schaich, E. Degenhard, R. Köhne-Volland, M. Brüggemann, O. Ottmann, H. Pfeifer, T. Burmeister, D. Nagorsen, M. Schmidt, R. Lutterbuese, C. Reinhardt, P. A. Baeuerle, M. Kneba, H. Einsele, G. Riethmüller, D. Hoelzer, G. Zugmaier, R. C. Bargou, Targeted therapy with the T-cell–engaging antibody blinatumomab of chemotherapyrefractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J. Clin. Oncol. 29, 2493–2498 (2011). 8. D. Nagorsen, P. Kufer, P. A. Baeuerle, R. Bargou, Blinatumomab: A historical perspective. Pharmacol. Ther. 136, 334–342 (2012). 9. D. T. Teachey, S. R. Rheingold, S. L. Maude, G. Zugmaier, D. M. Barrett, A. E. Seif, K. E. Nichols, E. K. Suppa, M. Kalos, R. A. Berg, J. C. Fitzgerald, R. Aplenc, L. Gore, S. A. Grupp, Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine-directed therapy. Blood 121, 5154–5157 (2013). 10. P. A. Moore, W. Zhang, G. J. Rainey, S. Burke, H. Li, L. Huang, S. Gorlatov, M. C. Veri, S. Aggarwal, Y. Yang, K. Shah, L. Jin, S. Zhang, L. He, T. Zhang, V. Ciccarone, S. Koenig, E. Bonvini, S. Johnson, Application of dual affinity retargeting molecules to achieve optimal redirected T-cell killing of B-cell lymphoma. Blood 117, 4542–4551 (2011). 11. L. M. Portner, K. Schonberg, M. Hejazi, D. Brunnert, F. Neumann, L. Galonska, U. Reusch, M. Little, R. Haas, M. Uhrberg, T and NK cells of B cell NHL patients exert cytotoxicity against lymphoma cells following binding of bispecific tetravalent antibody CD19 × CD3 or CD19 × CD16. Cancer Immunol. Immunother. 61, 1869–1875 (2012). 12. M. Stanglmaier, M. Faltin, P. Ruf, A. Bodenhausen, P. Schröder, H. Lindhofer, Bi20 (FBTA05), a novel trifunctional bispecific antibody (anti-CD20 × anti-CD3), mediates efficient killing of B-cell lymphoma cells even with very low CD20 expression levels. Int. J. Cancer 123, 1181–1189 (2008). 13. P. Strop, W. H. Ho, L. M. Boustany, Y. N. Abdiche, K. C. Lindquist, S. E. Farias, M. Rickert, C. T. Appah, E. Pascua, T. Radcliffe, J. Sutton, J. Chaparro-Riggers, W. Chen, M. G. Casas, S. M. Chin, O. K. Wong, S. H. Liu, G. Vergara, D. Shelton, A. Rajpal, J. Pons, Generating bispecific human IgG1 and IgG2 antibodies from any antibody pair. J. Mol. Biol. 420, 204–219 (2012). 14. C. Spiess, M. Merchant, A. Huang, Z. Zheng, N. Y. Yang, J. Peng, D. Ellerman, W. Shatz, D. Reilly, D. G. Yansura, J. M. Scheer, Bispecific antibodies with natural architecture produced by

www.ScienceTranslationalMedicine.org

13 May 2015

Vol 7 Issue 287 287ra70

9

Downloaded from stm.sciencemag.org on May 14, 2015

Ficoll separation. CD4+T and CD8+T cells were separated with Miltenyi kits according to the manufacturer’s instructions. Cryopreserved PBMCs from CLL patients were purchased from Conversant Bio. Cells were cultured in RPMI 1640 supplemented with 10% fetal bovine serum (SigmaAldrich). For B cell killing assays, live B cells were gated as PI−CD19+ B cells by FACS, and absolute cell count was obtained with fluorescein isothiocyanate beads added to the reaction mix as internal counting control. Activated T cells were detected by CD69 and CD25 surface expression. Intracellular granzyme B induction was detected by FACS. Perforin concentration in medium was detected by ELISA (eBioscience). All antibodies were purchased from BD Biosciences.

15.

16.

17.

18.

19.

20.

21.

22.

23. 24.

25.

26.

27.

28.

29.

30.

co-culture of bacteria expressing two distinct half-antibodies. Nat. Biotechnol. 31, 753–758 (2013). T. T. Junttila, J. Li, J. Johnston, M. Hristopoulos, R. Clark, D. Ellerman, B. E. Wang, Y. Li, M. Mathieu, G. Li, J. Young, E. Luis, P. G. Lewis, E. Stefanich, C. Spiess, A. Polson, B. Irving, J. M. Scheer, M. R. Junttila, M. S. Dennis, R. Kelley, K. Totpal, A. Ebens, Antitumor efficacy of a bispecific antibody that targets HER2 and activates T cells. Cancer Res. 74, 5561–5571 (2014). K. Brischwein, B. Schlereth, B. Guller, C. Steiger, A. Wolf, R. Lutterbuese, S. Offner, M. Locher, T. Urbig, T. Raum, P. Kleindienst, P. Wimberger, R. Kimmig, I. Fichtner, P. Kufer, R. Hofmeister, A. J. da Silva, P. A. Baeuerle, MT110: A novel bispecific single-chain antibody construct with high efficacy in eradicating established tumors. Mol. Immunol. 43, 1129–1143 (2006). B. Schlereth, I. Fichtner, G. Lorenczewski, P. Kleindienst, K. Brischwein, A. da Silva, P. Kufer, R. Lutterbuese, I. Junghahn, S. Kasimir-Bauer, P. Wimberger, R. Kimmig, P. A. Baeuerle, Eradication of tumors from a human colon cancer cell line and from ovarian cancer metastases in immunodeficient mice by a single-chain Ep-CAM-/CD3-bispecific antibody construct. Cancer Res. 65, 2882–2889 (2005). A. dela Hera, U. Muller, C. Olsson, S. Isaaz, A. Tunnacliffe, Structure of the T cell antigen receptor (TCR): Two CD3e subunits in a functional TCR/CD3 complex. J. Exp. Med. 173, 7–17 (1991). Q. Gong, Q. Ou, S. Ye, W. P. Lee, J. Cornelius, L. Diehl, W. Y. Lin, Z. Hu, Y. Lu, Y. Chen, Y. Wu, Y. G. Meng, P. Gribling, Z. Lin, K. Nguyen, T. Tran, Y. Zhang, H. Rosen, F. Martin, A. C. Chan, Importance of cellular microenvironment and circulatory dynamics in B cell immunotherapy. J. Immunol. 174, 817–826 (2005). F. Ishikawa, M. Yasukawa, B. Lyons, S. Yoshida, T. Miyamoto, G. Yoshimoto, T. Watanabe, K. Akashi, L. D. Shultz, M. Harada, Development of functional human blood and immune systems in NOD/SCID/IL2 receptor g chainnull mice. Blood 106, 1565–1573 (2005). J. Golay, M. Lazzari, V. Facchinetti, S. Bernasconi, G. Borleri, T. Barbui, A. Rambaldi, M. Introna, CD20 levels determine the in vitro susceptibility to rituximab and complement of B-cell chronic lymphocytic leukemia: Further regulation by CD55 and CD59. Blood 98, 3383–3389 (2001). D. Huhn, C. von Schilling, M. Wilhelm, A. D. Ho, M. Hallek, R. Kuse, W. Knauf, U. Riedel, A. Hinke, S. Srock, S. Serke, C. Peschel, B. Emmerich; German Chronic Lymphocytic Leukemia Study Group, Rituximab therapy of patients with B-cell chronic lymphocytic leukemia. Blood 98, 1326–1331 (2001). J. C. Riches, J. G. Gribben, Immunomodulation and immune reconstitution in chronic lymphocytic leukemia. Semin. Hematol. 51, 228–234 (2014). J. C. Riches, J. K. Davies, F. McClanahan, R. Fatah, S. Iqbal, S. Agrawal, A. G. Ramsay, J. G. Gribben, T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production. Blood 121, 1612–1621 (2013). A. G. Ramsay, A. J. Clear, R. Fatah, J. G. Gribben, Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: Establishing a reversible immune evasion mechanism in human cancer. Blood 120, 1412–1421 (2012). G. Görgün, T. A. Holderried, D. Zahrieh, D. Neuberg, J. G. Gribben, Chronic lymphocytic leukemia cells induce changes in gene expression of CD4 and CD8 T cells. J. Clin. Invest. 115, 1797–1805 (2005). D. Bagnara, M. S. Kaufman, C. Calissano, S. Marsilio, P. E. Patten, R. Simone, P. Chum, X. J. Yan, S. L. Allen, J. E. Kolitz, S. Baskar, C. Rader, H. Mellstedt, H. Rabbani, A. Lee, P. K. Gregersen, K. R. Rai, N. Chiorazzi, A novel adoptive transfer model of chronic lymphocytic leukemia suggests a key role for T lymphocytes in the disease. Blood 117, 5463–5472 (2011). N. Di Gaetano, E. Cittera, R. Nota, A. Vecchi, V. Grieco, E. Scanziani, M. Botto, M. Introna, J. Golay, Complement activation determines the therapeutic activity of rituximab in vivo. J. Immunol. 171, 1581–1587 (2003). D. M. Goldenberg, E. A. Rossi, R. Stein, T. M. Cardillo, M. S. Czuczman, F. J. Hernandez-Ilizaliturri, H. J. Hansen, C. H. Chang, Properties and structure-function relationships of veltuzumab (hA20), a humanized anti-CD20 monoclonal antibody. Blood 113, 1062–1070 (2009). M. A. Purbhoo, D. J. Irvine, J. B. Huppa, M. M. Davis, T cell killing does not require the formation of a stable mature immunological synapse. Nat. Immunol. 5, 524–530 (2004).

31. D. J. Irvine, M. A. Purbhoo, M. Krogsgaard, M. M. Davis, Direct observation of ligand recognition by T cells. Nature 419, 845–849 (2002). 32. Y. Sykulev, M. Joo, I. Vturina, T. J. Tsomides, H. N. Eisen, Evidence that a single peptide–MHC complex on a target cell can elicit a cytolytic T cell response. Immunity 4, 565–571 (1996). 33. C. Müller, N. Murawski, M. H. Wiesen, G. Held, V. Poeschel, S. Zeynalova, M. Wenger, C. Nickenig, N. Peter, E. Lengfelder, B. Metzner, T. Rixecker, C. Zwick, M. Pfreundschuh, M. Reiser, The role of sex and weight on rituximab clearance and serum elimination half-life in elderly patients with DLBCL. Blood 119, 3276–3284 (2012). 34. M. Klinger, C. Brandl, G. Zugmaier, Y. Hijazi, R. C. Bargou, M. S. Topp, N. Gökbuget, S. Neumann, M. Goebeler, A. Viardot, M. Stelljes, M. Brüggemann, D. Hoelzer, E. Degenhard, D. Nagorsen, P. A. Baeuerle, A. Wolf, P. Kufer, Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell–engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood 119, 6226–6233 (2012). 35. B. Zheng, R. N. Fuji, K. Elkins, S. F. Yu, F. K. Fuh, J. Chuh, C. Tan, J. A. Hongo, H. Raab, K. R. Kozak, M. Williams, E. McDorman, D. Eaton, A. Ebens, A. G. Polson, In vivo effects of targeting CD79b with antibodies and antibody-drug conjugates. Mol. Cancer Ther. 8, 2937–2946 (2009). 36. T. Dreier, P. A. Baeuerle, I. Fichtner, M. Grün, B. Schlereth, G. Lorenczewski, P. Kufer, R. Lutterbüse, G. Riethmüller, P. Gjorstrup, R. C. Bargou, T cell costimulus-independent and very efficacious inhibition of tumor growth in mice bearing subcutaneous or leukemic human B cell lymphoma xenografts by a CD19-/CD3- bispecific single-chain antibody construct. J. Immunol. 170, 4397–4402 (2003). 37. D. Rai, N. L. Pham, J. T. Harty, V. P. Badovinac, Tracking the total CD8 T cell response to infection reveals substantial discordance in magnitude and kinetics between inbred and outbred hosts. J. Immunol. 183, 7672–7681 (2009). 38. F. R. Schuster, M. Stanglmaier, W. Woessmann, B. Winkler, M. Siepermann, R. Meisel, P. G. Schlegel, J. Hess, H. Lindhofer, A. Borkhardt, R. Buhmann, Immunotherapy with the trifunctional anti-CD20 x anti-CD3 antibody FBTA05 (Lymphomun) in paediatric high-risk patients with recurrent CD20positive B cell malignancies. Br. J. Haematol. 169, 90–102 (2015). 39. S. Atwell, J. B. Ridgway, J. A. Wells, P. Carter, Stable heterodimers from remodeling the domain interface of a homodimer using a phage display library. J. Mol. Biol. 270, 26–35 (1997). Acknowledgments: We thank the donors and patients who consented to the use of their cells for the studies; B. Kelley, G. Nakamura, T. Sai, C. Spiess, T. Lombana, A. Yee, and T. Wong for their help in antibody generation; A. Oldendorp for facilitating the studies in cynomolgus monkeys; M. Byrtek for help with statistical analyses; and M. Nicoll and F. De Sauvage for critical reading of the manuscript and helpful discussions. Funding: Supported by Genentech Inc. Author contributions: L.L.S., Y.-W.C., and A.J.E. wrote the manuscript; L.L.S., E.S., H.W., N.D., N.C., T.J., K.T., and A.J.E. designed and supervised the studies; L.L.S., M. Hristopoulos, Y.L., X.Y., C.J., M. Huseni, A.R., E.M., J.Y., R.C., and P.W. executed the studies and analyzed assay data; and L.L.S., A.J.E., D.E., M.M., X.W., X.C., Y.C., J.M.S., and M.S.D. designed and produced the antibodies. Competing interests: L.L.S., M. Hristopoulos, D.E., M.M., Y.L., C.J., M. Huseni, A.R., E.S., E.M., J.Y., X.W., X.C., Y.C., R.C., H.W., N.D., Y-W.C., J.M.S., T.J., K.T., and M.S.D. are all employees of Genentech Inc.; X.Y. and N.C. have no conflict of interest to disclose; A.J.E. was an employee of Genentech at the time the studies were conducted and is currently an employee and shareholder at Juno Therapeutics. Genentech has filed patent applications related to this work on TDBs. Investigators may request materials from Genentech by submitting a request form at www.gene.com/gene/reagents-program/request.do. Submitted 11 December 2014 Accepted 18 March 2015 Published 13 May 2015 10.1126/scitranslmed.aaa4802 Citation: L. L. Sun, D. Ellerman, M. Mathieu, M. Hristopoulos, X. Chen, Y. Li, X. Yan, R. Clark, A. Reyes, E. Stefanich, E. Mai, J. Young, C. Johnson, M. Huseni, X. Wang, Y. Chen, P. Wang, H. Wang, N. Dybdal, Y.-W. Chu, N. Chiorazzi, J. M. Scheer, T. Junttila, K. Totpal, M. S. Dennis, A. J. Ebens, Anti-CD20/CD3 T cell–dependent bispecific antibody for the treatment of B cell malignancies. Sci. Transl. Med. 7, 287ra70 (2015).

www.ScienceTranslationalMedicine.org

13 May 2015

Vol 7 Issue 287 287ra70

10

Downloaded from stm.sciencemag.org on May 14, 2015

RESEARCH ARTICLE

Anti-CD20/CD3 T cell−dependent bispecific antibody for the treatment of B cell malignancies Liping L. Sun et al. Sci Transl Med 7, 287ra70 (2015); DOI: 10.1126/scitranslmed.aaa4802

Editor's Summary

Two-headed cancer therapy

A complete electronic version of this article and other services, including high-resolution figures, can be found at: http://stm.sciencemag.org/content/7/287/287ra70.full.html Supplementary Material can be found in the online version of this article at: http://stm.sciencemag.org/content/suppl/2015/05/11/7.287.287ra70.DC1.html Related Resources for this article can be found online at: http://stm.sciencemag.org/content/scitransmed/4/148/148rv9.full.html http://stm.sciencemag.org/content/scitransmed/2/17/17ps5.full.html Information about obtaining reprints of this article or about obtaining permission to reproduce this article in whole or in part can be found at: http://www.sciencemag.org/about/permissions.dtl

Science Translational Medicine (print ISSN 1946-6234; online ISSN 1946-6242) is published weekly, except the last week in December, by the American Association for the Advancement of Science, 1200 New York Avenue NW, Washington, DC 20005. Copyright 2015 by the American Association for the Advancement of Science; all rights reserved. The title Science Translational Medicine is a registered trademark of AAAS.

Downloaded from stm.sciencemag.org on May 14, 2015

Immunotherapeutic approaches harness either humoral (antibody-mediated) or cellular (T cell−mediated) immunity to fight cancer. Sun et al. combine these approaches by designing a CD3/CD20 TDB (T cell−dependent bispecific), a dual-targeted antibody that recruits T cells to CD20-expressing cells. Their humanized TDB induces T cells to kill primary patient leukemia and lymphoma cells both in vitro and in a mouse model and can deplete CD20-expressing B cells in a macaque model with similar properties as conventional antibodies. If these data hold true in clinical studies, this CD20/CD3 TDB could add to our expanding arsenal of cancer immunotherapeutics.

CD3 T cell-dependent bispecific antibody for the treatment of B cell malignancies.

Bispecific antibodies and antibody fragments in various formats have been explored as a means to recruit cytolytic T cells to kill tumor cells. Encour...
2MB Sizes 7 Downloads 19 Views