Pre-Clinical Development of a Humanized Anti-CD47 Antibody with Anti-Cancer Therapeutic Potential

Jie Liu, Lijuan Wang, Feifei Zhao, Serena Tseng, Cyndhavi Narayanan, Lei Shura, Stephen Willingham, Maureen Howard, Susan Prohaska, Jens Volkmer, Mark Chao, Irving L Weissman, Ravindra Majeti, Jie Liu, Lijuan Wang, Feifei Zhao, Serena Tseng, Cyndhavi Narayanan, Lei Shura, Stephen Willingham, Maureen Howard, Susan Prohaska, Jens Volkmer, Mark Chao, Irving L Weissman, Ravindra Majeti

Abstract

CD47 is a widely expressed cell surface protein that functions as a regulator of phagocytosis mediated by cells of the innate immune system, such as macrophages and dendritic cells. CD47 serves as the ligand for a receptor on these innate immune cells, SIRP-alpha, which in turn delivers an inhibitory signal for phagocytosis. We previously found increased expression of CD47 on primary human acute myeloid leukemia (AML) stem cells, and demonstrated that blocking monoclonal antibodies directed against CD47 enabled the phagocytosis and elimination of AML, non-Hodgkin's lymphoma (NHL), and many solid tumors in xenograft models. Here, we report the development of a humanized anti-CD47 antibody with potent efficacy and favorable toxicokinetic properties as a candidate therapeutic. A novel monoclonal anti-human CD47 antibody, 5F9, was generated, and antibody humanization was carried out by grafting its complementarity determining regions (CDRs) onto a human IgG4 format. The resulting humanized 5F9 antibody (Hu5F9-G4) bound monomeric human CD47 with an 8 nM affinity. Hu5F9-G4 induced potent macrophage-mediated phagocytosis of primary human AML cells in vitro and completely eradicated human AML in vivo, leading to long-term disease-free survival of patient-derived xenografts. Moreover, Hu5F9-G4 synergized with rituximab to eliminate NHL engraftment and cure xenografted mice. Finally, toxicokinetic studies in non-human primates showed that Hu5F9-G4 could be safely administered intravenously at doses able to achieve potentially therapeutic serum levels. Thus, Hu5F9-G4 is actively being developed for and has been entered into clinical trials in patients with AML and solid tumors (ClinicalTrials.gov identifier: NCT02216409).

Conflict of interest statement

Competing Interests: JL, RM, and ILW have filed International Patent Application WO 2011/143624 A2 entitled ‘Humanized and chimeric monoclonal antibodies to CD47’. JL, RM, ILW, SW, JV, MH, and SP have filed U.S. Patent Application number PCT/US2014/018743 entitled ‘Methods for achieving therapeutically effective doses of anti-CD47 agents’. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Cloning, humanization, and characterization of…
Fig 1. Cloning, humanization, and characterization of anti-human CD47 monoclonal antibody 5F9.
(A-B) Comparison of mouse and humanized 5F9 variable heavy (A) and light (B) regions. CDRs are marked as indicated. (C) Rat YB2/0 cells stably transfected with human CD47 and parental YB2/0 cells were stained with human IgG4 isotype control or Hu5F9-G4 and analyzed for surface binding by flow cytometry. (D) Biotinylated mouse 5F9 binding to human CD47/mFc was detected by ELISA in the absence or presence of increasing concentrations of unlabeled Hu5F9-G4 or isotype control. Each sample was assayed in duplicate and SEM values are presented. Indicated p-values were determined by Two-Way ANOVA in comparison. (E) Binding of Hu5F9-G4 to monomeric or dimeric CD47 was analyzed by surface plasmon resonance yielding the indicated traces and binding constants. Data were processed by subtracting responses from the reference surface as well as an average of buffer injections. The responses were globally fit to a 1:1 interaction model including a term for mass transport. The number in parentheses represents the standard error in the last reported digit.
Fig 2. Hu5F9-G4 induces potent macrophage-mediated phagocytosis…
Fig 2. Hu5F9-G4 induces potent macrophage-mediated phagocytosis of AML.
(A) Recombinant SIRPα/human Fc fusion was coated in 96-well plates and CD47/mouse Fc fusion protein was added either in the absence or presence of an equal amount, 5, 10, 25, 50 or 100-times more Hu5F9-G4. The binding activity of CD47 to SIRPα was measured by an HRP-conjugated anti-mouse Fc specific secondary antibody. Each sample was performed in triplicate and SEM values are presented. (B) HL-60 and seven primary human AML cells were labeled with CFSE and incubated with human peripheral blood-derived macrophages in the presence of 10 ug/ml Hu5F9-G4 or isotype control. Two hours later, macrophages were imaged by fluorescence microscopy to determine the phagocytic index (number of target cells ingested per 100 macrophages) in duplicate. Lines indicate mean values and the indicated p-value was determined using a two-sided t-test. (C) The relationship between CD47 receptor occupancy and phagocytosis was determined with primary human AML cells. AML cells were first incubated with increasing concentrations of unlabeled Hu5F9-G4 (0.01–10 ug/ml), and after 2 hours, free CD47 receptor was measured using a saturating concentration (40 ug/ml) of Alexa 488-labeled Hu5F9-G4 followed by flow cytometry. In parallel, the AML cells were evaluated for susceptibility to phagocytosis via addition of human peripheral blood-derived macrophages and determination of the phagocytic index for each condition. (D) HL-60 cells were incubated with increasing concentrations of the indicated isotype control antibodies, chimeric B6H12-IgG1 as a positive control, or Hu5F9-G4, and ADCC activity was determined with human PBMC effector cells. The experiment was repeated 3 times. *** p

Fig 3. Hu5F9-G4 eradicates primary human AML…

Fig 3. Hu5F9-G4 eradicates primary human AML and synergizes with rituximab to eliminate lymphoma.

(A)…

Fig 3. Hu5F9-G4 eradicates primary human AML and synergizes with rituximab to eliminate lymphoma.
(A) Primary human AML cells (5e6) from samples SU028 and SU048 were engrafted into 10 NSG mice per the indicated scheme. Engrafted mice were assigned to treatment with daily injections of either control IgG or Hu5F9-G4 for 2 weeks (n = 5 mice per group). The percent of hCD45+CD33+ leukemic blasts in the bone marrow was determined pre- and post-treatment in all mice by flow cytometry. The indicated p-values were determined by paired t-test. (B) At the end of treatment, mice were monitored for an additional 22 weeks with periodic assessment of human leukemic engraftment in the bone marrow. For the two SU028 mice with recurrence of disease, additional treatment and response is indicated. (C) Kaplan-Meier plot of overall survival of SU048 and SU028 cohorts treated with control IgG or Hu5F9-G4 is indicated. The indicated p-values were derived by log rank test. All mice treated with Hu5F9-G4 were sacrificed on Day 159 for histology and bone marrow analysis. (D) H&E sections of representative mouse bone marrow from mice engrafted with SU028 or SU048 post-treatment with either control IgG or Hu5F9-G4 antibody. (E) Luciferase-labeled Raji cells were transplanted intravenously into NSG mice as a model of disseminated lymphoma. 7 days after transplantation, mice were assigned to daily treatment with control IgG (200 ug), Hu5F9-G4 (100 ug), rituximab (200 ug), or combination of Hu5F9-G4 (100 ug) and rituximab (100 ug) for 21 days. Mice were imaged repeatedly up to more than 200 days to determine the bioluminescent radiance. Relative to the IgG control, p-value of tumor growth inhibition is 0.0013, 0.0065, and 0.0013 on day 21 in Hu5F9-G4-, rituximab-, and combination-treated groups, respectively. Relative to Hu5F9-G4- and rituximab-treated groups, p-value of tumor growth inhibition is 0.0112 and 0.0003 on day 21 in combination-treated groups, respectively. On day 28, p-value of tumor growth inhibition is 0.0059 in Hu5F9-G4 and combination-treated groups. P-values were determined by t-test. (F) Kaplan-Meier plot of overall survival of Raji-engrafted cohorts from panel E. The indicated p-values were derived by log rank test.

Fig 4. Non-human primate pharmacokinetic and toxicology…

Fig 4. Non-human primate pharmacokinetic and toxicology studies show no serious adverse events associated with…

Fig 4. Non-human primate pharmacokinetic and toxicology studies show no serious adverse events associated with Hu5F9-G4.
(A) Individual cynomolgus monkeys were administered single intravenous infusions of Hu5F9-G4 at the indicated doses. The hemoglobin level was monitored over 3 weeks. The shaded bar indicates the range of hemoglobin that might trigger transfusion in humans. (B) Serum Hu5F9-G4 levels were determined from the monkeys dosed in panel A. The shaded bar indicates the range of serum Hu5F9-G4 associated with efficacy against human cancer in xenograft studies. (C) Individual cynomolgus monkeys that received either no pre-treatment (red) or pre-treatment with a single dose of EPO (black) were administered Hu5F9-G4 in a dose escalation study with the doses and time points indicated. Hemoglobin was serially measured to monitor anemia. The shaded bar indicates the range of hemoglobin in humans that might trigger transfusion. (D) Serum Hu5F9-G4 levels were determined from the monkeys dosed in panel C. The shaded bar indicates the range of serum Hu5F9-G4 associated with potent efficacy against primary human AML in xenograft studies. (E) Cynomolgus monkeys were administered a priming dose (PD) on Day 1 of either 1 or 3 mg/kg, followed by once weekly maintenance doses (MD) of 30 mg/kg at the indicated time points. Hemoglobin was serially measured to monitor anemia, and the shaded bar indicates the range of hemoglobin in humans that might require transfusion. (F) Serum Hu5F9-G4 levels were determined from the monkeys dosed in panel E; the shaded bar indicates the range of serum Hu5F9-G4 associated with efficacy against human cancer in xenograft studies. (G) Pharmacokinetic parameters in cynomolgus monkeys dosed in panel E. T1/2: half-life, Tmax: time of Cmax, Cmax: maximum observed concentration, AUC: area under the curve, CL: clearance.
Fig 3. Hu5F9-G4 eradicates primary human AML…
Fig 3. Hu5F9-G4 eradicates primary human AML and synergizes with rituximab to eliminate lymphoma.
(A) Primary human AML cells (5e6) from samples SU028 and SU048 were engrafted into 10 NSG mice per the indicated scheme. Engrafted mice were assigned to treatment with daily injections of either control IgG or Hu5F9-G4 for 2 weeks (n = 5 mice per group). The percent of hCD45+CD33+ leukemic blasts in the bone marrow was determined pre- and post-treatment in all mice by flow cytometry. The indicated p-values were determined by paired t-test. (B) At the end of treatment, mice were monitored for an additional 22 weeks with periodic assessment of human leukemic engraftment in the bone marrow. For the two SU028 mice with recurrence of disease, additional treatment and response is indicated. (C) Kaplan-Meier plot of overall survival of SU048 and SU028 cohorts treated with control IgG or Hu5F9-G4 is indicated. The indicated p-values were derived by log rank test. All mice treated with Hu5F9-G4 were sacrificed on Day 159 for histology and bone marrow analysis. (D) H&E sections of representative mouse bone marrow from mice engrafted with SU028 or SU048 post-treatment with either control IgG or Hu5F9-G4 antibody. (E) Luciferase-labeled Raji cells were transplanted intravenously into NSG mice as a model of disseminated lymphoma. 7 days after transplantation, mice were assigned to daily treatment with control IgG (200 ug), Hu5F9-G4 (100 ug), rituximab (200 ug), or combination of Hu5F9-G4 (100 ug) and rituximab (100 ug) for 21 days. Mice were imaged repeatedly up to more than 200 days to determine the bioluminescent radiance. Relative to the IgG control, p-value of tumor growth inhibition is 0.0013, 0.0065, and 0.0013 on day 21 in Hu5F9-G4-, rituximab-, and combination-treated groups, respectively. Relative to Hu5F9-G4- and rituximab-treated groups, p-value of tumor growth inhibition is 0.0112 and 0.0003 on day 21 in combination-treated groups, respectively. On day 28, p-value of tumor growth inhibition is 0.0059 in Hu5F9-G4 and combination-treated groups. P-values were determined by t-test. (F) Kaplan-Meier plot of overall survival of Raji-engrafted cohorts from panel E. The indicated p-values were derived by log rank test.
Fig 4. Non-human primate pharmacokinetic and toxicology…
Fig 4. Non-human primate pharmacokinetic and toxicology studies show no serious adverse events associated with Hu5F9-G4.
(A) Individual cynomolgus monkeys were administered single intravenous infusions of Hu5F9-G4 at the indicated doses. The hemoglobin level was monitored over 3 weeks. The shaded bar indicates the range of hemoglobin that might trigger transfusion in humans. (B) Serum Hu5F9-G4 levels were determined from the monkeys dosed in panel A. The shaded bar indicates the range of serum Hu5F9-G4 associated with efficacy against human cancer in xenograft studies. (C) Individual cynomolgus monkeys that received either no pre-treatment (red) or pre-treatment with a single dose of EPO (black) were administered Hu5F9-G4 in a dose escalation study with the doses and time points indicated. Hemoglobin was serially measured to monitor anemia. The shaded bar indicates the range of hemoglobin in humans that might trigger transfusion. (D) Serum Hu5F9-G4 levels were determined from the monkeys dosed in panel C. The shaded bar indicates the range of serum Hu5F9-G4 associated with potent efficacy against primary human AML in xenograft studies. (E) Cynomolgus monkeys were administered a priming dose (PD) on Day 1 of either 1 or 3 mg/kg, followed by once weekly maintenance doses (MD) of 30 mg/kg at the indicated time points. Hemoglobin was serially measured to monitor anemia, and the shaded bar indicates the range of hemoglobin in humans that might require transfusion. (F) Serum Hu5F9-G4 levels were determined from the monkeys dosed in panel E; the shaded bar indicates the range of serum Hu5F9-G4 associated with efficacy against human cancer in xenograft studies. (G) Pharmacokinetic parameters in cynomolgus monkeys dosed in panel E. T1/2: half-life, Tmax: time of Cmax, Cmax: maximum observed concentration, AUC: area under the curve, CL: clearance.

References

    1. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144: 646–674. 10.1016/j.cell.2011.02.013
    1. Chao MP, Majeti R, Weissman IL (2012) Programmed cell removal: a new obstacle in the road to developing cancer. Nat Rev Cancer 12: 58–67.
    1. Chao MP, Jaiswal S, Weissman-Tsukamoto R, Alizadeh AA, Gentles AJ, Volkmer J, et al. (2010) Calreticulin Is the Dominant Pro-Phagocytic Signal on Multiple Human Cancers and Is Counterbalanced by CD47. Science Translational Medicine 2.
    1. Estey E, Dohner H (2006) Acute myeloid leukaemia. Lancet 368: 1894–1907.
    1. Lowenberg B, Downing JR, Burnett A (1999) Medical progress—Acute myeloid leukemia. New England Journal of Medicine 341: 1051–1062.
    1. Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs KD, et al. (2009) CD47 Is an Adverse Prognostic Factor and Therapeutic Antibody Target on Human Acute Myeloid Leukemia Stem Cells. Cell 138: 286–299. 10.1016/j.cell.2009.05.045
    1. Majeti R, Chao MP, Alizadeh AA, Pang WW, Weissman IL (2008) CD47 Is An Independent Prognostic Factor and Therapeutic Antibody Target on Human Acute Myeloid Leukemia Stem Cells. Blood 112: 284–284.
    1. Jordan CT, Guzman ML, Noble M (2006) Mechanisms of disease—Cancer stem cells. New England Journal of Medicine 355: 1253–1261.
    1. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, cancer, and cancer stem cells. Nature 414: 105–111.
    1. Majeti R (2010) Monoclonal antibody therapy directed against human acute myeloid leukemia stem cells oncogene 1.
    1. Brown EJ, Frazier WA (2001) Integrin-associated protein (CD47) and its ligands. Trends in Cell Biology 11: 130–135.
    1. Jaiswal S, Jamieson CHM, Pang WW, Park CY, Chao MP, Majeti R, et al. (2009) CD47 Is Upregulated on Circulating Hematopoietic Stem Cells and Leukemia Cells to Avoid Phagocytosis. Cell 138: 271–285. 10.1016/j.cell.2009.05.046
    1. Barclay AN, Brown MH (2006) The SIRP family of receptors and immune regulation. Nature Reviews Immunology 6: 457–464.
    1. Blazar BR, Lindberg FP, Ingulli E, Panoskaltsis-Mortari A, Oldenborg PA, Lizuka K, et al. (2001) CD47 (integrin-associated protein) engagement of dendritic cell and macrophage counterreceptors is required to prevent the clearance of donor lymphohematopoietic cells. Journal of Experimental Medicine 194: 541–549.
    1. Okazawa H, Motegi SI, Ohyama N, Ohnishi H, Tomizawa T, Kaneko Y, et al. (2005) Negative regulation of phagocytosis in macrophages by the CD47-SHPS-1 system. Journal of Immunology 174: 2004–2011.
    1. Oldenborg PA, Gresham HD, Lindberg FP (2001) CD47-signal regulatory protein alpha (SIRP alpha) regulates Fc gamma and complement receptor-mediated phagocytosis. Journal of Experimental Medicine 193: 855–861.
    1. Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD, Lindberg FP. (2000) Role of CD47 as a marker of self on red blood cells. Science 288: 2051–2054.
    1. Okazawa H, Motegi S, Ohyama N, Ohnishi H, Tomizawa T, Kaneko Y, et al. (2005) Negative regulation of phagocytosis in macrophages by the CD47-SHPS-1 system. J Immunol 174: 2004–2011.
    1. Fujioka Y, Matozaki T, Noguchi T, Iwamatsu A, Yamao T, Takahashi N, et al. (1996) A novel membrane glycoprotein, SHPS-1, that binds the SH2-domain-containing protein tyrosine phosphatase SHP-2 in response to mitogens and cell adhesion. Mol Cell Biol 16: 6887–6899.
    1. Kharitonenkov A, Chen Z, Sures I, Wang H, Schilling J, Ullrich A. (1997) A family of proteins that inhibit signalling through tyrosine kinase receptors. Nature 386: 181–186.
    1. Timms JF, Carlberg K, Gu H, Chen H, Kamatkar S, Nadler MJ, et al. (1998) Identification of major binding proteins and substrates for the SH2-containing protein tyrosine phosphatase SHP-1 in macrophages. Mol Cell Biol 18: 3838–3850.
    1. Tsai RK, Discher DE (2008) Inhibition of "self" engulfment through deactivation of myosin-II at the phagocytic synapse between human cells. J Cell Biol 180: 989–1003. 10.1083/jcb.200708043
    1. Oldenborg PA (2004) Role of CD47 in erythroid cells and in autoimmunity. Leuk Lymphoma 45: 1319–1327.
    1. Jaiswal S, Chao MP, Majeti R, Weissman IL (2010) Macrophages as mediators of tumor immunosurveillance. Trends Immunol 31: 212–219. 10.1016/j.it.2010.04.001
    1. Chao MP, Alizadeh AA, Tang C, Myklebust JH, Varghese B, Gill S, et al. (2010) Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 142: 699–713. 10.1016/j.cell.2010.07.044
    1. Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, et al. (2012) The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A 109: 6662–6667. 10.1073/pnas.1121623109
    1. Chao MP, Alizadeh AA, Tang CZ, Myklebust JH, Varghese B, Jan M, et al. (2009) Therapeutic Antibody Targeting of CD47 Synergizes with Rituximab to Completely Eradicate Human B-Cell Lymphoma Xenografts. Blood 114: 1063–1064.
    1. Chao MP, Alizadeh AA, Tang C, Jan M, Weissman-Tsukamoto R, Zhao F, et al. (2011) Therapeutic antibody targeting of CD47 eliminates human acute lymphoblastic leukemia. Cancer Res 71: 1374–1384. 10.1158/0008-5472.CAN-10-2238
    1. Edris B, Weiskopf K, Volkmer AK, Volkmer JP, Willingham SB, Contreras-Trujillo H, et al. (2012) Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma. Proc Natl Acad Sci U S A 109: 6656–6661. 10.1073/pnas.1121629109
    1. Kim D, Wang J, Willingham SB, Martin R, Wernig G, Weissman IL. (2012) Anti-CD47 antibodies promote phagocytosis and inhibit the growth of human myeloma cells. Leukemia 26: 2538–2545. 10.1038/leu.2012.141
    1. Queen C, Schneider WP, Selick HE, Payne PW, Landolfi NF, Duncan JF, et al. (1989) A Humanized Antibody That Binds to the Interleukin-2 Receptor. Proceedings of the National Academy of Sciences of the United States of America 86: 10029–10033.
    1. Angal S, King DJ, Bodmer MW, Turner A, Lawson AD, Roberts G, et al. (1993) A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (IgG4) antibody. Mol Immunol 30: 105–108.
    1. Bloom JW, Madanat MS, Marriott D, Wong T, Chan SY (1997) Intrachain disulfide bond in the core hinge region of human IgG4. Protein Sci 6: 407–415.
    1. Schuurman J, Perdok GJ, Gorter AD, Aalberse RC (2001) The inter-heavy chain disulfide bonds of IgG4 are in equilibrium with intra-chain disulfide bonds. Mol Immunol 38: 1–8.
    1. Kikuchi Y, Uno S, Kinoshita Y, Yoshimura Y, Iida S, Wakahara Y, et al. (2005) Apoptosis inducing bivalent single-chain antibody fragments against CD47 showed antitumor potency for multiple myeloma. Leuk Res 29: 445–450.
    1. Kikuchi Y, Uno S, Yoshimura Y, Otabe K, Iida S, Oheda M, et al. (2004) A bivalent single-chain Fv fragment against CD47 induces apoptosis for leukemic cells. Biochem Biophys Res Commun 315: 912–918.
    1. Mateo V, Lagneaux L, Bron D, Biron G, Armant M, Delespesse G, et al. (1999) CD47 ligation induces caspase-independent cell death in chronic lymphocytic leukemia. Nat Med 5: 1277–1284.
    1. Uno S, Kinoshita Y, Azuma Y, Tsunenari T, Yoshimura Y, Iida S, et al. (2007) Antitumor activity of a monoclonal antibody against CD47 in xenograft models of human leukemia. Oncol Rep 17: 1189–1194.
    1. Hatherley D, Graham SC, Turner J, Harlos K, Stuart DI, Barclay AN. (2008) Paired receptor specificity explained by structures of signal regulatory proteins alone and complexed with CD47. Mol Cell 31: 266–277. 10.1016/j.molcel.2008.05.026
    1. Chao MP, Weissman IL, Majeti R (2012) The CD47-SIRPalpha pathway in cancer immune evasion and potential therapeutic implications. Curr Opin Immunol 24: 225–232. 10.1016/j.coi.2012.01.010
    1. Soto-Pantoja DR, Miller TW, Frazier WA, Roberts DD (2012) Inhibitory signaling through signal regulatory protein-alpha is not sufficient to explain the antitumor activities of CD47 antibodies. Proc Natl Acad Sci U S A 109: E2842; author reply E2844–2845. 10.1073/pnas.1205441109
    1. Kim MJ, Lee JC, Lee JJ, Kim S, Lee SG, Park SW, et al. (2008) Association of CD47 with natural killer cell-mediated cytotoxicity of head-and-neck squamous cell carcinoma lines. Tumour Biol 29: 28–34. 10.1159/000132568
    1. Zhao XW, van Beek EM, Schornagel K, Van der Maaden H, Van Houdt M, Otten MA, et al. (2011) CD47-signal regulatory protein-alpha (SIRPalpha) interactions form a barrier for antibody-mediated tumor cell destruction. Proc Natl Acad Sci U S A 108: 18342–18347. 10.1073/pnas.1106550108
    1. Ravetch JV, Kinet JP (1991) Fc receptors. Annu Rev Immunol 9: 457–492.
    1. Antonelou MH, Kriebardis AG, Papassideri IS (2010) Aging and death signalling in mature red cells: from basic science to transfusion practice. Blood Transfus 8 Suppl 3: s39–47. 10.2450/2010.007S
    1. Burger P, Hilarius-Stokman P, de Korte D, van den Berg TK, van Bruggen R (2012) CD47 functions as a molecular switch for erythrocyte phagocytosis. Blood 119: 5512–5521. 10.1182/blood-2011-10-386805
    1. van Meer PJ, Kooijman M, Brinks V, Gispen-de Wied CC, Silva-Lima B, Moors EH, et al. (2013) Immunogenicity of mAbs in non-human primates during nonclinical safety assessment. MAbs 5: 810–816. 10.4161/mabs.25234

Source: PubMed

3
Prenumerera