Toxicological and pharmacological assessment of AGEN1884, a novel human IgG1 anti-CTLA-4 antibody

Randi B Gombos, Ana Gonzalez, Mariana Manrique, Dhan Chand, David Savitsky, Benjamin Morin, Ekaterina Breous-Nystrom, Christopher Dupont, Rebecca A Ward, Cornelia Mundt, Benjamin Duckless, Hao Tang, Mark A Findeis, Andrea Schuster, Jeremy D Waight, Dennis Underwood, Christopher Clarke, Gerd Ritter, Taha Merghoub, David Schaer, Jedd D Wolchok, Marc van Dijk, Jennifer S Buell, Jean-Marie Cuillerot, Robert Stein, Elise E Drouin, Nicholas S Wilson, Randi B Gombos, Ana Gonzalez, Mariana Manrique, Dhan Chand, David Savitsky, Benjamin Morin, Ekaterina Breous-Nystrom, Christopher Dupont, Rebecca A Ward, Cornelia Mundt, Benjamin Duckless, Hao Tang, Mark A Findeis, Andrea Schuster, Jeremy D Waight, Dennis Underwood, Christopher Clarke, Gerd Ritter, Taha Merghoub, David Schaer, Jedd D Wolchok, Marc van Dijk, Jennifer S Buell, Jean-Marie Cuillerot, Robert Stein, Elise E Drouin, Nicholas S Wilson

Abstract

CTLA-4 and CD28 exemplify a co-inhibitory and co-stimulatory signaling axis that dynamically sculpts the interaction of antigen-specific T cells with antigen-presenting cells. Anti-CTLA-4 antibodies enhance tumor-specific immunity through a variety of mechanisms including: blockade of CD80 or CD86 binding to CTLA-4, repressing regulatory T cell function and selective elimination of intratumoral regulatory T cells via an Fcγ receptor-dependent mechanism. AGEN1884 is a novel IgG1 antibody targeting CTLA-4. It potently enhanced antigen-specific T cell responsiveness that could be potentiated in combination with other immunomodulatory antibodies. AGEN1884 was well-tolerated in non-human primates and enhanced vaccine-mediated antigen-specific immunity. AGEN1884 combined effectively with PD-1 blockade to elicit a T cell proliferative response in the periphery. Interestingly, an IgG2 variant of AGEN1884 revealed distinct functional differences that may have implications for optimal dosing regimens in patients. Taken together, the pharmacological properties of AGEN1884 support its clinical investigation as a single therapeutic and combination agent.

Conflict of interest statement

Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: Randi B. Gombos, Ana Gonzalez, Mariana Manrique, Dhan Chand, David Savitsky, Benjamin Morin, Ekaterina Breous-Nystrom, Christopher Dupont, Rebecca A. Ward, Mark A. Findeis, Jeremy D. Waight, Dennis Underwood, Christopher Clarke, Marc van Dijk, Jennifer S. Buell, Jean-Marie Cuillerot, Robert Stein, Elise E. Drouin and Nicholas S. Wilson have ownership of equity securities and/or are currently employed by Agenus Inc. or a subsidiary thereof. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. AGEN1884 binds CTLA-4 and blocks…
Fig 1. AGEN1884 binds CTLA-4 and blocks CTLA-4 from interacting with CD80 and CD86.
(A) Binding of fluorescently-labeled CD80-Fc or CD86-Fc (1 nM) in the presence of increasing concentrations of AGEN1884 or an IgG1 isotype control. Binding to CTLA-4-linked microspheres was assessed using Luminex. (B) CTLA-4-expressing CHO cells were pre-incubated with increasing concentrations of AGEN1884 or an IgG1 isotype control followed by the addition of a fixed concentration of fluorescently-labeled CD80-Fc or CD86-Fc (0.625 μg/ml). Binding of CD80-Fc or CD86-Fc to the CHO cells was assessed by flow cytometry. (C-D) AGEN1884 binding to a (C) Jurkat cell line genetically engineered to express human CTLA-4 or (D) wildtype (CTLA-4-negative) Jurkat cell line. Expression of CD28 was also assessed using an anti-CD28 antibody (empty histogram) compared to an isotype control (filled histogram) on the (E) CTLA-4-expressing and (F) wildtype (CTLA-4-negative) cells lines. (A-D) Representative data from one of three experiments indicate the mean ± SEM in each treatment group.
Fig 2. AGEN1884 epitope.
Fig 2. AGEN1884 epitope.
(A) Mapping by hydrogen-deuterium exchange mass spectrometry (HDX-MS) on the CTLA-4 structure (PDB 1I8L). Ribbon representation of CTLA-4 highlighting residues identified as having reduced HDX by HDX-MS: residues 80–82 (QVT, magenta), 135–139 (YPPPY, cyan), 140–141 (YL, dark blue), 142–149 (GIGNGTQI, pale blue). (B) Structure of the human co-stimulatory complex CD80 (grey)/CTLA-4 (yellow) (PDB 1I8L). Residues having reduced HDX are indicated as in panel (A). The view of the structure highlights the loop region comprised of residues at positions 135–149, which encompasses a turn-loop that directly interacts with CD80. (C) Sequence alignment between human and cynomolgus macaque CTLA-4. An asterisk indicates identical residues, a colon indicates conservation between groups of strongly similar properties and a period indicates conservation between groups of weakly similar properties. Solid line boxes indicate residues identified as having reduced HDX.
Fig 3. AGEN1884 increases T cell activation…
Fig 3. AGEN1884 increases T cell activation alone and in combination with other immunomodulatory antibodies.
(A) A Jurkat T cell line genetically engineered to express CTLA-4 with an IL-2-dependent luciferase reporter was co-cultured with CD80/CD86-positive artificial APCs in the presence of increasing concentrations of AGEN1884 or an isotype control antibody. The fold increase in relative light units (RLU) relative to baseline is shown. (B) CTLA-4 expression was measured on T cells on days 1, 2, 4, 8 and 10 following stimulation with a sub-maximal concentration of SEA peptide (100 ng/mL). Primary human PBMC were stimulated with a sub-maximal concentration of the SEA peptide (100 ng/mL) and (C) increasing doses of AGEN1884, (D) single dose of AGEN1884 (10 μg/mL), (E) AGEN1884 (10 mg/mL) ± nivolumab (anti-PD-1 antagonist antibody), pembrolizumab (anti-PD-1 antagonist antibody), 25F7 (anti-LAG-3 antagonist antibody), 10C7 (anti-CD137 agonist antibody) (10 mg/mL) or (F) AGEN1884 (10 mg/mL) ± AGEN2034 (anti-PD-1 antagonist antibody) (10 mg/mL). Replicate cell supernatants were collected after 5 days for measurement of IL-2. Representative data from one of three experiments indicate the mean ± SEM in each treatment group (n = 3). (D-F) Data were analyzed using a Student’s t-test. Significant differences depicted were p

Fig 4. Fcγ receptor signaling and antibody-dependent…

Fig 4. Fcγ receptor signaling and antibody-dependent cellular cytotoxicity of CTLA-4-expressing cells with AGEN1884.

(A-C)…

Fig 4. Fcγ receptor signaling and antibody-dependent cellular cytotoxicity of CTLA-4-expressing cells with AGEN1884.
(A-C) Jurkat cell lines genetically engineered to express FcγRs upstream of a NFAT-dependent luciferase reporter were co-cultured with CTLA-4-expressing cells and increasing doses of AGEN1884 or an isotype control. Signaling through (A) FcγRIIIA-V158-, (B) FcγRIIIA-F158-, or (C) FcγRIIA-H131-expressing cells was assessed based upon luciferase expression, which is shown as relative light units (RLU). (D) The cytotoxicity of CTLA-4high Jurkat cells by CD16-expressing NK-92 cells based on lactate dehydrogenase (LDH) release in the presence of increasing concentrations of AGEN1884 or an isotype control antibody. (E-F) Extracellular CTLA-4 expression (empty histogram) compared to the isotype control (filled histogram) was measured on (E) CD3+FoxP3+ or (F) CD3+FoxP3- cells. The lysis of (G) CTLA-4highCD3+FoxP3+ target cells or (H) CTLA-4lowCD3+FoxP3- target cells by primary NK cells in the presence of increasing concentrations of AGEN1884 compared to an isotype control. Representative data from one of three experiments indicate the mean ± SEM in each treatment group (n = 2–3). (G, H) Data were analyzed using a Student’s t-test for each dose of AGEN1884 compared to the isotype. Significant differences depicted were p<0.05 (*).

Fig 5. Pharmacokinetic profile of AGEN1884 and…

Fig 5. Pharmacokinetic profile of AGEN1884 and T cell-dependent antibody response and immune cell response…

Fig 5. Pharmacokinetic profile of AGEN1884 and T cell-dependent antibody response and immune cell response to systemic co-administration of AGEN1884 with an ENGERIX-B® and KLH vaccine in cynomolgus macaques.
AGEN1884 titers were measured from serum samples collected from five groups of animals (n = 3 per group) following (A) a single intravenous (IV) dose at either 3 mg/kg or 100 mg/kg or (B) five weekly IV doses at 5 mg/kg, 30 mg/kg, or 100 mg/kg. Cynomolgus macaques (n = 6 per group) were given 10 mg/kg of AGEN1884 via IV administration with an ENGERIX-B® and KLH vaccine on days 1 and 29. Duplicate samples were analyzed for (C) anti-HBsAg-specific IgG serum titers or (D) the frequency of IFN-γ-producing HBsAg-specific cells in PBMC based on the number of spot forming units (SFU) per million PBMC. Anti-KLH (E) IgG and (F) IgM serum titers were also measured.

Fig 6. AGEN1884 in combination with anti-PD-1…

Fig 6. AGEN1884 in combination with anti-PD-1 further potentiates T cell proliferation in vivo .

Cynomolgus…

Fig 6. AGEN1884 in combination with anti-PD-1 further potentiates T cell proliferation in vivo.
Cynomolgus macaques (n = 6 per group) were administered 3 mg/kg of nivolumab alone or in combination with 10 mg/kg of AGEN1884 via intravenous (IV) infusion. Duplicate samples of PBMC were analyzed for Ki67 expression in CD4+ and CD8+ T cells at 7 days prior to treatment or 3, 10, 14, 18 and 22 days after treatment. (A-B) Representative dot plots of CD4+ and CD8+ T cells isolated from PBMC from cynomolgus macaques given (A) 3 mg/kg of nivolumab alone or (B) a combination of 10 mg/kg of AGEN1884 with 3 mg/kg of nivolumab. (C) Representative dot plots of naïve, central memory and effector memory CD4+ and CD8+ T cells isolated from PBMC from cynomolgus macaques given a combination of 10 mg/kg of AGEN1884 with 3 mg/kg of nivolumab.

Fig 7. Fcγ receptor signaling and antibody-dependent…

Fig 7. Fcγ receptor signaling and antibody-dependent cellular cytotoxicity of CTLA-4-expressing cells with AGEN2041 and…

Fig 7. Fcγ receptor signaling and antibody-dependent cellular cytotoxicity of CTLA-4-expressing cells with AGEN2041 and increased T cell activation in the presence of AGEN2041.
(A-C) Jurkat cell lines genetically engineered to express FcγRs upstream of a NFAT-dependent luciferase reporter were co-cultured with CTLA-4-expressing cells and increasing doses of AGEN2041 or an isotype control. Signaling through (A) FcγRIIIA-V158-, (B) FcγRIIIA-F158-, or (C) FcγRIIA-H131-expressing cells was assessed based upon luciferase expression which is shown as relative light units (RLU). (D) The cytotoxicity of CTLA-4high Jurkat cells by CD16-expressing NK-92 cells based on lactate dehydrogenase (LDH) release in the presence of increasing concentrations of AGEN2041, AGEN1884 (positive control), or an IgG2 isotype control antibody. (E-F) Primary human PBMC were stimulated with a sub-maximal concentration of the SEA peptide (100 ng/mL) and increasing doses of (E) AGEN2041 versus (F) AGEN1884. Cell supernatants were collected after 5 days for measurement of IL-2. Representative data indicate the mean ± SEM in each treatment group (n = ≥2).
All figures (7)
Fig 4. Fcγ receptor signaling and antibody-dependent…
Fig 4. Fcγ receptor signaling and antibody-dependent cellular cytotoxicity of CTLA-4-expressing cells with AGEN1884.
(A-C) Jurkat cell lines genetically engineered to express FcγRs upstream of a NFAT-dependent luciferase reporter were co-cultured with CTLA-4-expressing cells and increasing doses of AGEN1884 or an isotype control. Signaling through (A) FcγRIIIA-V158-, (B) FcγRIIIA-F158-, or (C) FcγRIIA-H131-expressing cells was assessed based upon luciferase expression, which is shown as relative light units (RLU). (D) The cytotoxicity of CTLA-4high Jurkat cells by CD16-expressing NK-92 cells based on lactate dehydrogenase (LDH) release in the presence of increasing concentrations of AGEN1884 or an isotype control antibody. (E-F) Extracellular CTLA-4 expression (empty histogram) compared to the isotype control (filled histogram) was measured on (E) CD3+FoxP3+ or (F) CD3+FoxP3- cells. The lysis of (G) CTLA-4highCD3+FoxP3+ target cells or (H) CTLA-4lowCD3+FoxP3- target cells by primary NK cells in the presence of increasing concentrations of AGEN1884 compared to an isotype control. Representative data from one of three experiments indicate the mean ± SEM in each treatment group (n = 2–3). (G, H) Data were analyzed using a Student’s t-test for each dose of AGEN1884 compared to the isotype. Significant differences depicted were p<0.05 (*).
Fig 5. Pharmacokinetic profile of AGEN1884 and…
Fig 5. Pharmacokinetic profile of AGEN1884 and T cell-dependent antibody response and immune cell response to systemic co-administration of AGEN1884 with an ENGERIX-B® and KLH vaccine in cynomolgus macaques.
AGEN1884 titers were measured from serum samples collected from five groups of animals (n = 3 per group) following (A) a single intravenous (IV) dose at either 3 mg/kg or 100 mg/kg or (B) five weekly IV doses at 5 mg/kg, 30 mg/kg, or 100 mg/kg. Cynomolgus macaques (n = 6 per group) were given 10 mg/kg of AGEN1884 via IV administration with an ENGERIX-B® and KLH vaccine on days 1 and 29. Duplicate samples were analyzed for (C) anti-HBsAg-specific IgG serum titers or (D) the frequency of IFN-γ-producing HBsAg-specific cells in PBMC based on the number of spot forming units (SFU) per million PBMC. Anti-KLH (E) IgG and (F) IgM serum titers were also measured.
Fig 6. AGEN1884 in combination with anti-PD-1…
Fig 6. AGEN1884 in combination with anti-PD-1 further potentiates T cell proliferation in vivo.
Cynomolgus macaques (n = 6 per group) were administered 3 mg/kg of nivolumab alone or in combination with 10 mg/kg of AGEN1884 via intravenous (IV) infusion. Duplicate samples of PBMC were analyzed for Ki67 expression in CD4+ and CD8+ T cells at 7 days prior to treatment or 3, 10, 14, 18 and 22 days after treatment. (A-B) Representative dot plots of CD4+ and CD8+ T cells isolated from PBMC from cynomolgus macaques given (A) 3 mg/kg of nivolumab alone or (B) a combination of 10 mg/kg of AGEN1884 with 3 mg/kg of nivolumab. (C) Representative dot plots of naïve, central memory and effector memory CD4+ and CD8+ T cells isolated from PBMC from cynomolgus macaques given a combination of 10 mg/kg of AGEN1884 with 3 mg/kg of nivolumab.
Fig 7. Fcγ receptor signaling and antibody-dependent…
Fig 7. Fcγ receptor signaling and antibody-dependent cellular cytotoxicity of CTLA-4-expressing cells with AGEN2041 and increased T cell activation in the presence of AGEN2041.
(A-C) Jurkat cell lines genetically engineered to express FcγRs upstream of a NFAT-dependent luciferase reporter were co-cultured with CTLA-4-expressing cells and increasing doses of AGEN2041 or an isotype control. Signaling through (A) FcγRIIIA-V158-, (B) FcγRIIIA-F158-, or (C) FcγRIIA-H131-expressing cells was assessed based upon luciferase expression which is shown as relative light units (RLU). (D) The cytotoxicity of CTLA-4high Jurkat cells by CD16-expressing NK-92 cells based on lactate dehydrogenase (LDH) release in the presence of increasing concentrations of AGEN2041, AGEN1884 (positive control), or an IgG2 isotype control antibody. (E-F) Primary human PBMC were stimulated with a sub-maximal concentration of the SEA peptide (100 ng/mL) and increasing doses of (E) AGEN2041 versus (F) AGEN1884. Cell supernatants were collected after 5 days for measurement of IL-2. Representative data indicate the mean ± SEM in each treatment group (n = ≥2).

References

    1. Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, Sharpe AH. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3(5):541–7. .
    1. Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, et al. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science. 1995;270(5238):985–8. .
    1. Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT, et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science. 2014;345(6204):1623–7. Epub 2014/09/13. doi: . ; PubMed Central PMCID: PMCPMC4371526.
    1. Wilson NS, Villadangos JA. Regulation of antigen presentation and cross-presentation in the dendritic cell network: facts, hypothesis, and immunological implications. Adv Immunol. 2005;86:241–305. doi: . .
    1. Acuto O, Michel F. CD28-mediated co-stimulation: a quantitative support for TCR signalling. Nat Rev Immunol. 2003;3(12):939–51. doi: . .
    1. Egen JG, Allison JP. Cytotoxic T lymphocyte antigen-4 accumulation in the immunological synapse is regulated by TCR signal strength. Immunity. 2002;16(1):23–35. .
    1. Valk E, Leung R, Kang H, Kaneko K, Rudd CE, Schneider H. T cell receptor-interacting molecule acts as a chaperone to modulate surface expression of the CTLA-4 coreceptor. Immunity. 2006;25(5):807–21. doi:
    1. van der Merwe PA, Bodian DL, Daenke S, Linsley P, Davis SJ. CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics. J Exp Med. 1997;185(3):393–403. ; PubMed Central PMCID: PMC2196039.
    1. Xu Z, Juan V, Ivanov A, Ma Z, Polakoff D, Powers DB, et al. Affinity and cross-reactivity engineering of CTLA4-Ig to modulate T cell costimulation. J Immunol. 2012;189(9):4470–7. doi: . .
    1. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science. 2008;322(5899):271–5. doi: . .
    1. Qureshi OS, Zheng Y, Nakamura K, Attridge K, Manzotti C, Schmidt EM, et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science. 2011;332(6029):600–3. doi: . ; PubMed Central PMCID: PMC3198051.
    1. Tai X, Van Laethem F, Pobezinsky L, Guinter T, Sharrow SO, Adams A, et al. Basis of CTLA-4 function in regulatory and conventional CD4(+) T cells. Blood. 2012;119(22):5155–63. doi: . ; PubMed Central PMCID: PMCPMC3369608.
    1. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A. 2010;107(9):4275–80. doi: . ; PubMed Central PMCID: PMC2840093.
    1. Grosso JF, Jure-Kunkel MN. CTLA-4 blockade in tumor models: an overview of preclinical and translational research. Cancer Immun. 2013;13:5 ; PubMed Central PMCID: PMC3559193.
    1. Bulliard Y, Jolicoeur R, Windman M, Rue SM, Ettenberg S, Knee DA, et al. Activating Fc gamma receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J Exp Med. 2013;210(9):1685–93. doi: . ; PubMed Central PMCID: PMC3754864.
    1. Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T, Srinivasan M, et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res. 2013;1(1):32–42. doi: . .
    1. Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210(9):1695–710. doi: . ; PubMed Central PMCID: PMC3754863.
    1. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371(23):2189–99. doi: . ; PubMed Central PMCID: PMC4315319.
    1. Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, et al. Pooled Analysis of Long-Term Survival Data From Phase II and Phase III Trials of Ipilimumab in Unresectable or Metastatic Melanoma. J Clin Oncol. 2015;33(17):1889–94. doi: . .
    1. Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–17. doi: . .
    1. Robert L, Harview C, Emerson R, Wang X, Mok S, Homet B, et al. Distinct immunological mechanisms of CTLA-4 and PD-1 blockade revealed by analyzing TCR usage in blood lymphocytes. Oncoimmunology. 2014;3:e29244 Epub 2014/08/02. doi: . ; PubMed Central PMCID: PMCPMC4108466.
    1. Romano E, Kusio-Kobialka M, Foukas PG, Baumgaertner P, Meyer C, Ballabeni P, et al. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A. 2015;112(19):6140–5. doi: . ; PubMed Central PMCID: PMC4434760.
    1. Boutros C, Tarhini A, Routier E, Lambotte O, Ladurie FL, Carbonnel F, et al. Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev Clin Oncol. 2016;13(8):473–86. doi: . .
    1. Callahan MK, Postow MA, Wolchok JD. CTLA-4 and PD-1 Pathway Blockade: Combinations in the Clinic. Front Oncol. 2014;4:385 doi: . ; PubMed Central PMCID: PMC4295550.
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med. 2015;373(1):23–34. doi: . .
    1. Breous-Nystrom E, Schultze K, Meier M, Flueck L, Holzer C, Boll M, et al. Retrocyte Display(R) technology: generation and screening of a high diversity cellular antibody library. Methods. 2014;65(1):57–67. doi: . .
    1. Van Dijk MA, Mundt CA, Ritter G, Schaer DA, Wolchok JD, Merghoub T, et al., inventors; Agenus Inc.; Ludwig Institute for Cancer Research, Ltd; Memorial Sloan Kettering Cancer Center, assignee. Anti-ctla-4 antibodies and methods of use thereof. United States Patent Application Publication No. 20160368989. 2016 Dec 22.
    1. Van Dijk MA, Mundt CA, Ritter G, Wolchok JD, Merghoub T, Zappasodi R, et al., inventors; Agenus Inc; Ludwig Institute for Cancer Research, Ltd; Memorial Sloan Kettering Cancer Center, assignee. Anti-pd-1 antibodies and methods of use thereof. United States Patent Application Publication No. 20170081409. 2017 Mar 23.
    1. Spindler N, Diestel U, Stump JD, Wiegers AK, Winkler TH, Sticht H, et al. Structural basis for the recognition of human cytomegalovirus glycoprotein B by a neutralizing human antibody. PLoS Pathog. 2014;10(10):e1004377 doi: . ; PubMed Central PMCID: PMC4192593.
    1. Grawunder U, Mach M, Martin-Parras L, Potzsch S, Spindler N, Sticht H, et al., inventors; 4-Antibody AG, assignee. Binding members for human cytomegalovirus. United States Patent No. 9346874. 2016 May 24.
    1. Thudium K, Selby M, Zens KD, Yamanaka M, Korman A, Leblanc H, inventors; Medarex, Inc, assignee. Human antibodies that bind lymphocyte activation gene-3 (lag-3) and uses thereof. United States Patent Application Publication No. 20110150892. 2011 Jun 23.
    1. Jure-Kunkel M, Hefta LJ, Santoro M, Ganguly S, Halk EL, inventors; Bristol-Myers Squibb Company, assignee. Fully human antibodies against human 4-1bb. United States Patent No. 7288638. 2007 Oct 30.
    1. Petersson K, Thunnissen M, Forsberg G, Walse B. Crystal structure of a SEA variant in complex with MHC class II reveals the ability of SEA to crosslink MHC molecules. Structure. 2002;10(12):1619–26. .
    1. Levy R, Rotfogel Z, Hillman D, Popugailo A, Arad G, Supper E, et al. Superantigens hyperinduce inflammatory cytokines by enhancing the B7-2/CD28 costimulatory receptor interaction. Proc Natl Acad Sci U S A. 2016;113(42):E6437–E46. doi: . ; PubMed Central PMCID: PMC5081635.
    1. Moreau T, Coles A, Wing M, Isaacs J, Hale G, Waldmann H, et al. Transient increase in symptoms associated with cytokine release in patients with multiple sclerosis. Brain. 1996;119 (Pt 1):225–37. .
    1. Curti BD, Kovacsovics-Bankowski M, Morris N, Walker E, Chisholm L, Floyd K, et al. OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Res. 2013;73(24):7189–98. doi: . ; PubMed Central PMCID: PMC3922072.
    1. Sharpe AH, Freeman GJ. The B7-CD28 superfamily. Nat Rev Immunol. 2002;2(2):116–26. doi: . .
    1. Azuma M, Ito D, Yagita H, Okumura K, Phillips JH, Lanier LL, et al. B70 antigen is a second ligand for CTLA-4 and CD28. Nature. 1993;366(6450):76–9. doi: . .
    1. Freedman AS, Freeman GJ, Rhynhart K, Nadler LM. Selective induction of B7/BB-1 on interferon-gamma stimulated monocytes: a potential mechanism for amplification of T cell activation through the CD28 pathway. Cell Immunol. 1991;137(2):429–37. .
    1. Hathcock KS, Laszlo G, Dickler HB, Bradshaw J, Linsley P, Hodes RJ. Identification of an alternative CTLA-4 ligand costimulatory for T cell activation. Science. 1993;262(5135):905–7. .
    1. Stamper CC, Zhang Y, Tobin JF, Erbe DV, Ikemizu S, Davis SJ, et al. Crystal structure of the B7-1/CTLA-4 complex that inhibits human immune responses. Nature. 2001;410(6828):608–11. doi: . .
    1. Garcia-Sanz JA, Lenig D. Translational control of interleukin 2 messenger RNA as a molecular mechanism of T cell anergy. J Exp Med. 1996;184(1):159–64. ; PubMed Central PMCID: PMC2192667.
    1. Guntermann C, Alexander DR. CTLA-4 suppresses proximal TCR signaling in resting human CD4(+) T cells by inhibiting ZAP-70 Tyr(319) phosphorylation: a potential role for tyrosine phosphatases. J Immunol. 2002;168(9):4420–9. .
    1. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369(2):122–33. doi: . .
    1. Das R, Verma R, Sznol M, Boddupalli CS, Gettinger SN, Kluger H, et al. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 2015;194(3):950–9. doi: . ; PubMed Central PMCID: PMC4380504.
    1. Sznol M, Hodi FS, Margolin K, McDermott D, Ernstoff MS, Kirkwood JM, et al. Phase I study of BMS-663513, a fully human anti-CD137 agonist monoclonal antibody, in patients (pts) with advanced cancer (CA) J Clin Oncol. 2008;26(15_suppl):3007.
    1. Marabelle A, Kohrt H, Sagiv-Barfi I, Ajami B, Axtell RC, Zhou G, et al. Depleting tumor-specific Tregs at a single site eradicates disseminated tumors. J Clin Invest. 2013;123(6):2447–63. doi: . ; PubMed Central PMCID: PMC3668834.
    1. Kim JM, Ashkenazi A. Fcgamma receptors enable anticancer action of proapoptotic and immune-modulatory antibodies. J Exp Med. 2013;210(9):1647–51. doi: . ; PubMed Central PMCID: PMC3754862.
    1. Oflazoglu E, Stone IJ, Brown L, Gordon KA, van Rooijen N, Jonas M, et al. Macrophages and Fc-receptor interactions contribute to the antitumour activities of the anti-CD40 antibody SGN-40. Br J Cancer. 2009;100(1):113–7. doi: . ; PubMed Central PMCID: PMC2634668.
    1. Chung S, Lin YL, Reed C, Ng C, Cheng ZJ, Malavasi F, et al. Characterization of in vitro antibody-dependent cell-mediated cytotoxicity activity of therapeutic antibodies—impact of effector cells. J Immunol Methods. 2014;407:63–75. doi: . .
    1. Leidi M, Gotti E, Bologna L, Miranda E, Rimoldi M, Sica A, et al. M2 macrophages phagocytose rituximab-opsonized leukemic targets more efficiently than m1 cells in vitro. J Immunol. 2009;182(7):4415–22. doi: . .
    1. Finco D, Grimaldi C, Fort M, Walker M, Kiessling A, Wolf B, et al. Cytokine release assays: current practices and future directions. Cytokine. 2014;66(2):143–55. doi: . .
    1. Vessillier S, Eastwood D, Fox B, Sathish J, Sethu S, Dougall T, et al. Cytokine release assays for the prediction of therapeutic mAb safety in first-in man trials—Whole blood cytokine release assays are poorly predictive for TGN1412 cytokine storm. J Immunol Methods. 2015;424:43–52. doi: . ; PubMed Central PMCID: PMC4768082.
    1. Piccotti JR, Alvey JD, Reindel JF, Guzman RE. T-cell-dependent antibody response: assay development in cynomolgus monkeys. J Immunotoxicol. 2005;2(4):191–6. doi: . .
    1. Antonia S, Goldberg SB, Balmanoukian A, Chaft JE, Sanborn RE, Gupta A, et al. Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol. 2016;17(3):299–308. doi: . .
    1. Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown. J Cell Physiol. 2000;182(3):311–22. doi: . .
    1. Buchbinder EI, Desai A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am J Clin Oncol. 2016;39(1):98–106. doi: . ; PubMed Central PMCID: PMC4892769.
    1. Eagar TN, Karandikar NJ, Bluestone JA, Miller SD. The role of CTLA-4 in induction and maintenance of peripheral T cell tolerance. Eur J Immunol. 2002;32(4):972–81. doi: . .
    1. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J Med. 2012;366(26):2517–9. doi: . .
    1. Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017;355(6332):1428–33. doi: . .
    1. Hellmann MD, Rizvi NA, Goldman JW, Gettinger SN, Borghaei H, Brahmer JR, et al. Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study. Lancet Oncol. 2017;18(1):31–41. doi: . .
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74. doi: . .
    1. Kvistborg P, Philips D, Kelderman S, Hageman L, Ottensmeier C, Joseph-Pietras D, et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med. 2014;6(254):254ra128 doi: . .
    1. Montler R, Bell RB, Thalhofer C, Leidner R, Feng Z, Fox BA, et al. OX40, PD-1 and CTLA-4 are selectively expressed on tumor-infiltrating T cells in head and neck cancer. Clin Transl Immunology. 2016;5(4):e70 doi: . ; PubMed Central PMCID: PMC4855266.
    1. Bulliard Y, Jolicoeur R, Zhang J, Dranoff G, Wilson NS, Brogdon JL. OX40 engagement depletes intratumoral Tregs via activating FcgammaRs, leading to antitumor efficacy. Immunol Cell Biol. 2014;92(6):475–80. doi: . .
    1. Wilson NS, Yang B, Yang A, Loeser S, Marsters S, Lawrence D, et al. An Fcgamma receptor-dependent mechanism drives antibody-mediated target-receptor signaling in cancer cells. Cancer Cell. 2011;19(1):101–13. doi: . .
    1. Brezski RJ, Oberholtzer A, Strake B, Jordan RE. The in vitro resistance of IgG2 to proteolytic attack concurs with a comparative paucity of autoantibodies against peptide analogs of the IgG2 hinge. MAbs. 2011;3(6):558–67. doi: . ; PubMed Central PMCID: PMC3242842.
    1. Trist HM, Tan PS, Wines BD, Ramsland PA, Orlowski E, Stubbs J, et al. Polymorphisms and interspecies differences of the activating and inhibitory FcgammaRII of Macaca nemestrina influence the binding of human IgG subclasses. J Immunol. 2014;192(2):792–803. doi: . ; PubMed Central PMCID: PMC4080885.
    1. Warncke M, Calzascia T, Coulot M, Balke N, Touil R, Kolbinger F, et al. Different adaptations of IgG effector function in human and nonhuman primates and implications for therapeutic antibody treatment. J Immunol. 2012;188(9):4405–11. doi: . .
    1. Funt SA, Page DB, Wolchok JD, Postow MA. CTLA-4 antibodies: new directions, new combinations. Oncology (Williston Park). 2014;28 Suppl 3:6–14. .
    1. Le Mercier I, Lines JL, Noelle RJ. Beyond CTLA-4 and PD-1, the Generation Z of Negative Checkpoint Regulators. Front Immunol. 2015;6:418 doi: . ; PubMed Central PMCID: PMC4544156.

Source: PubMed

3
Abonner