Immunocytokine

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 360 Experts worldwide ranked by ideXlab platform

Dario Neri - One of the best experts on this subject based on the ideXlab platform.

  • the Immunocytokine l19 tnf eradicates sarcomas in combination with chemotherapy agents or with immune check point inhibitors
    Anti-Cancer Drugs, 2020
    Co-Authors: Riccardo Corbellari, Dario Neri, Alessandra Villa, Lisa Nadal, Roberto De Luca
    Abstract:

    Antibody-cytokine fusion proteins (also called 'Immunocytokines') represent an emerging class of biopharmaceutical products, which are being considered for cancer immunotherapy. When used as single agents, pro-inflammatory Immunocytokines are rarely capable of inducing complete and durable cancer regression in mouse models and in patients. However, the combination treatment with conventional chemotherapy or with other immune-stimulatory agents typically increases the therapeutic efficacy of Immunocytokines. In this article, we describe combination treatments of a tumor-targeting antibody-cytokine fusion protein based on the L19 antibody (specific to a splice isoform of fibronectin) fused to murine tumor necrosis factor with standard chemotherapy (dacarbazine, trabectedin or melphalan) or with an immune check-point inhibitor (anti-PD-1) in a BALB/c derived immunocompetent murine model of sarcoma (WEHI-164). All combination treatments led to improved tumor remission compared to single-agent treatments, suggesting that these combination partners may be suitable for further clinical development in sarcoma patients.

  • the Immunocytokine l19 tnf eradicates sarcomas in combination with chemotherapy or with immune check point inhibitors
    bioRxiv, 2019
    Co-Authors: Riccardo Corbellari, Dario Neri, Alessandra Villa, Roberto De Luca
    Abstract:

    Antibody-cytokine fusion proteins (also called "Immunocytokines") represent an emerging class of biopharmaceutical products, which are being considered for cancer immunotherapy. When used as single agents, pro-inflammatory Immunocytokines are rarely capable of induce complete and durable cancer regression in mouse models and in patients. However, the combination treatment with conventional chemotherapy or with other immune-stimulatory agents typically increases the therapeutic efficacy of Immunocytokines. In this article, we describe combination treatments of a tumor-targeting antibody-cytokine fusion protein based on the L19 antibody (specific to a splice isoform of fibronectin) fused to murine tumor necrosis factor (TNF) with standard chemotherapy (dacarbazine, trabectedin or melphalan) or with an immune check-point inhibitor (anti-PD-1) in an immunocompetent murine model of sarcoma (WEHI-164). All combination treatments led to improved tumor remission compared to monotherapy treatments, suggesting that these combination partners may be suitable for further clinical development in sarcoma patients.

  • targeted enhancement of the therapeutic window of l19 tnf by transient and selective inhibition of ripk1 signaling cascade
    bioRxiv, 2019
    Co-Authors: Sheila Dakhel, Tiziano Ongaro, Dario Neri, Alessandra Villa, Mattia Matasci, Baptiste Gouyou, Samuele Cazzamalli
    Abstract:

    Abstract Introduction Cytokine-based products are gaining importance for cancer immunotherapy. L19-TNF is a clinical-stage antibody-cytokine fusion protein that selectively accumulates to tumors and displays potent anticancer activity in preclinical models. Here, we describe an innovative approach to transiently inhibit off-target toxicity of L19-TNF, while maintaining antitumor activity. Methods GSK’963, a potent small molecule inhibitor of RIPK1, was tested in tumor-bearing mice for its ability to reduce acute toxicity associated with TNF signaling. The biological effects of L19-TNF on tumor cells, lymphocytes and tumor vessels were investigated with the aim to enable the administration of TNF doses, which would otherwise be lethal. Results Transient inhibition of RIPK1 allowed to increase the maximal tolerated dose of L19-TNF. The protective effect of GSK’963 did not affect the selective localization of the Immunocytokine to tumors as evidenced by quantitative biodistribution analysis and allowed to reach high local TNF concentrations around tumor blood vessels, causing diffused vascular shutdown and hemorrhagic necrosis within the neoplastic mass. Conclusions The selective inhibition of RIPK1 with small molecule inhibitors can be used as a pharmaceutical tool to transiently mask TNF activity and improve the therapeutic window of TNF-based biopharmaceuticals. Similar approaches may be applicable to other pro-inflammatory cytokines.

  • Abstract 5553: A novel Immunocytokine for the treatment of cancer
    Cancer Research, 2018
    Co-Authors: Mattia Matasci, Emanuele Puca, Tiziano Ongaro, Sarah Wulhfard, Alessandra Villa, Dario Neri
    Abstract:

    Interleukin-12 (IL12) is an immunomodulatory cytokine, which offers unique opportunities for cancer therapy due to its stimulatory function on cell-mediated immunity and its anti-angiogenic activity. IL12 was shown to polarize CD4+ T cells into a TH1 type and to be a key activator of NK cells and CD8+ T cells. It also induces the production of interferon-gamma by T cells and NK cells and subsequently of the anti-angiogenic chemokines CXCL10/IP-10, and CXCL9/Mig. However the potent anti-tumor IL12 activity, which has been reported in mice, has not yet been successfully translated into clinical development, mainly because of early reports of severe toxicity and low response rates in human. Following our pioneering work which started in 2002 with the first description of an IL12 based antibody-cytokine fusion protein (i.e. Immunocytokine), we have extensively explored and perfected alternative molecular formats, with the aim to further improve biodistribution properties and therapeutic activity of IL12-based Immunocytokines. Here we describe the development and evaluation of new targeted variants of both murine and human IL-12 with enhanced therapeutic efficacy and improved safety profile. To this end we combined the immunomodulatory properties of the IL12 payload with the tumor-homing activity of the L19 antibody. L19 is a clinical grade fully human antibody, which recognizes with identical affinity in both mouse and human, the alternatively spliced EDB domain of fibronectin. EDB represent an optimal target for anti-cancer pharmacodelivery, due to its pan-tumoral over-expression nature combined with very low expression levels in normal tissues. This has also been confirmed by extensive Nuclear Medicine work in which radiolabeled L19 has been administered to more than 150 patients, making L19 one of the best validated tumor-targeting agent. Our novel Immunocytokine, termed IL12-L19-L19, relies on the L19 antibody in tandem diabody format, with a monomeric IL12 moiety expressed as single-chain polypeptide at the N-terminal extremity. Recombinant IL12-L19-L19 proteins based either on human or murine IL12, were efficiently expressed in CHO cells and purified to high quality. The tumor-targeting properties of both variants were validated in tumor-bearing mice, using radioiodinated protein preparations. In preclinical therapy studies IL12-L19-L19 showed potent anti-cancer activity when used either as single agent or in combination with other anticancer agents. PK studies in Cynomolgus Monkey using the fully human IL12-L19-IL19 product, revealed a favorable profile, which is compatible with other clinical-stage Immunocytokines based on antibody-fragments. These results strongly support the further development of the fully human IL12-L19-L19 product for future clinical investigations. Citation Format: Mattia Matasci, Emanuele Puca, Tiziano Ongaro, Sarah Wulhfard, Alessandra Villa, Dario Neri. A novel Immunocytokine for the treatment of cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5553.

  • fri0118 dekavil f8il10 update on the results of clinical trials investigating the Immunocytokine in patients with rheumatoid arthritis
    Annals of the Rheumatic Diseases, 2018
    Co-Authors: Mauro Galeazzi, G D Sebastiani, Reinhard E Voll, Ombretta Viapiana, J Dudler, Enrico Selvi, Pascal Zufferey, Stephanie Finzel, F S Bootz, Dario Neri
    Abstract:

    Background The targeted pharmacodelivery of cytokines by means of antibody-fusion proteins aims at selective accumulation of the active compound at the site of disease while sparing healthy tissues. Dekavil (F8IL10) is a fully human fusion protein composed of the vascular targeting antibody F8 fused to the cytokine interleukin-10. A phase 1 dose escalation study in patients with rheumatoid arthritis (RA) has recently been completed. Dekavil is currently under investigation in a phase 2 clinical study for the treatment of RA. Objectives The phase 1 study investigated safety, tolerability and the maximum tolerated dose of Dekavil when administered in combination with methotrexate (MTX). The currently ongoing phase 2 (in patients with active RA despite MTX therapy who had failed anti-TNF treatment) measures the mean change from baseline in DAS28-CRP between F8IL10 and placebo arms as primary endpoint. Methods The target population of both RA studies comprises patients with active RA despite MTX therapy who had failed anti-TNF treatment. Dekavil is administered once weekly for 8 consecutive weeks by s.c. injection in combination with a fixed dose of MTX (10–25 mg). The recently completed phase 1 study investigated escalating doses of Dekavil (6–600 µg/kg +MTX). The ongoing randomised multicenter, double-blind, placebo-controlled phase 2 trial assesses therapeutic activity in two treatment groups (Dekavil 30 or 160 µg/kg plus MTX) and one placebo group (placebo plus MTX). Results Dekavil was well tolerated up to the highest investigated dose (600 µg/kg) and an MTD was not reached. In 34 out of 35 patients treated in the phase 1 study no SAEs and no SUSARs were reported. One subject (cohort 9, 450 µg/kg) experienced a DLT (G2 purpura), which was accompanied by an SAE (G2 dyspnea, not drug related). The patient fully recovered within one week following corticosteroid administration. The most frequently observed adverse event was mild (G1) injection site reaction and occurred in 60% of the patients. Furthermore, two cases of drug related anaemia (G3 and G2; 160 µg/kg and 450 µg/kg, respectively) were reported in this study. All adverse reactions resolved completely. After 4 cycles of treatment, 36.4% of patients (12/33) exhibited ACR responses. The fraction of responses increased to 45.8% (11/24) after 8 cycles. Two patients achievedan ACR70 for more than 12 months after the last drug administration. As of January 2018, 25 patients have been treated in the phase 2 clinical study and neither SUSARs nor treatment-related deaths were recorded. Immunogenicity assessment is completed for the first 20 study subjects did not reveal an antibody response specific to F8IL10. An interim read-out after 45 patients will provide a more thorough understanding of the therapeutic activity of F8IL10. Conclusions The data obtained in the population studied to date suggest that Dekavil may be a safe and well tolerated novel therapeutic for the potential treatment of RA. Disclosure of Interest M. Galeazzi: None declared, G. Sebastiani: None declared, R. Voll: None declared, O. Viapiana: None declared, J. Dudler: None declared, P. Zufferey: None declared, E. Selvi: None declared, S. Finzel: None declared, F. Bootz Employee of: Philogen Group (Sponsor of the study), D. Neri Shareholder of: Philogen Group (Sponsor of the study)

Stephen D Gillies - One of the best experts on this subject based on the ideXlab platform.

  • in situ vaccination with local radiation and intratumoral Immunocytokine to elicit a tumor specific memory b cell response
    Journal of Clinical Oncology, 2017
    Co-Authors: Claire C Baniel, Alexander L. Rakhmilevich, Jacquelyn A. Hank, Stephen D Gillies, Emily I Guy, Paul M Sondel, Alan J Korman, Hans Loibner, Paul M Harari, Zachary S Morris
    Abstract:

    69Background: In a murine melanoma (MEL) model, we reported an in situ vaccination response to combined radiation (RT) and intra-tumor (IT) injection of anti-GD2 hu14.18-IL2 Immunocytokine (IC). Th...

  • enhanced binding of necrosis targeting Immunocytokine nhs il12 after local tumour irradiation in murine xenograft models
    Cancer Immunology Immunotherapy, 2016
    Co-Authors: Franziska Eckert, Stephen D Gillies, Julia Schmitt, Daniel Zips, Marcel A Krueger, Bernd J Pichler, Wolfgang Strittmatter, Rupert Handgretinger, Karin Schilbach
    Abstract:

    NHS-IL12 is an Immunocytokine targeting necrotic tumour areas. IL12 shows anti-tumour activity. As local irradiation might induce additional necrosis in solid tumours, we aimed to evaluate the increase in intratumoural accumulation of NHS-IL12 after irradiation and correlate the findings with diffusion-weighted MRI studies in two xenograft models. Human rhabdomyosarcoma (A204) and prostate cancer (PC3) cells were studied in vitro and as subcutaneous xenografts. Radiation sensitivity of the cell lines was assessed in vitro by colony formation assays. In vivo tumour necrosis was assessed based on apparent diffusion coefficients (ADC). Biodistribution of NHS-IL12 was evaluated with and without tumour irradiation using in vivo small-animal PET and ex vivo biodistribution. A204 and PC3 differed in their intrinsic radiation sensitivity. Accordingly, radiation-induced tumour necrosis was found only in A204 xenografts. In comparison with control, ADC was significantly increased after irradiation of A204 tumours with 1 × 8.0 Gy and 5 × 2.0 Gy, whereas no change in ADC was observed in PC3 xenografts in all irradiation regimes. ADC correlated with histology. An enhanced uptake of radiolabelled NHS-IL12 in A204 tumours was detected by PET and ex vivo biodistribution after tumour irradiation. In PC3 tumours, no increase in NHS-IL12 uptake was observed. In dependence of the tumour model, tumour irradiation enhanced tumour necrosis measured in MRI and histology. In vivo PET and ex vivo biodistribution showed enhanced binding of NHS-IL12 in rhabdomyosarcoma xenografts. Thus, enhanced binding of necrosis-targeting Immunocytokines might be a novel mechanism of additive effects in combination with irradiation.

  • Immunocytokine augments local and abscopal response and animal survival when added to radiation and ctla 4 checkpoint inhibition in a murine melanoma model
    Journal for ImmunoTherapy of Cancer, 2015
    Co-Authors: Zachary S Morris, Jacquelyn A. Hank, Stephen D Gillies, Emily I Guy, David M Francis, Monica M Gressett, Eric A Armstrong, Shyhmin Huang, Alan J Korman, Alexander L. Rakhmilevich
    Abstract:

    Meeting abstracts We have identified a cooperative interaction between radiation and intratumoral injection of anti-GD2 Immunocytokine (hu14.18-IL2) in murine tumor models. In a moderate size (~200mm3), single tumor, B78 melanoma model this combination results in complete tumor regression in 71% of

  • in vivo synergy of radiation and hu14 18 il2 Immunocytokine results in a memory t cell response in a syngeneic murine melanoma model
    Journal for ImmunoTherapy of Cancer, 2014
    Co-Authors: Zachary S Morris, Alexander L. Rakhmilevich, Stephen D Gillies, Emily I Guy, David M Francis, Monica M Gressett, Richard K Yang, Paul M Harari, J A Hank, Paul M Sondel
    Abstract:

    Purpose Tumor-specific monoclonal antibodies (mAb) are a common type of immunotherapy capable of engaging innate immune cells to elicit antibody-dependent cell-mediated cytotoxicity (ADCC). We recently demonstrated in vivo synergy between radiation (RT) and ADCC using the anti-GD2 hu14.18 mAb. We now investigate the potential of hu14.18-IL2 Immunocytokine (IC) to augment this synergy.

  • immunologic and clinical responses to a cd20 targeted Immunocytokine di leu16 il2 in relapsed non hodgkin lymphoma
    Blood, 2013
    Co-Authors: Ryotaro Nakamura, David Colcher, Stephen D Gillies, Robert T Chen, Joycelynne Palmer, Karl Gaal, Auayporn Nademanee, Leslie Popplewell, James E Talmadge, Stephen J Forman
    Abstract:

    Background Tumor-targeted antibody-cytokine fusion proteins, known as Immunocytokines (ICKs) have potent antitumor activity in several preclinical tumor models. We have engineered an anti-CD20-interleukin 2 (IL-2) ICK based on the Leu16 anti-CD20 antibody with de-immunized variable (V) region and junction between the heavy (H) chain constant region and IL-2 (DI-Leu16-IL2). In-vitro studies of DI-Leu16-IL2 have demonstrated CD20 targeting, IL2 bioactivities, and antibody-dependent cell-mediated cytotoxicity (ADCC) functions. In a SCID mouse model, DI-Leu16-IL2 showed superior anti-lymphoma effects over rituximab or rituximab combined with systemic IL-2. Methods We have initiated a pilot trial of DI-Leu16-IL20 in CD20+ B-cell non-Hodgkin lymphoma (NHL) under an IRB-approved protocol (clinicaltrials.gov: [NCT00720135][1]). A total of 7 patients have been treated with DI-Leu16-IL20 at 0.5mg/m2. The first 2 cases received DI-Leu16-IL2 by intravenous infusion on days 2, 4 and 23, 25 after peripheral B cell depletion with low-dose rituximab (50mg/m2) on days 1 and 22. Five remaining patients received DI-Leu16-IL2 weekly for 4 weeks (days 2, 9, 16, 23) after B cell depletion with low-dose rituximab on days 1, 8, 15, and/or 22 as necessary, based on serum rituximab levels. Clinical Responses The treatment has been generally well tolerated with the majority of adverse events (AE) grade 1 or 2. Non-DLT grade 3 AE included one case of hyperglycemia, infection (herpes zoster), and prolonged QTc. One patient developed grade 3 hyponatremia, which was the only DLT in the study to date. One patient with diffuse large B cell NHL of the lower extremity achieved a CR after 2 cycles (significant PR after cycle 1; [Figure 1][2]). Another patient showed a temporal increase in FDG uptake on PET after cycle 1, which decreased after cycle 2 in 2 of the 3 responding lymph nodes, suggesting a local immunologic reaction with corresponding FDG uptake (immune-mediated FDG flare). A lymph node biopsy demonstrated a dense infiltration of T cells. One patient who had her residual tumor biopsied after DI-Leu16-IL2 treatment demonstrated loss of CD20 expression. Pre-ICK B Cell Depletion and Pharmacologic Data : We have successfully maintained minimal or absent peripheral blood B cells, with a level-guided dosing of low-dose rituximab. The Cmax ranged between 75 and 125 ng/ml following each infusion of DI-Leu16-IL2. The half life was 6-8 hours and did not change significantly between doses. We have detected no anti-DI-Leu16-IL2 antibodies in any of the 7 patients treated on this protocol. ![Figure][3] Immunologic Responses DI-Leu16-IL2 was highly immunologically active. There was a significant increase in local infiltrating T cells (predominantly CD8 T cells) in lymph node samples taken after DI-Leu16-IL2 in 3 of the 4 cases whose samples were available. Soluble IL2 receptor levels spiked with each DI-Leu16-IL2 infusion, up to 3 times above baseline. Among the cytokines and chemokines we evaluated using the Luminex platform, IL-5, IL-7, IL-10, IP10, IL-15, and IFN-gamma demonstrated high spikes (>2-fold increase) within 12 hours from each DI-Leu16-IL2 infusion, while IL-1, IL-12, MIG, TNF-alpha, IFN-alpha, and GCSF levels had a lesser degree of spikes (<2-fold increase). The data on the CR patient seem to show higher peak levels of IL15, IL7, and IP10, while MCP1, IL5, IL10, and MCP appeared lower compared with others. Flow cytometry-based cellular analyses showed no significant increase in regulatory T cells or myeloid-derived suppressor cells. Peripheral blood T cells were analyzed for TCR CDR3 sequence, and demonstrated oligoclonal T cell expansions, suggestive of antigen-driven T cell expansion. Conclusion This study demonstrated the clinical and immunologic data consistent with a proof-of-principle that DI-Leu16-IL2 is capable of inducing both local and systemic immune responses at a dose 0.5mg/m2, and was associated with a CR in a patient who had highly refractory NHL. A multi-center phase I trial of subcutaneously administered DI-Leu16-IL2 is currently underway. Disclosures: Gillies: Provenance Biopharmaceuticals Corp (employment and equity ownership), Alopexx Oncology (Consultancy) : Consultancy, Employment, Equity Ownership. [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT00720135&atom=%2Fbloodjournal%2F122%2F21%2F1808.atom [2]: #F1 [3]: pending:yes

Christian Klein - One of the best experts on this subject based on the ideXlab platform.

  • abstract 1552 a novel pd1 il2v Immunocytokine for preferential cis activation of il 2r signaling on pd 1 expressing t cell subsets strongly potentiates anti tumor t cell activity of pd 1 checkpoint inhibition and il 2r beta gamma agonism
    Immunology, 2019
    Co-Authors: Christian Klein, Johannes Sam, Valeria G Nicolini, Laura Codarrideak, Stefan Seeber, Laura Lauener, Marine Richard, Esther Bommer, Maria Karagianni, Ramona Schlenker
    Abstract:

    High-dose IL-2 is approved for patients with metastatic melanoma and renal cell cancer, but is associated with significant toxicity. We have described tumor-targeted CEA-IL2v and FAP-IL2v Immunocytokines that are based on an engineered IL2v moiety with abolished binding to IL-2Ra (CD25) to avoid undesired CD25-mediated toxicities and Treg expansion. Their antibodies bind with high affinity to either CEA or FAP, which are broadly expressed in various tumors, and mediate retention/accumulation in malignant lesions. In an effort to further maximize the potency of IL-2R activation of T cells (aka signal 3) without the side effects of wildtype IL-2, we have generated PD1-IL2v that binds to the checkpoint inhibitor PD-1 and delivers IL2v preferentially in cis to PD-1+ T cells substituting binding to CD25. This enables high affinity IL2R signaling selectively in recently primed antigen specific TILs and stem-cell like subsets. Binding/competition experiments demonstrated that PD1-IL2v is 50-fold more potent than FAP-IL2v in inducing p-STAT5 in PD-1 positive T cells, whereas it has equivalent potency on PD-1 negative T cells. Notably, although Tregs can express low constitutive levels of PD-1, PD1-IL2v shows preferential binding to Teff vs. Tregs. Furthermore, PD1-IL2v can overcome Treg suppression of Tconv effector cells to a greater extent than PD-1, FAP-IL2v or their combination. For use in syngeneic mouse models muPD1-IL2v was generated. In an orthotopic pancreatic Panc02-Luciferase model administration of muPD1-IL2v (1 mg/kg IV, qw) resulted in rapid and complete elimination of tumor cells (loss of luciferase signal) and in long term survival (> 140 days) in 7/7 treated animals with protection from tumor cell re-challenge, whereas only 1/7 animals in the muPD-1 + muFAP-IL2v combination group (10 mg/kg + 2.5 mg/kg IV, qw) and none of the animals treated with the respective monotherapies showed long term survival. IHC showed that the improved outcome with muPD1-IL2v was related to a strong intra-tumoral increase of CD3+ CD8+ T cells expressing PD-1 and GrzB. In an immuno-PD study with s.c. Panc02 tumors muPD1-IL2v strongly increased the number of IFNg+ TNFa+ multifunctional and cytotoxic GrzB+ PD-1+ T cells in the tumor accompanied with a 20-fold increase in CD8/CD4 ratio and a strong increase in the CD8+ TEM population resulting in subsequent tumor control or remission in 50% of the animals, respectively. In summary, our data demonstrate that preferential cis-targeting of IL2v to PD-1+ antigen specific T cells with PD1-IL2v results in a strong potentiation of T cell response and anti-tumor efficacy as compared to the combination of PD-1 checkpoint inhibition with FAP-IL2v. These preclinical data establish PD1-IL2v as a promising next generation IL-2 for cancer immunotherapy. Citation Format: Christian Klein, Laura Codarri-Deak, Valeria Nicolini, Stefan Seeber, Laura Lauener, Marine Richard, Esther Bommer, Maria Karagianni, Johannes Sam, Ramona Schlenker, Marisa Mariani, Petra Petra Schwalie, Sylvia Herter, Marina Bacac, Inja Waldhauer, Anne Freimoser-Grundschober, Volker Teichgraeber, Pablo Umana. A novel PD1-IL2v Immunocytokine for preferential cis-activation of IL-2R signaling on PD-1 expressing T cell subsets strongly potentiates anti-tumor T cell activity of PD-1 checkpoint inhibition and IL-2R-beta-gamma agonism [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1552.

  • prediction of the optimal dosing regimen using a mathematical model of tumor uptake for Immunocytokine based cancer immunotherapy
    Clinical Cancer Research, 2018
    Co-Authors: Benjamin Ribba, Stefan Evers, Christian Klein, Tapan K Nayak, Christophe Boetsch, Hans Peter Grimm, Jehad Charo, Jean Tessier, Jean Eric Charoin, Alex Phipps
    Abstract:

    Purpose: Optimal dosing is critical for Immunocytokine-based cancer immunotherapy to maximize efficacy and minimize toxicity. Cergutuzumab amunaleukin (CEA-IL2v) is a novel CEA-targeted Immunocytokine. We set out to develop a mathematical model to predict intratumoral CEA-IL2v concentrations following various systemic dosing intensities.Experimental Design: Sequential measurements of CEA-IL2v plasma concentrations in 74 patients with solid tumors were applied in a series of differential equations to devise a model that also incorporates the peripheral concentrations of IL2 receptor-positive cell populations (i.e., CD8+, CD4+, NK, and B cells), which affect tumor bioavailability of CEA-IL2v. Imaging data from a subset of 14 patients were subsequently utilized to additionally predict antibody uptake in tumor tissues.Results: We created a pharmacokinetic/pharmacodynamic mathematical model that incorporates the expansion of IL2R-positive target cells at multiple dose levels and different schedules of CEA-IL2v. Model-based prediction of drug levels correlated with the concentration of IL2R-positive cells in the peripheral blood of patients. The pharmacokinetic model was further refined and extended by adding a model of antibody uptake, which is based on drug dose and the biological properties of the tumor. In silico predictions of our model correlated with imaging data and demonstrated that a dose-dense schedule comprising escalating doses and shortened intervals of drug administration can improve intratumoral drug uptake and overcome consumption of CEA-IL2v by the expanding population of IL2R-positive cells.Conclusions: The model presented here allows simulation of individualized treatment plans for optimal dosing and scheduling of Immunocytokines for anticancer immunotherapy. Clin Cancer Res; 24(14); 3325-33. ©2018 AACRSee related commentary by Ruiz-Cerda et al., p. 3236.

  • abstract 2774 the triple combination of the fap il2v Immunocytokine with pd l1 checkpoint inhibitory and cd40 agonistic antibodies results in long term tumor control in the orthotopic panco2 model
    Cancer Research, 2018
    Co-Authors: Valeria G Nicolini, Inja Waldhauer, Anne Freimoser, Emily Corse, Jahad Charo, Pablo Umana, Christian Klein
    Abstract:

    Introduction: We have previously described the FAP-targeted Immunocytokine FAP-IL2v and its combination with PD-L1 checkpoint inhibition. Here we show that the anti-tumoral efficacy of FAP-IL2v can be strongly enhanced by combination with a CD40 agonistic antibody as well as the triple combination of FAP-IL2v with a CD40 agonistic and a PD-L1 checkpoint inhibitory antibody in the syngeneic PancO2 model. Methods: Anti-tumoral efficacy was assessed in the orthotopic intra-pancreatic PancO2 model transfected with Luciferase in C57BL/6 mice. Mice were injected intra-pancreatically on study day 0 with 1x10E5 Panc02-Fluc cells, randomized and weighed. One week after tumor cell injection mice were injected i.p. with the murine specific surrogate Immunocytokine muFAP-muIL2v (muIgG1 DAPG) (2 mg/kg, q1wx3), anti-muPD-L1 surrogate antibody (muIgG1 DAPG) (10 mg/kg, q1wx3) and/or the anti-muCD40 surrogate antibody FGK4.5 (muIgG1) (10mg/kg, q1wx1). Bioluminescence imaging (BLI) was performed after ip injection of 150 mg/kg D-Luciferin 10 min before acquisition using IVIS® SPECTRUM. An analogous experiment was performed in C57BL/6 B cell knockout mice. Results: In the orthotopic PancO2-Fluc model the triple combination of FAP-IL2v plus anti-PD-L1 plus anti-CD40 showed superior outcome compared to vehicle and the respective monotherapies in terms of median survival and overall survival in the majority of animals followed by the combination of FAP-IL2v with anti-CD40 and the combinations of anti-PD-L1 with anti-CD40 and FAP-IL2v with anti-PD-L1, respectively. Bioluminescence imaging confirmed the survival outcome of the respective groups by decrease in/absence of bioluminescence signals in the tumor area. Tumor re-challenge experiments showed that the animals with long-term survival were protected both in the C57BL/6 and the C57BL/6 B cell knockout mice. Conclusions: These data show that the anti-tumoral efficacy of FAP-IL2v in the syngeneic PancO2 model can be strongly enhanced by combining it with a CD40 agonistic antibody on top of a PD-L1 checkpoint inhibitory antibody. Most notably, the triple combination of FAP-IL2v with CD40 agonism and PD-L1 checkpoint inhibition resulted in long term survival of the majority of animals. These data provide the preclinical rationale for further clinical investigation of the combination of FAP-IL2v (RG7461) with the PD-L1 antibody atezolizumab and the CD40 agonistic antibody selicrelumab (RG7876). Citation Format: Valeria Nicolini, Inja Waldhauer, Anne Freimoser, Emily Corse, Jahad Charo, Pablo Umana, Christian Klein. The triple combination of the FAP-IL2v Immunocytokine with PD-L1 checkpoint inhibitory and CD40 agonistic antibodies results in long-term tumor control in the orthotopic PancO2 model [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2774.

  • Cergutuzumab amunaleukin (CEA-IL2v), a CEA-targeted IL-2 variant-based Immunocytokine for combination cancer immunotherapy: Overcoming limitations of aldesleukin and conventional IL-2-based Immunocytokines.
    Oncoimmunology, 2017
    Co-Authors: Christian Klein, Tapan K Nayak, Inja Waldhauer, Nicolini Valeria G, Anne Freimoser-grundschober, Danielle J. Vugts, Claire Dunn, Marije Bolijn, Jörg Benz, Martine Stihle
    Abstract:

    We developed cergutuzumab amunaleukin (CEA-IL2v, RG7813), a novel monomeric CEA-targeted Immunocytokine, that comprises a single IL-2 variant (IL2v) moiety with abolished CD25 binding, fused to the C-terminus of a high affinity, bivalent carcinoembryonic antigen (CEA)-specific antibody devoid of Fc-mediated effector functions. Its molecular design aims to (i) avoid preferential activation of regulatory T-cells vs. immune effector cells by removing CD25 binding; (ii) increase the therapeutic index of IL-2 therapy by (a) preferential retention at the tumor by having a lower dissociation rate from CEA-expressing cancer cells vs. IL-2R-expressing cells, (b) avoiding any FcγR-binding and Fc effector functions and (c) reduced binding to endothelial cells expressing CD25; and (iii) improve the pharmacokinetics, and thus convenience of administration, of IL-2. The crystal structure of the IL2v-IL-2Rβγ complex was determined and CEA-IL2v activity was assessed using human immune effector cells. Tumor targeting was investigated in tumor-bearing mice using 89Zr-labeled CEA-IL2v. Efficacy studies were performed in (a) syngeneic mouse models as monotherapy and combined with anti-PD-L1, and in (b) xenograft mouse models in combination with ADCC-mediating antibodies. CEA-IL2v binds to CEA with pM avidity but not to CD25, and consequently did not preferentially activate Tregs. In vivo, CEA-IL2v demonstrated superior pharmacokinetics and tumor targeting compared with a wild-type IL-2-based CEA Immunocytokine (CEA-IL2wt). CEA-IL2v strongly expanded NK and CD8+ T cells, skewing the CD8+:CD4+ ratio toward CD8+ T cells both in the periphery and in the tumor, and mediated single agent efficacy in syngeneic MC38-CEA and PancO2-CEA models. Combination with trastuzumab, cetuximab and imgatuzumab, all of human IgG1 isotype, resulted in superior efficacy compared with the monotherapies alone. Combined with anti-PD-L1, CEA-IL2v mediated superior efficacy over the respective monotherapies, and over the combination with an untargeted control Immunocytokine. These preclinical data support the ongoing clinical investigation of the cergutuzumab amunaleukin Immunocytokine with abolished CD25 binding for the treatment of CEA-positive solid tumors in combination with PD-L1 checkpoint blockade and ADCC competent antibodies.

  • abstract 2592 tumor targeting and pharmacodynamics of the novel targeted Immunocytokine fap il2v in a tumor bearing rhesus monkey
    Cancer Research, 2014
    Co-Authors: Stefan Evers, Pradeep Garg, Mark J Cline, Sudha Garg, Greg Dugan, Anne Freimosergrundschober, Natalie D Keirstead, Christian Klein, Tapan K Nayak
    Abstract:

    FAP-IL2v is a novel monomeric FAP-targeted Immunocytokine where a single, engineered IL-2 variant (IL-2v) with abolished IL-2Ralpha (CD25) binding is fused to the C-terminus of an high affinity anti-FAP antibody (1). FAP-IL2v was designed to selectively bind to the tumor thereby avoiding the systemic toxicity inherent to systemic administration of cytokines. The goal of this study was to assess the tumor targeting and pharmacodynamic properties of FAP-IL2v in an appropriate tumor bearing animal model. Explanted tumor xenografts in mice do not have high expression of FAP and lack the complete immune system which is important to assess the safety, efficacy, and biodistribution of these targeted Immunocytokines. Therefore, we elected to use monkeys with spontaneous tumors for this study. Herein, we report a first of kind study in monkeys with spontaneous tumors to better understand tumor targeting and pharmacodynamics of FAP-IL2v. Three monkeys with a history of cancer and suspected relapse were identified. Immunohistochemistry was done on archival tumor tissue to assess target (FAP) expression and FDG-PET performed to detect metastatic lesions. Based on the positive FAP expression on tumor tissue and detectable tumor lesions and lesion locations from FDG-PET scans, a rhesus monkey with metastatic breast cancer was selected for further experimentation with FAP-IL2v. The monkey was administered with 0.5 mg/kg of FAP-IL2v mixed with tracer amounts of 89Zr labeled FAP-IL2v for assessment of tumor targeting using PET imaging. Baseline and on-treatment blood samples and tumor biopsies were collected after single-administration for assessment of pharmacodynamic effects. The study revealed that FAP-IL2v selectively targets and accumulates in the tumor lesions with minimal uptake in the lungs, spleen and lymphoid tissues (major sink organs and potential risk factors). All three FDG-avid lesions showed accumulation of FAP-IL2v over 6 days with peak uptake of SUV= 3.1 at day 3. After single administration, transient lymphocytosis and peripheral expansion of immune cells were observed. Analyses of tumor biopsies suggested increased immune cell infiltration. No signs of distress or cytokine release were observed during the treatment period. In conclusion, using a monkey with spontaneous tumors, we demonstrated that the FAP-IL2v targeted Immunocytokine can selectively be delivered to tumor and induce intended changes in immune microenvironment without major adverse events. (1) C. Klein et al., Tumor-targeted, engineered IL-2 variant (IL-2v)-based Immunocytokines for the immunotherapy of cancer, AACR 2013, Poster 486 Citation Format: Stefan Evers, Pradeep K. Garg, J. Mark Cline, Sudha Garg, Greg Dugan, Anne Freimoser-Grundschober, Natalie D. Keirstead, Christian Klein, Tapan Nayak. Tumor targeting and pharmacodynamics of the novel targeted Immunocytokine FAP-IL2v in a tumor-bearing Rhesus monkey. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2592. doi:10.1158/1538-7445.AM2014-2592

Nicolle H Rekers - One of the best experts on this subject based on the ideXlab platform.

  • the Immunocytokine l19 il2 an interplay between radiotherapy and long lasting systemic anti tumour immune responses
    OncoImmunology, 2017
    Co-Authors: Nicolle H Rekers, Dario Neri, Veronica Olivo Pimentel, Ala Yaromina, Natasja G Lieuwes, Rianne Biemans, Catharina M L Zegers, Wilfred T V Germeraad, Evert J Van Limbergen, Ludwig Dubois
    Abstract:

    ABSTRACTRecently, we have shown that the administration of the tumour-targeted antibody-based Immunocytokine L19-IL2 after radiotherapy (RT) resulted in synergistic anti-tumour effect. Here we show that RT and L19-IL2 can activate a curative abscopal effect, with a long-lasting immunological memory. Ionizing radiation (single dose of 15Gy, 5 × 2Gy or 5 × 5Gy) was delivered to primary C51 colon tumour-bearing immunocompetent mice in combination with L19-IL2 and response of secondary non-irradiated C51 or CT26 colon tumours was evaluated. 15Gy + L19-IL2 triggered a curative (20%) abscopal effect, which was T cell dependent. Moreover, 10Gy + L19-IL2 treated and cured mice were re-injected after 150 days with C51 tumour cells and tumour uptake was assessed. Age-matched controls (matrigel injected mice treated with 10Gy + L19-IL2, mice cured after treatment with surgery + L19-IL2 and mice cured after high dose RT 40Gy + vehicle) were included. Several immunological parameters in blood, tumours, lymph nodes and...

  • combination of radiotherapy with the Immunocytokine l19 il2 additive effect in a nk cell dependent tumour model
    Radiotherapy and Oncology, 2015
    Co-Authors: Nicolle H Rekers, Ala Yaromina, Natasja G Lieuwes, Rianne Biemans, Catharina M L Zegers, Wilfred T V Germeraad, Evert J Van Limbergen, Birgit L M G Sendengijsbers, Mario Losen, Dario Neri
    Abstract:

    Abstract Background and purpose Recently, we have shown that radiotherapy (RT) combined with the Immunocytokine L19-IL2 can induce long-lasting antitumour effects, dependent on ED-B expression and infiltration of cytotoxic T cells. On the other hand, in certain tumours, IL2 treatment can trigger a natural killer cell (NK) immune response. The aim of this study is to investigate the therapeutic effect of our combination therapy in the ED-B positive F9 teratocarcinoma model, lacking MHCI expression and known to be dependent on NK immune responses. Material and methods In syngeneic F9 tumour bearing 129/FvHsd mice tumour growth delay was evaluated after local tumour irradiation (10 Gy) combined with systemic administration of L19-IL2. Immunological responses were investigated using flow cytometry. Results Tumour growth delay of L19-IL2 can be further improved by a single dose of RT administered before immunotherapy, but not during immunotherapy. Furthermore, treatment of L19-IL2 favours a NK response and lacks cytotoxic T cell tumour infiltrating immune cells, which may be explained by the absence of MHCI expression. Conclusion An additive effect can be detected when the NK dependent F9 tumour model is treated with radiotherapy and L19-IL2 and therefore this combination could be useful in the absence of tumoural MHCI expression.

  • long lasting antitumor effects provided by radiotherapy combined with the Immunocytokine l19 il2
    OncoImmunology, 2015
    Co-Authors: Nicolle H Rekers, Catharina M L Zegers, Wilfred T V Germeraad, Ludwig Dubois, Philippe Lambin
    Abstract:

    Recently, we have shown that radiotherapy (RT) combined with L19-IL2 can induce a long-lasting antitumor effect, dependent on ED-B expression and infiltration of cytotoxic T cells. These findings will be translated to a Phase I clinical study (NCT02086721) in patients with oligometastatic solid tumors. See this link for the animation: http://youtu.be/xHbwQuCTkRc.

  • radiotherapy combined with the Immunocytokine l19 il2 provides long lasting antitumor effects
    Clinical Cancer Research, 2015
    Co-Authors: Catharina M L Zegers, Nicolle H Rekers, Ala Yaromina, Natasja G Lieuwes, Wilfred T V Germeraad, Dana H F Quaden, Lotte Wieten, Erik A L Biessen, Louis Boon, Dario Neri
    Abstract:

    Purpose: Radiotherapy modifies the tumor microenvironment and causes the release of tumor antigens, which can enhance the effect of immunotherapy. L19 targets the extra domain B (ED-B) of fibronectin, a marker for tumor neoangiogenesis, and can be used as Immunocytokine when coupled to IL2. We hypothesize that radiotherapy in combination with L19-IL2 provides an enhanced antitumor effect, which is dependent on ED-B expression. Experimental Design: Mice were injected with syngeneic C51 colon carcinoma, Lewis lung carcinoma (LLC), or 4T1 mammary carcinoma cells. Tumor growth delay, underlying immunologic parameters, and treatment toxicity were evaluated after single-dose local tumor irradiation and systemic administration of L19-IL2 or equimolar controls. Results: ED-B expression was high, intermediate, and low for C51, LLC, and 4T1, respectively. The combination therapy showed (i) a long-lasting synergistic effect for the C51 model with 75% of tumors being cured, (ii) an additive effect for the LLC model, and (iii) no effect for the 4T1 model. The combination treatment resulted in a significantly increased cytotoxic (CD8 + ) T-cell population for both C51 and LLC. Depletion of CD8 + T cells abolished the benefit of the combination therapy. Conclusions: These data provide the first evidence for an increased therapeutic potential by combining radiotherapy with L19-IL2 in ED-B–positive tumors. This new opportunity in cancer treatment will be investigated in a phase I clinical study for patients with an oligometastatic solid tumor (NCT02086721). An animation summarizing our results is available at https://www.youtube.com/watch?v=xHbwQuCTkRc. Clin Cancer Res; 21(5); 1151–60. ©2014 AACR .

Jacquelyn A. Hank - One of the best experts on this subject based on the ideXlab platform.

  • pilot trial of the hu14 18 il2 Immunocytokine in patients with completely resectable recurrent stage iii or stage iv melanoma
    Cancer Immunology Immunotherapy, 2018
    Co-Authors: Mark R Albertini, Jacquelyn A. Hank, Richard K Yang, Erik A Ranheim, Cindy L Zuleger, Sharon M Weber, Heather B Neuman, Greg Hartig, Tracey L Weigel, David M Mahvi
    Abstract:

    Phase I testing of the hu14.18-IL2 Immunocytokine (IC) in melanoma patients showed immune activation, reversible toxicities, and a maximal tolerated dose of 7.5 mg/m2/day. Preclinical data in IC-treated tumor-bearing mice with low tumor burden documented striking antitumor effects. Patients with completely resectable recurrent stage III or stage IV melanoma were scheduled to receive 3 courses of IC at 6 mg/m2/day i.v. on days 1, 2 and 3 of each 28-day course. Patients were randomized to complete surgical resection either following neoadjuvant (Group A) or prior to adjuvant (Group B) IC course 1. Primary objectives were to: (1) evaluate histological evidence of anti-tumor activity and (2) evaluate recurrence-free survival (RFS) and OS. Twenty melanoma patients were randomized to Group A (11 patients) or B (9 patients). Two Group B patients did not receive IC due to persistent disease following surgery. Six of 18 IC-treated patients remained free of recurrence, with a median RFS of 5.7 months (95% confidence interval (CI) 1.8-not reached). The 24-month RFS rate was 38.9% (95% CI 17.5–60.0%). The median follow-up of surviving patients was 50.0 months (range: 31.8–70.4). The 24-month OS rate was 65.0% (95% CI 40.3–81.5%). Toxicities were similar to those previously reported. Exploratory tumor-infiltrating lymphocyte (TIL) analyses suggest prognostic value of TILs from Group A patients. Prolonged tumor-free survival was seen in some melanoma patients at high risk for recurrence who were treated with IC.

  • in situ vaccination with local radiation and intratumoral Immunocytokine to elicit a tumor specific memory b cell response
    Journal of Clinical Oncology, 2017
    Co-Authors: Claire C Baniel, Alexander L. Rakhmilevich, Jacquelyn A. Hank, Stephen D Gillies, Emily I Guy, Paul M Sondel, Alan J Korman, Hans Loibner, Paul M Harari, Zachary S Morris
    Abstract:

    69Background: In a murine melanoma (MEL) model, we reported an in situ vaccination response to combined radiation (RT) and intra-tumor (IT) injection of anti-GD2 hu14.18-IL2 Immunocytokine (IC). Th...

  • Immunocytokine augments local and abscopal response and animal survival when added to radiation and ctla 4 checkpoint inhibition in a murine melanoma model
    Journal for ImmunoTherapy of Cancer, 2015
    Co-Authors: Zachary S Morris, Jacquelyn A. Hank, Stephen D Gillies, Emily I Guy, David M Francis, Monica M Gressett, Eric A Armstrong, Shyhmin Huang, Alan J Korman, Alexander L. Rakhmilevich
    Abstract:

    Meeting abstracts We have identified a cooperative interaction between radiation and intratumoral injection of anti-GD2 Immunocytokine (hu14.18-IL2) in murine tumor models. In a moderate size (~200mm3), single tumor, B78 melanoma model this combination results in complete tumor regression in 71% of

  • differential internalization of hu14 18 il2 Immunocytokine by nk and tumor cell impact on conjugation cytotoxicity and targeting
    Journal of Leukocyte Biology, 2011
    Co-Authors: Ilia N. Buhtoiarov, Jacquelyn A. Hank, Jacek Gan, Zane C Neal, Brett H Yamane, Tatiana N Buhtoiarova, Manish S Patankar, Jennifer A A Gubbels, Alexander L. Rakhmilevich
    Abstract:

    The hu14.18-IL2 (EMD 273063) IC, consisting of a GD(2)-specific mAb genetically engineered to two molecules of IL-2, is in clinical trials for treatment of GD(2)-expressing tumors. Anti-tumor activity of IC in vivo and in vitro involves NK cells. We studied the kinetics of retention of IC on the surface of human CD25(+)CD16(-) NK cell lines (NKL and RL12) and GD(2)(+) M21 melanoma after IC binding to the cells via IL-2R and GD(2), respectively. For NK cells, ∼ 50% of IC was internalized by 3 h and ∼ 90% by 24 h of cell culture. The decrease of surface IC levels on NK cells correlated with the loss of their ability to bind to tumor cells and mediate antibody-dependent cellular cytotoxicity in vitro. Unlike NK cells, M21 cells retained ∼ 70% of IC on the surface following 24 h of culture and maintained the ability to become conjugated and lysed by NK cells. When NKL cells were injected into M21-bearing SCID mice, IT delivery of IC augmented NK cell migration into the tumor. These studies demonstrate that once IC binds to the tumor, it is present on the tumor surface for a prolonged time, inducing the recruitment of NK cells to the tumor site, followed by tumor cell killing.

  • genotypes of nk cell kir receptors their ligands and fcγ receptors in the response of neuroblastoma patients to hu14 18 il2 immunotherapy
    Cancer Research, 2010
    Co-Authors: David C Delgado, Jacquelyn A. Hank, Stephen D Gillies, Jacek Gan, Songwon Seo, Kyungmann Kim, Jill M Kolesar, David Lorentzen, Suzanne Shusterman, Ralph A. Reisfeld
    Abstract:

    Response to Immunocytokine (IC) therapy is dependent on natural killer cells in murine neuroblastoma (NBL) models. Furthermore, killer immunoglobulin-like receptor (KIR)/KIR-ligand mismatch is associated with improved outcome to autologous stem cell transplant for NBL. Additionally, clinical antitumor response to monoclonal antibodies has been associated with specific polymorphic-FcγR alleles. Relapsed/refractory NBL patients received the hu14.18-IL2 IC (humanized anti-GD2 monoclonal antibody linked to human IL2) in a Children's Oncology Group phase II trial. In this report, these patients were genotyped for KIR, HLA, and FcR alleles to determine whether KIR receptor-ligand mismatch or specific FcγR alleles were associated with antitumor response. DNA samples were available for 38 of 39 patients enrolled: 24 were found to have autologous KIR/KIR-ligand mismatch; 14 were matched. Of the 24 mismatched patients, 7 experienced either complete response or improvement of their disease after IC therapy. There was no response or comparable improvement of disease in patients who were matched. Thus KIR/KIR-ligand mismatch was associated with response/improvement to IC (P = 0.03). There was a trend toward patients with the FcγR2A 131-H/H genotype showing a higher response rate than other FcγR2A genotypes (P = 0.06). These analyses indicate that response or improvement of relapsed/refractory NBL patients after IC treatment is associated with autologous KIR/KIR-ligand mismatch, consistent with a role for natural killer cells in this clinical response.