Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
MAbs ; 16(1): 2395499, 2024.
Article de Anglais | MEDLINE | ID: mdl-39205483

RÉSUMÉ

T cell engagers (TCEs) are becoming an integral class of biological therapeutic owing to their highly potent ability to eradicate cancer cells. Nevertheless, the widespread utility of classical CD3-targeted TCEs has been limited by narrow therapeutic index (TI) linked to systemic CD4+ T cell activation and aberrant cytokine release. One attractive approach to circumvent the systemic activation of pan CD3+ T cells and reduce the risk of cytokine release syndrome is to redirect specific subsets of T cells. A promising strategy is the use of peptide-major histocompatibility class I bispecific antibodies (pMHC-IgGs), which have emerged as an intriguing modality of TCE, based on their ability to selectively redirect highly reactive viral-specific effector memory cytotoxic CD8+ T cells to eliminate cancer cells. However, the relatively low frequency of these effector memory cells in human peripheral blood mononuclear cells (PBMCs) may hamper their redirection as effector cells for clinical applications. To mitigate this potential limitation, we report here the generation of a pMHC-IgG derivative known as guided-pMHC-staging (GPS) carrying a covalent fusion of a monovalent interleukin-2 (IL-2) mutein (H16A, F42A). Using an anti-epidermal growth factor receptor (EGFR) arm as a proof-of-concept, tumor-associated antigen paired with a single-chain HLA-A *02:01/CMVpp65 pMHC fusion moiety, we demonstrate in vitro that the IL-2-armored GPS modality robustly expands CMVpp65-specific CD8+ effector memory T cells and induces potent cytotoxic activity against target cancer cells. Similar to GPS, IL-2-armored GPS molecules induce modulated T cell activation and reduced cytokine release profile compared to an analogous CD3-targeted TCE. In vivo we show that IL-2-armored GPS, but not the corresponding GPS, effectively expands grafted CMVpp65 CD8+ T cells from unstimulated human PBMCs in an NSG mouse model. Lastly, we demonstrate that the IL-2-armored GPS modality exhibits a favorable developability profile and monoclonal antibody-like pharmacokinetic properties in human neonatal Fc receptor transgenic mice. Overall, IL-2-armored GPS represents an attractive approach for treating cancer with the potential for inducing vaccine-like antiviral T cell expansion, immune cell redirection as a TCE, and significantly widened TI due to reduced cytokine release.


Sujet(s)
Anticorps bispécifiques , Lymphocytes T CD8+ , Interleukine-2 , Anticorps bispécifiques/immunologie , Anticorps bispécifiques/pharmacologie , Humains , Animaux , Souris , Lymphocytes T CD8+/immunologie , Interleukine-2/immunologie , Cytokines/immunologie , Cytokines/métabolisme , Protéines de la matrice virale/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Lignée cellulaire tumorale , Mémoire immunologique , Antigènes d'histocompatibilité de classe I/immunologie
2.
Nat Commun ; 15(1): 1700, 2024 Feb 24.
Article de Anglais | MEDLINE | ID: mdl-38402224

RÉSUMÉ

The Ataxia telangiectasia and Rad3-related (ATR) inhibitor ceralasertib in combination with the PD-L1 antibody durvalumab demonstrated encouraging clinical benefit in melanoma and lung cancer patients who progressed on immunotherapy. Here we show that modelling of intermittent ceralasertib treatment in mouse tumor models reveals CD8+ T-cell dependent antitumor activity, which is separate from the effects on tumor cells. Ceralasertib suppresses proliferating CD8+ T-cells on treatment which is rapidly reversed off-treatment. Ceralasertib causes up-regulation of type I interferon (IFNI) pathway in cancer patients and in tumor-bearing mice. IFNI is experimentally found to be a major mediator of antitumor activity of ceralasertib in combination with PD-L1 antibody. Improvement of T-cell function after ceralasertib treatment is linked to changes in myeloid cells in the tumor microenvironment. IFNI also promotes anti-proliferative effects of ceralasertib on tumor cells. Here, we report that broad immunomodulatory changes following intermittent ATR inhibition underpins the clinical therapeutic benefit and indicates its wider impact on antitumor immunity.


Sujet(s)
Lymphocytes T CD8+ , Indoles , Morpholines , Tumeurs , Pyrimidines , Sulfonamides , Humains , Animaux , Souris , Antigène CD274 , Microenvironnement tumoral , Lignée cellulaire tumorale , Immunothérapie , Modèles animaux de maladie humaine , Protéines mutées dans l'ataxie-télangiectasie
3.
Mol Cancer Ther ; 20(9): 1723-1734, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34224361

RÉSUMÉ

A recombinant Newcastle Disease Virus (NDV), encoding either a human (NDVhuGM-CSF, MEDI5395) or murine (NDVmuGM-CSF) GM-CSF transgene, combined broad oncolytic activity with the ability to significantly modulate genes related to immune functionality in human tumor cells. Replication in murine tumor lines was significantly diminished relative to human tumor cells. Nonetheless, intratumoral injection of NDVmuGM-CSF conferred antitumor effects in three syngeneic models in vivo; with efficacy further augmented by concomitant treatment with anti-PD-1/PD-L1 or T-cell agonists. Ex vivo immune profiling, including T-cell receptor sequencing, revealed profound immune-contexture changes consistent with priming and potentiation of adaptive immunity and tumor microenvironment (TME) reprogramming toward an immune-permissive state. CRISPR modifications rendered CT26 tumors significantly more permissive to NDV replication, and in this setting, NDVmuGM-CSF confers immune-mediated effects in the noninjected tumor in vivo Taken together, the data support the thesis that MEDI5395 primes and augments cell-mediated antitumor immunity and has significant utility as a combination partner with other immunomodulatory cancer treatments.


Sujet(s)
Facteur de stimulation des colonies de granulocytes et de macrophages/administration et posologie , Immunomodulation , Immunothérapie/méthodes , Virus de la maladie de Newcastle/génétique , Thérapie virale de cancers/instrumentation , Microenvironnement tumoral , Animaux , Apoptose , Prolifération cellulaire , Tumeurs du côlon/immunologie , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/thérapie , Femelle , Humains , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
4.
J Clin Invest ; 131(16)2021 08 16.
Article de Anglais | MEDLINE | ID: mdl-34228641

RÉSUMÉ

Myeloid-derived suppressor cells (MDSCs) are major negative regulators of immune responses in cancer and chronic infections. It remains unclear if regulation of MDSC activity in different conditions is controlled by similar mechanisms. We compared MDSCs in mice with cancer and lymphocytic choriomeningitis virus (LCMV) infection. Chronic LCMV infection caused the development of monocytic MDSCs (M-MDSCs) but did not induce polymorphonuclear MDSCs (PMN-MDSCs). In contrast, both MDSC populations were present in cancer models. An acquisition of immune-suppressive activity by PMN-MDSCs in cancer was controlled by IRE1α and ATF6 pathways of the endoplasmic reticulum (ER) stress response. Abrogation of PMN-MDSC activity by blockade of the ER stress response resulted in an increase in tumor-specific immune response and reduced tumor progression. In contrast, the ER stress response was dispensable for suppressive activity of M-MDSCs in cancer and LCMV infection. Acquisition of immune-suppressive activity by M-MDSCs in spleens was mediated by IFN-γ signaling. However, it was dispensable for suppressive activity of M-MDSCs in tumor tissues. Suppressive activity of M-MDSCs in tumors was retained due to the effect of IL-6 present at high concentrations in the tumor site. These results demonstrate disease- and population-specific mechanisms of MDSC accumulation and the need for targeting different pathways to achieve inactivation of these cells.


Sujet(s)
Cellules myéloïdes suppressives/immunologie , Tumeurs/immunologie , Maladies virales/immunologie , Animaux , Lignée cellulaire tumorale , Maladie chronique , Stress du réticulum endoplasmique/génétique , Stress du réticulum endoplasmique/immunologie , Femelle , Humains , Tolérance immunitaire/génétique , Interféron gamma/immunologie , Chorioméningite lymphocytaire/génétique , Chorioméningite lymphocytaire/immunologie , Chorioméningite lymphocytaire/virologie , Virus de la chorioméningite lymphocytaire/classification , Virus de la chorioméningite lymphocytaire/immunologie , Virus de la chorioméningite lymphocytaire/pathogénicité , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Cellules myéloïdes suppressives/classification , Cellules myéloïdes suppressives/métabolisme , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs expérimentales/génétique , Tumeurs expérimentales/immunologie , Tumeurs expérimentales/métabolisme , Transcriptome , Maladies virales/génétique , Maladies virales/métabolisme
5.
Cancer Discov ; 11(11): 2828-2845, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34230008

RÉSUMÉ

Mutations in the STK11 (LKB1) gene regulate resistance to PD-1/PD-L1 blockade. This study evaluated this association in patients with nonsquamous non-small cell lung cancer (NSCLC) enrolled in three phase I/II trials. STK11 mutations were associated with resistance to the anti-PD-L1 antibody durvalumab (alone/with the anti-CTLA4 antibody tremelimumab) independently of KRAS mutational status, highlighting STK11 as a potential driver of resistance to checkpoint blockade. Retrospective assessments of tumor tissue, whole blood, and serum revealed a unique immune phenotype in patients with STK11 mutations, with increased expression of markers associated with neutrophils (i.e., CXCL2, IL6), Th17 contexture (i.e., IL17A), and immune checkpoints. Associated changes were observed in the periphery. Reduction of STAT3 in the tumor microenvironment using an antisense oligonucleotide reversed immunotherapy resistance in preclinical STK11 knockout models. These results suggest that STK11 mutations may hinder response to checkpoint blockade through mechanisms including suppressive myeloid cell biology, which could be reversed by STAT3-targeted therapy. SIGNIFICANCE: Patients with nonsquamous STK11-mutant (STK11mut) NSCLC are less likely than STK11 wild-type (STK11wt) patients to respond to anti-PD-L1 ± anti-CTLA4 immunotherapies, and their tumors show increased expression of genes and cytokines that activate STAT3 signaling. Preclinically, STAT3 modulation reverses this resistance, suggesting STAT3-targeted agents as potential combination partners for immunotherapies in STK11mut NSCLC.This article is highlighted in the In This Issue feature, p. 2659.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , AMP-activated protein kinase kinases , Anticorps monoclonaux , Anticorps monoclonaux humanisés , Antigène CD274/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mutation , Protein-Serine-Threonine Kinases/génétique , Études rétrospectives , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme , Microenvironnement tumoral
6.
J Immunother Cancer ; 9(6)2021 06.
Article de Anglais | MEDLINE | ID: mdl-34145033

RÉSUMÉ

BACKGROUND: Immuno-oncology therapies are now part of the standard of care for cancer in many indications. However, durable objective responses remain limited to a subset of patients. As such, there is a critical need to identify biomarkers that can predict or enrich for treatment response. So far, the majority of putative biomarkers consist of features of the tumor microenvironment (TME). However, in preclinical mouse models, the collection of tumor tissue for this type of analysis is a terminal procedure, obviating the ability to directly link potential biomarkers to long-term treatment outcomes. METHODS: To address this, we developed and validated a novel non-terminal tumor sampling method to enable biopsy of the TME in mouse models based on fine needle aspiration. RESULTS: We show that this technique enables repeated in-life sampling of subcutaneous flank tumors and yields sufficient material to support downstream analyses of tumor-infiltrating immune cells using methods such as flow cytometry and single-cell transcriptomics. Moreover, using this technique we demonstrate that we can link TME biomarkers to treatment response outcomes, which is not possible using the current method of terminal tumor sampling. CONCLUSION: Thus, this minimally invasive technique is an important refinement for the pharmacodynamic analysis of the TME facilitating paired evaluation of treatment response biomarkers with outcomes and reducing the number of animals used in preclinical research.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Cytoponction/méthodes , Immunothérapie/méthodes , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Humains , Souris
7.
Cancer Discov ; 11(5): 1100-1117, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33419761

RÉSUMÉ

The clinical benefit of PD-1 blockade can be improved by combination with CTLA4 inhibition but is commensurate with significant immune-related adverse events suboptimally limiting the doses of anti-CTLA4 mAb that can be used. MEDI5752 is a monovalent bispecific antibody designed to suppress the PD-1 pathway and provide modulated CTLA4 inhibition favoring enhanced blockade on PD-1+ activated T cells. We show that MEDI5752 preferentially saturates CTLA4 on PD-1+ T cells versus PD-1- T cells, reducing the dose required to elicit IL2 secretion. Unlike conventional PD-1/CTLA4 mAbs, MEDI5752 leads to the rapid internalization and degradation of PD-1. Moreover, we show that MEDI5752 preferentially localizes and accumulates in tumors providing enhanced activity when compared with a combination of mAbs targeting PD-1 and CTLA4 in vivo. Following treatment with MEDI5752, robust partial responses were observed in two patients with advanced solid tumors. MEDI5752 represents a novel immunotherapy engineered to preferentially inhibit CTLA4 on PD-1+ T cells. SIGNIFICANCE: The unique characteristics of MEDI5752 represent a novel immunotherapy engineered to direct CTLA4 inhibition to PD-1+ T cells with the potential for differentiated activity when compared with current conventional mAb combination strategies targeting PD-1 and CTLA4. This molecule therefore represents a step forward in the rational design of cancer immunotherapy.See related commentary by Burton and Tawbi, p. 1008.This article is highlighted in the In This Issue feature, p. 995.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Adénocarcinome/traitement médicamenteux , Adénocarcinome à cellules claires/traitement médicamenteux , Antigène CTLA-4/métabolisme , Humains , Immunothérapie , Tumeurs du rein/traitement médicamenteux , Mâle , Adulte d'âge moyen , Récepteur-1 de mort cellulaire programmée/métabolisme , Tumeurs de l'estomac/traitement médicamenteux , Lymphocytes T/immunologie
8.
Clin Cancer Res ; 26(23): 6284-6298, 2020 12 01.
Article de Anglais | MEDLINE | ID: mdl-32817076

RÉSUMÉ

PURPOSE: While immune checkpoint inhibitors such as anti-PD-L1 are rapidly becoming the standard of care in the treatment of many cancers, only a subset of treated patients have long-term responses. IL12 promotes antitumor immunity in mouse models; however, systemic recombinant IL12 had significant toxicity and limited efficacy in early clinical trials. EXPERIMENTAL DESIGN: We therefore designed a novel intratumoral IL12 mRNA therapy to promote local IL12 tumor production while mitigating systemic effects. RESULTS: A single intratumoral dose of mouse (m)IL12 mRNA induced IFNγ and CD8+ T-cell-dependent tumor regression in multiple syngeneic mouse models, and animals with a complete response demonstrated immunity to rechallenge. Antitumor activity of mIL12 mRNA did not require NK and NKT cells. mIL12 mRNA antitumor activity correlated with TH1 tumor microenvironment (TME) transformation. In a PD-L1 blockade monotherapy-resistant model, antitumor immunity induced by mIL12 mRNA was enhanced by anti-PD-L1. mIL12 mRNA also drove regression of uninjected distal lesions, and anti-PD-L1 potentiated this response. Importantly, intratumoral delivery of mRNA encoding membrane-tethered mIL12 also drove rejection of uninjected lesions with very limited circulating IL12p70, supporting the hypothesis that local IL12 could induce a systemic antitumor immune response against distal lesions. Furthermore, in ex vivo patient tumor slice cultures, human IL12 mRNA (MEDI1191) induced dose-dependent IL12 production, downstream IFNγ expression and TH1 gene expression. CONCLUSIONS: These data demonstrate the potential for intratumorally delivered IL12 mRNA to promote TH1 TME transformation and robust antitumor immunity.See related commentary by Cirella et al., p. 6080.


Sujet(s)
Tumeurs colorectales/prévention et contrôle , Interleukine-12/administration et posologie , Lymphocytes TIL/immunologie , Mélanome/prévention et contrôle , ARN messager/administration et posologie , Lymphocytes auxiliaires Th1/immunologie , Microenvironnement tumoral/immunologie , Animaux , Anticorps monoclonaux/pharmacologie , Apoptose , Antigène CD274/antagonistes et inhibiteurs , Lymphocytes T CD8+ , Prolifération cellulaire , Tumeurs colorectales/génétique , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Résistance aux médicaments antinéoplasiques , Femelle , Humains , Interleukine-12/génétique , Mélanome/génétique , Mélanome/immunologie , Mélanome/anatomopathologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris nude , Souris SCID , ARN messager/génétique , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Mol Cancer Ther ; 17(5): 1024-1038, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29545330

RÉSUMÉ

Ligation of OX40 (CD134, TNFRSF4) on activated T cells by its natural ligand (OX40L, CD252, TNFSF4) enhances cellular survival, proliferation, and effector functions such as cytokine release and cellular cytotoxicity. We engineered a recombinant human OX40L IgG4P Fc fusion protein termed MEDI6383 that assembles into a hexameric structure and exerts potent agonist activity following engagement of OX40. MEDI6383 displayed solution-phase agonist activity that was enhanced when the fusion protein was clustered by Fc gamma receptors (FcγRs) on the surface of adjacent cells. The resulting costimulation of OX40 on T cells induced NFκB promoter activity in OX40-expressing T cells and induced Th1-type cytokine production, proliferation, and resistance to regulatory T cell (Treg)-mediated suppression. MEDI6383 enhanced the cytolytic activity of tumor-reactive T cells and reduced tumor growth in the context of an alloreactive human T cell:tumor cell admix model in immunocompromised mice. Consistent with the role of OX40 costimulation in the expansion of memory T cells, MEDI6383 administered to healthy nonhuman primates elicited peripheral blood CD4 and CD8 central and effector memory T-cell proliferation as well as B-cell proliferation. Together, these results suggest that OX40 agonism has the potential to enhance antitumor immunity in human malignancies. Mol Cancer Ther; 17(5); 1024-38. ©2018 AACR.


Sujet(s)
Fragments Fc des immunoglobulines/immunologie , Immunoglobuline G/immunologie , Ligand de OX40/immunologie , Protéines de fusion recombinantes/immunologie , Animaux , Lignée cellulaire tumorale , Cytokines/immunologie , Cytokines/métabolisme , Cytotoxicité immunologique/effets des médicaments et des substances chimiques , Cytotoxicité immunologique/immunologie , Femelle , Cellules HEK293 , Humains , Fragments Fc des immunoglobulines/génétique , Fragments Fc des immunoglobulines/métabolisme , Immunoglobuline G/génétique , Immunoglobuline G/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Macaca mulatta , Ligand de OX40/génétique , Ligand de OX40/métabolisme , Multimérisation de protéines/immunologie , Récepteur au OX40/agonistes , Récepteur au OX40/immunologie , Récepteur au OX40/métabolisme , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/pharmacologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme
10.
Mol Ther ; 25(8): 1917-1932, 2017 08 02.
Article de Anglais | MEDLINE | ID: mdl-28578991

RÉSUMÉ

Vesicular stomatitis virus encoding the IFNß transgene (VSV-IFNß) is a mediator of potent oncolytic activity and is undergoing clinical evaluation for the treatment of solid tumors. Emerging preclinical and clinical data suggest treatment of tumors with oncolytic viruses may sensitize tumors to checkpoint inhibitors and increase the anti-tumor immune response. New generations of immuno-oncology molecules including T cell agonists are entering clinical development and could be hypothesized to enhance the activity of oncolytic viruses, including VSV-IFNß. Here, we show that VSV-IFNß exhibits multiple mechanisms of action, including direct cell killing, stimulation of an innate immune response, recruitment of CD8 T cells, and depletion of T regulatory cells. Moreover, VSV-IFNß promotes the establishment of a CD8 T cell response to endogenous tumor antigens. Our data demonstrate a significant enhancement of anti-tumor function for VSV-IFNß when combined with checkpoint inhibitors, but not OX40 agonists. While the addition of checkpoint inhibitors to VSV-IFNß generated robust tumor growth inhibition, it resulted in no increase in viral replication, transgene expression, or immunophenotypic changes beyond treatment with VSV-IFNß alone. We hypothesize that tumor-specific T cells generated by VSV-IFNß retain activity due to a lack of immune exhaustion when checkpoint inhibitors were used.


Sujet(s)
Thérapie génétique , Vecteurs génétiques/génétique , Immunothérapie , Tumeurs/génétique , Tumeurs/immunologie , Thérapie virale de cancers , Virus oncolytiques/génétique , Virus de la stomatite vésiculeuse de type Indiana/génétique , Animaux , Anticorps monoclonaux/pharmacologie , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Marqueurs biologiques tumoraux , Association thérapeutique , Modèles animaux de maladie humaine , Expression des gènes , Thérapie génétique/méthodes , Humains , Immunomodulation , Immunothérapie/méthodes , Interféron bêta/génétique , Interféron bêta/métabolisme , Interférons/génétique , Interférons/métabolisme , Mélanome expérimental , Souris , Tumeurs/anatomopathologie , Tumeurs/thérapie , Récepteur au OX40/agonistes , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Transduction génétique , Transgènes , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Réplication virale , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Cancer Res ; 77(10): 2686-2698, 2017 05 15.
Article de Anglais | MEDLINE | ID: mdl-28283653

RÉSUMÉ

Immunogenic cell death (ICD) is the process by which certain cytotoxic drugs induce apoptosis of tumor cells in a manner that stimulates the immune system. In this study, we investigated whether antibody-drug conjugates (ADCS) conjugated with pyrrolobenzodiazepine dimer (PBD) or tubulysin payloads induce ICD, modulate the immune microenvironment, and could combine with immuno-oncology drugs to enhance antitumor activity. We show that these payloads on their own induced an immune response that prevented the growth of tumors following subsequent tumor cell challenge. ADCs had greater antitumor activity in immunocompetent versus immunodeficient mice, demonstrating a contribution of the immune system to the antitumor activity of these ADCs. ADCs also induced immunologic memory. In the CT26 model, depletion of CD8+ T cells abrogated the activity of ADCs when used alone or in combination with a PD-L1 antibody, confirming a role for T cells in antitumor activity. Combinations of ADCs with immuno-oncology drugs, including PD-1 or PD-L1 antibodies, OX40 ligand, or GITR ligand fusion proteins, produced synergistic antitumor responses. Importantly, synergy was observed in some cases with suboptimal doses of ADCs, potentially providing an approach to achieve potent antitumor responses while minimizing ADC-induced toxicity. Immunophenotyping studies in different tumor models revealed broad immunomodulation of lymphoid and myeloid cells by ADC and ADC/immuno-oncology combinations. These results suggest that it may be possible to develop novel combinatorial therapies with PBD- and tubulysin-based ADC and immuno-oncology drugs that may increase clinical responses. Cancer Res; 77(10); 2686-98. ©2017 AACR.


Sujet(s)
Antinéoplasiques/pharmacologie , Benzodiazépines/pharmacologie , Immunoconjugués/pharmacologie , Facteurs immunologiques/pharmacologie , Pyrroles/pharmacologie , Animaux , Anticorps monoclonaux/immunologie , Marqueurs biologiques , Vaccins anticancéreux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Synergie des médicaments , Femelle , Humains , Mémoire immunologique , Immunophénotypage , Immunothérapie , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Souris , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Rats , Sous-populations de lymphocytes T/effets des médicaments et des substances chimiques , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Clin Cancer Res ; 23(13): 3416-3427, 2017 07 01.
Article de Anglais | MEDLINE | ID: mdl-28069723

RÉSUMÉ

Purpose: To generate and characterize a murine GITR ligand fusion protein (mGITRL-FP) designed to maximize valency and the potential to agonize the GITR receptor for cancer immunotherapy.Experimental Design: The EC50 value of the mGITRL-FP was compared with an anti-GITR antibody in an in vitro agonistic cell-based reporter assay. We assessed the impact of dose, schedule, and Fc isotype on antitumor activity and T-cell modulation in the CT26 tumor model. The activity of the mGITRL-FP was compared with an agonistic murine OX40L-FP targeting OX40, in CT26 and B16F10-Luc2 tumor models. Combination of the mGITRL-FP with antibodies targeting PD-L1, PD-1, or CTLA-4 was analyzed in mice bearing CT26 tumors.Results: The mGITRL-FP had an almost 50-fold higher EC50 value compared with an anti-murine GITR antibody. Treatment of CT26 tumor-bearing mice with mGITRL-FP-mediated significant antitumor activity that was dependent on isotype, dose, and duration of exposure. The antitumor activity could be correlated with the increased proliferation of peripheral CD8+ and CD4+ T cells and a significant decrease in the frequency of intratumoral Tregs. The combination of mGITRL-FP with mOX40L-FP or checkpoint inhibitor antagonists enhanced antitumor immunity above that of monotherapy treatment.Conclusions: These results suggest that therapeutically targeting GITR represents a unique approach to cancer immunotherapy and suggests that a multimeric fusion protein may provide increased agonistic potential versus an antibody. In addition, these data provide, for the first time, early proof of concept for the potential combination of GITR targeting agents with OX40 agonists and PD-L1 antagonists. Clin Cancer Res; 23(13); 3416-27. ©2017 AACR.


Sujet(s)
Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Mélanome expérimental/immunologie , Protéines de fusion oncogènes/administration et posologie , Facteurs de nécrose tumorale/immunologie , Animaux , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Modèles animaux de maladie humaine , Protéine associée au récepteur du TNF induit par les corticoïdes/administration et posologie , Humains , Mélanome expérimental/génétique , Mélanome expérimental/thérapie , Glycoprotéines membranaires/agonistes , Glycoprotéines membranaires/immunologie , Souris , Ligand de OX40 , Protéines de fusion oncogènes/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Facteurs de nécrose tumorale/agonistes , Facteurs de nécrose tumorale/génétique
13.
Sci Rep ; 7: 40098, 2017 01 09.
Article de Anglais | MEDLINE | ID: mdl-28067257

RÉSUMÉ

Bispecific antibodies are considered attractive bio-therapeutic agents owing to their ability to target two distinct disease mediators. Cross-arm avidity targeting of antigen double-positive cancer cells over single-positive normal tissue is believed to enhance the therapeutic efficacy, restrict major escape mechanisms and increase tumor-targeting selectivity, leading to reduced systemic toxicity and improved therapeutic index. However, the interplay of factors regulating target selectivity is not well understood and often overlooked when developing clinically relevant bispecific therapeutics. We show in vivo that dual targeting alone is not sufficient to endow selective tumor-targeting, and report the pivotal roles played by the affinity of the individual arms, overall avidity and format valence. Specifically, a series of monovalent and bivalent bispecific IgGs composed of the anti-HER2 trastuzumab moiety paired with affinity-modulated VH and VL regions of the anti-EGFR GA201 mAb were tested for selective targeting and eradication of double-positive human NCI-H358 non-small cell lung cancer target tumors over single-positive, non-target NCI-H358-HER2 CRISPR knock out tumors in nude mice bearing dual-flank tumor xenografts. Affinity-reduced monovalent bispecific variants, but not their bivalent bispecific counterparts, mediated a greater degree of tumor targeting selectivity, while the overall efficacy against the targeted tumor was not substantially affected.


Sujet(s)
Anticorps bispécifiques/immunologie , Affinité des anticorps , Spécificité des anticorps , Carcinome pulmonaire non à petites cellules/immunologie , Trastuzumab/immunologie , Animaux , Anticorps bispécifiques/administration et posologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Lignée cellulaire tumorale , Récepteurs ErbB/immunologie , Humains , Souris nude , Récepteur ErbB-2/immunologie , Trastuzumab/administration et posologie , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Oncoimmunology ; 5(8): e1208875, 2016 Aug.
Article de Anglais | MEDLINE | ID: mdl-27622077

RÉSUMÉ

MEDI9447 is a human monoclonal antibody that is specific for the ectoenzyme CD73 and currently undergoing Phase I clinical trials. Here we show that MEDI9447 is a potent inhibitor of CD73 ectonucleotidase activity, with wide ranging immune regulatory consequences. MEDI9447 results in relief from adenosine monophosphate (AMP)-mediated lymphocyte suppression in vitro and inhibition of mouse syngeneic tumor growth in vivo. In contrast with other cancer immunotherapy agents such as checkpoint inhibitors or T-cell agonists, MEDI9447 drives changes in both myeloid and lymphoid infiltrating leukocyte populations within the tumor microenvironment of mouse models. Changes include significant alterations in a number of tumor micro-environmental subpopulations including increases in CD8(+) effector cells and activated macrophages. Furthermore, these changes correlate directly with responder and non-responder subpopulations within animal studies using syngeneic tumors. Combination data showing additive activity between MEDI9447 and anti-PD-1 antibodies using human cells in vitro and mouse tumor models further demonstrate the potential value of relieving adenosine-mediated immunosuppression. Based on these data, a Phase I study to test the safety, tolerability, and clinical activity of MEDI9447 in cancer patients was initiated (NCT02503774).

15.
Cancer Immunol Res ; 3(9): 1052-62, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-25943534

RÉSUMÉ

Programmed cell-death 1 ligand 1 (PD-L1) is a member of the B7/CD28 family of proteins that control T-cell activation. Many tumors can upregulate expression of PD-L1, inhibiting antitumor T-cell responses and avoiding immune surveillance and elimination. We have identified and characterized MEDI4736, a human IgG1 monoclonal antibody that binds with high affinity and specificity to PD-L1 and is uniquely engineered to prevent antibody-dependent cell-mediated cytotoxicity. In vitro assays demonstrate that MEDI4736 is a potent antagonist of PD-L1 function, blocking interaction with PD-1 and CD80 to overcome inhibition of primary human T-cell activation. In vivo MEDI4736 significantly inhibits the growth of human tumors in a novel xenograft model containing coimplanted human T cells. This activity is entirely dependent on the presence of transplanted T cells, supporting the immunological mechanism of action for MEDI4736. To further determine the utility of PD-L1 blockade, an anti-mouse PD-L1 antibody was investigated in immunocompetent mice. Here, anti-mouse PD-L1 significantly improved survival of mice implanted with CT26 colorectal cancer cells. The antitumor activity of anti-PD-L1 was enhanced by combination with oxaliplatin, which resulted in increased release of HMGB1 within CT26 tumors. Taken together, our results demonstrate that inhibition of PD-L1 function can have potent antitumor activity when used as monotherapy or in combination in preclinical models, and suggest it may be a promising therapeutic approach for the treatment of cancer. MEDI4736 is currently in several clinical trials both alone and in combination with other agents, including anti-CTLA-4, anti-PD-1, and inhibitors of IDO, MEK, BRAF, and EGFR.


Sujet(s)
Anticorps monoclonaux/métabolisme , Anticorps monoclonaux/pharmacologie , Antigène CD274/antagonistes et inhibiteurs , Animaux , Anticorps monoclonaux/administration et posologie , Cytotoxicité à médiation cellulaire dépendante des anticorps/effets des médicaments et des substances chimiques , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Antigène CD80/métabolisme , Antigène CD274/métabolisme , Fixation compétitive , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Femelle , Humains , Activation des lymphocytes/effets des médicaments et des substances chimiques , Test de culture lymphocytaire mixte , Mélanome/immunologie , Mélanome/anatomopathologie , Mélanome/prévention et contrôle , Souris de lignée BALB C , Souris de lignée C57BL , Souris de lignée NOD , Composés organiques du platine/administration et posologie , Oxaliplatine , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/prévention et contrôle , Récepteur-1 de mort cellulaire programmée/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
16.
MAbs ; 6(6): 1571-84, 2014.
Article de Anglais | MEDLINE | ID: mdl-25484061

RÉSUMÉ

Individual or combinations of somatic mutations found in genes from colorectal cancers can redirect the effects of chemotherapy and targeted agents on cancer cell survival and, consequently, on clinical outcome. Novel therapeutics with mechanisms of action that are independent of mutational status would therefore fulfill a current unmet clinical need. Here the CEA and CD3 bispecific single-chain antibody MEDI-565 (also known as MT111 and AMG 211) was evaluated for its ability to activate T cells both in vitro and in vivo and to kill human tumor cell lines harboring various somatic mutations commonly found in colorectal cancers. MEDI-565 specifically bound to normal and malignant tissues in a CEA-specific manner, and only killed CEA positive cells. The BiTE® antibody construct mediated T cell-directed killing of CEA positive tumor cells within 6 hours, at low effector-to-target ratios which were independent of high concentrations of soluble CEA. The potency of in vitro lysis was dependent on CEA antigen density but independent of the mutational status in cancer cell lines. Importantly, individual or combinations of mutated KRAS and BRAF oncogenes, activating PI3KCA mutations, loss of PTEN expression, and loss-of-function mutations in TP53 did not reduce the activity in vitro. MEDI-565 also prevented growth of human xenograft tumors which harbored various mutations. These findings suggest that MEDI-565 represents a potential treatment option for patients with CEA positive tumors of diverse origin, including those with individual or combinations of somatic mutations that may be less responsive to chemotherapy and other targeted agents.


Sujet(s)
Anticorps bispécifiques/pharmacologie , Cytotoxicité immunologique/effets des médicaments et des substances chimiques , Activation des lymphocytes/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Lymphocytes T/effets des médicaments et des substances chimiques , Adénocarcinome/traitement médicamenteux , Adénocarcinome/génétique , Adénocarcinome/immunologie , Animaux , Anticorps bispécifiques/immunologie , Antigènes CD3/immunologie , Cellules CHO , Antigène carcinoembryonnaire/immunologie , Lignée cellulaire tumorale , Cellules cultivées , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/immunologie , Cricetinae , Cricetulus , Cytotoxicité immunologique/immunologie , Femelle , Cellules HT29 , Cellules HeLa , Humains , Activation des lymphocytes/immunologie , Mâle , Souris SCID , Mutation , Tumeurs/génétique , Tumeurs/immunologie , Liaison aux protéines/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Neoplasia ; 11(6): 509-17, 2 p following 517, 2009 Jun.
Article de Anglais | MEDLINE | ID: mdl-19484140

RÉSUMÉ

EphA2 is a receptor tyrosine kinase that has been shown to be overexpressed in a variety of human tumor types. Previous studies demonstrated that agonist monoclonal antibodies targeting EphA2 induced the internalization and degradation of the receptor, thereby abolishing its oncogenic effects. In this study, the in vitro and in vivo antibody-dependent cell-mediated cytotoxicity (ADCC) activity of EphA2 effector-enhanced agonist monoclonal antibodies was evaluated. With tumor cell lines and healthy human peripheral blood monocytes, the EphA2 antibodies demonstrated approximately 80% tumor cell killing. In a dose-dependent manner, natural killer (NK) cells were required for the in vitro ADCC activity and became activated as demonstrated by the induction of cell surface expression of CD107a. To assess the role of NK cells on antitumor efficacy in vivo, the EphA2 antibodies were evaluated in xenograft models in severe compromised immunodeficient (SCID) mice (which have functional NK cells and monocytes) and SCID nonobese diabetic (NOD) mice (which largely lack functional NK cells and monocytes). Dosing of EphA2 antibody in the SCID murine tumor model resulted in a 6.2-fold reduction in tumor volume, whereas the SCID/nonobese diabetic model showed a 1.6-fold reduction over the isotype controls. Together, these results demonstrate that the anti-EphA2 monoclonal antibodies may function through at least two mechanisms of action: EphA2 receptor activation and ADCC-mediated activity. These novel EphA2 monoclonal antibodies provide additional means by which host effector mechanisms can be activated for selective destruction of EphA2-expressing tumor cells.


Sujet(s)
Anticorps monoclonaux/immunologie , Cytotoxicité à médiation cellulaire dépendante des anticorps/immunologie , Tumeurs/immunologie , Récepteur EphA2/immunologie , Animaux , Anticorps monoclonaux/pharmacologie , Cytotoxicité à médiation cellulaire dépendante des anticorps/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Femelle , Génotype , Humains , Fragments Fc des immunoglobulines/immunologie , Cellules tueuses naturelles/cytologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/immunologie , Protéine de membrane-1 associée au lysosome/immunologie , Tumeurs expérimentales de la mamelle/traitement médicamenteux , Tumeurs expérimentales de la mamelle/immunologie , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris de lignée NOD , Souris nude , Souris SCID , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Phosphorylation/effets des médicaments et des substances chimiques , Polymorphisme génétique , Récepteur EphA2/agonistes , Récepteur EphA2/métabolisme , Récepteurs du fragment Fc des IgG/génétique , Résonance plasmonique de surface , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
18.
J Immunother ; 32(4): 341-52, 2009 May.
Article de Anglais | MEDLINE | ID: mdl-19342971

RÉSUMÉ

Carcinoembryonic antigen (CEA, CD66e) is a well-characterized tumor-associated antigen that is frequently overexpressed in tumors. Phospholipases release CEA from tumor cells resulting in high circulating serum levels of soluble CEA (sCEA) that has been validated as marker for progression of colorectal, breast, and lung cancers. sCEA also acts as a competitive inhibitor for anticancer strategies targeting membrane-bound CEA. As a novel therapeutic approach for treatment of tumors expressing CEA on their cell surface, we constructed a series of bispecific single-chain antibodies (bscAb) combining various single-chain variable fragments recognizing human CEA with a deimmunized single-chain variable fragments recognizing human CD3. CEA/CD3-bscAbs redirected human T cells to lyse CEA-expressing tumor cells in vitro and in vivo. Efficient tumor cell lysis was achieved in vitro at bscAb concentrations from 1 pg/mL (19 fM) to 8.9 pg/mL with preactivated CD8 T cells, and 200 to 500 pg/mL with unstimulated peripheral blood mononuclear cell. The cytotoxic activity of a subset of CEA/CD3-bscAbs was not competitively inhibited by sCEA at concentrations that exceeded levels found in the serum of most cancer patients. Treatment with CEA/CD3-bscAbs prevented the growth of human colorectal cancer lines in a severe combined immunodeficiency mouse model modified to show human T cell killing of tumors. A murine surrogate CEA/CD3-bscAb capable of recruiting murine T cells for redirected tumor lysis in immunocompetent mice prevented the growth of lung tumors expressing human CEA. Together, our results reveal a unique opportunity for targeting cytotoxic T cells toward CEA-expressing tumors without being competitively inhibited by sCEA and establish CEA/CD3-bscAb as a promising and potent therapeutic approach.


Sujet(s)
Anticorps bispécifiques/usage thérapeutique , Antigènes CD3/immunologie , Lymphocytes T CD8+/immunologie , Antigène carcinoembryonnaire/immunologie , Tumeurs colorectales/thérapie , Protéines de fusion recombinantes/usage thérapeutique , Animaux , Anticorps bispécifiques/immunologie , Lymphocytes T CD8+/métabolisme , Cellules CHO , Antigène carcinoembryonnaire/sang , Cricetinae , Cricetulus , Humains , Immunothérapie , Souris , Souris SCID , Protéines de fusion recombinantes/immunologie , Dihydrofolate reductase/génétique , Dihydrofolate reductase/immunologie
19.
Mol Cancer Ther ; 5(12): 3122-9, 2006 Dec.
Article de Anglais | MEDLINE | ID: mdl-17172415

RÉSUMÉ

The humanized monoclonal antibody Abegrin, currently in phase II trials for treatment of solid tumors, specifically recognizes the integrin alphavbeta3. Due to its high expression on mature osteoclasts, angiogenic endothelial cells, and tumor cells, integrin alphavbeta3 functions in several pathologic processes important to tumor growth and metastasis. Targeting of this integrin with Abegrin results in antitumor, antiangiogenic, and antiosteolytic activities. Here, we exploit the species specificity of Abegrin to evaluate the effects of direct targeting of tumor cells (independent of targeting of endothelia or osteoclasts). Flow cytometry analysis of human tumor cell lines shows high levels of alphavbeta3 on many solid tumors, including cancers of the prostate, skin, ovary, kidney, lung, and breast. We also show that tumor growth of alphavbeta3-expressing tumor cells is inhibited by Abegrin in a dose-dependent manner. We present a novel finding that high-dose administration can actively impair the antitumor activity of Abegrin. We also provide evidence that antibody-dependent cellular cytotoxicity contributes to in vitro and in vivo antitumor activity. Finally, it was observed that peak biological activity of Abegrin arises at serum levels that are consistent with those achieved in clinical trials. These results support a concept that Abegrin can be used to achieve selective targeting of the many tumor cells that express alphavbeta3 integrin. In combination with the well-established concept that alphavbeta3 plays a key role in cancer-associated angiogenesis and osteolytic activities, this triad of activity could provide new opportunities for therapeutic targeting of cancer.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Intégrine alphaVbêta3/immunologie , Tumeurs/thérapie , Animaux , Anticorps monoclonaux/immunologie , Anticorps monoclonaux humanisés , Cytotoxicité à médiation cellulaire dépendante des anticorps , Lignée cellulaire tumorale , Relation dose-réponse (immunologie) , Femelle , Humains , Intégrine alphaVbêta3/biosynthèse , Souris , Souris nude , Souris SCID , Tumeurs/immunologie , Spécificité d'espèce , Tests d'activité antitumorale sur modèle de xénogreffe
20.
Hum Gene Ther ; 14(8): 777-87, 2003 May 20.
Article de Anglais | MEDLINE | ID: mdl-12804140

RÉSUMÉ

Adenoviral vectors used in gene therapy are predominantly derived from adenovirus serotype 5 (Ad5), which infects a broad range of cells. Ad5 cell entry involves interactions with the coxsackie-adenovirus receptor (CAR) and integrins. To assess these receptors in vivo, we mutated amino acid residues in fiber and penton that are involved in receptor interaction and showed that CAR and integrins play a minor role in hepatic transduction but that integrins can influence gene delivery to other tissues. These data suggest that an alternative entry pathway exists for hepatocyte transduction in vivo that is more important than CAR or integrins. In vitro data suggest a role for heparan sulfate glycosaminoglycans (HSG) in adenovirus transduction. The role of the fiber shaft in liver uptake was examined by introducing specific amino acid changes into a putative HSG-binding motif contained within the shaft or by preparing fiber shaft chimeras between Ad5 and Ad35 fibers. Results were obtained that demonstrate that the Ad5 fiber shaft can influence gene transfer both in vitro and to the liver in vivo. These observations indicate that the currently accepted two-step entry pathway, which involves CAR and integrins, described for adenoviral infection in vitro, is not used for hepatic gene transfer in vivo. In contrast, alpha(v) integrins influence gene delivery to the lung, spleen, heart, and kidney. The detargeted vector constructs described here may provide a foundation for the development of targeted adenoviral vectors.


Sujet(s)
Adenoviridae/génétique , Protéines de capside/génétique , Vecteurs génétiques/pharmacocinétique , Transduction génétique , Motifs d'acides aminés , Animaux , Technique de Western , Protéines de capside/métabolisme , Lignée cellulaire , Cellules cultivées , Protéine membranaire apparentée au récepteur des coxsackievirus et adénovirus , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/sang , Glycosaminoglycanes/physiologie , Hépatocytes/métabolisme , Humains , Intégrines/métabolisme , Mâle , Souris , Souris de lignée C57BL , Mutation , Oligopeptides/génétique , Oligopeptides/métabolisme , Récepteurs viraux/métabolisme , Virion/métabolisme , beta-Galactosidase/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE