Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 97
Filtrer
1.
Cancer Immunol Res ; 12(8): 1022-1038, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38842347

RÉSUMÉ

Despite clinical evidence of antitumor activity, the development of cytokine therapies has been hampered by a narrow therapeutic window and limited response rates. Two cytokines of high interest for clinical development are interleukin 2 (IL2) and interleukin 12 (IL12), which potently synergize to promote the activation and proliferation of T cells and NK cells. However, the only approved human IL2 therapy, Proleukin, is rarely used in the clinic due to systemic toxicities, and no IL12 product has been approved to date due to severe dose-limiting toxicities. Here, we describe CLN-617, a first-in-class therapeutic for intratumoral (IT) injection that co-delivers IL2 and IL12 on a single molecule in a safe and effective manner. CLN-617 is a single-chain fusion protein comprised of IL2, leukocyte-associated immunoglobulin-like receptor 2 (LAIR2), human serum albumin (HSA), and IL12. LAIR2 and HSA function to retain CLN-617 in the treated tumor by binding collagen and increasing molecular weight, respectively. We found that IT administration of a murine surrogate of CLN-617, mCLN-617, eradicated established treated and untreated tumors in syngeneic models, significantly improved response to anti-PD1 checkpoint therapy, and generated a robust abscopal response dependent on cellular immunity and antigen cross-presentation. CLN-617 is being evaluated in a clinical trial in patients with advanced solid tumors (NCT06035744).


Sujet(s)
Interleukine-12 , Interleukine-2 , Animaux , Femelle , Humains , Souris , Lignée cellulaire tumorale , Interleukine-12/métabolisme , Interleukine-2/usage thérapeutique , Interleukine-2/pharmacologie , Souris de lignée C57BL , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Protéines de fusion recombinantes/pharmacologie , Protéines de fusion recombinantes/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
2.
J Exp Med ; 221(5)2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38587494

RÉSUMÉ

Jennifer S. Michaelson, Chief Scientific Officer at Cullinan Oncology, and Patrick A. Baeuerle, scientific advisor to Cullinan Oncology and honorary professor in immunology at Ludwig Maximilians University Munich, discuss the use of CD19-specific T cell-engaging antibody therapies (TCEs) as therapeutics for autoimmune diseases.


Sujet(s)
Maladies auto-immunes , Humains , Maladies auto-immunes/thérapie , Protéines adaptatrices de la transduction du signal , Antigènes CD19 , Lymphocytes T
3.
Cancer Immunol Immunother ; 72(12): 4195-4207, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37848682

RÉSUMÉ

T cells expressing a mesothelin (MSLN)-specific T cell receptor fusion construct (TRuC®), called TC-210, have demonstrated robust antitumor activity in preclinical models of mesothelioma, ovarian cancer, and lung cancer. However, they are susceptible to suppression by the programmed cell death protein 1 (PD-1)/programmed cell death protein ligand 1 (PD-L1) axis and lack intrinsic costimulatory signaling elements. To enhance the function of anti-MSLN TRuC-T cells, chimeric switch receptors (CSRs) have been designed to co-opt the immunosuppressive PD-1/PD-L1 axis and to deliver a CD28-mediated costimulatory signal. Here, we report that coexpression of the PD1-CD28 CSR in TRuC-T cells enhanced T cell receptor signaling, increased proinflammatory effector cytokines, decreased anti-inflammatory cytokines, and sustained effector function in the presence of PD-L1 when compared with TC-210. Anti-MSLN TRuC-T cells engineered to coexpress PD1-CD28 CSRs comprising the ectodomain of PD-1 and the intracellular domain of CD28 linked by the transmembrane domain of PD-1 were selected for integration into an anti-MSLN TRuC-T cell therapy product called TC-510. In vitro, TC-510 showed significant improvements in persistence and resistance to exhaustion upon chronic stimulation by tumor cells expressing MSLN and PD-L1 when compared with TC-210. In vivo, TC-510 showed a superior ability to provide durable protection following tumor rechallenge, versus TC-210. These data demonstrate that integration of a PD1-CD28 CSR into TRuC-T cells improves effector function, resistance to exhaustion, and prolongs persistence. Based on these findings, TC-510 is currently being evaluated in patients with MSLN-expressing solid tumors.


Sujet(s)
Antigène CD28 , Mésothéliome , Humains , Mésothéline , Récepteur-1 de mort cellulaire programmée/métabolisme , Antigène CD274/génétique , Antigène CD274/métabolisme , Lymphocytes T/métabolisme , Récepteurs aux antigènes des cellules T/métabolisme , Cytokines/métabolisme
4.
J Immunother Cancer ; 11(8)2023 08.
Article de Anglais | MEDLINE | ID: mdl-37586770

RÉSUMÉ

BACKGROUND: Despite significant progress in the development of T cell-engaging therapies for various B-cell malignancies, a high medical need remains for the refractory disease setting, often characterized by suboptimal target levels. METHODS: To address this issue, we have developed a 65-kDa multispecific antibody construct, CLN-978, with affinities tuned to optimize the killing of low-CD19 expressing tumor cells. CLN-978 bound to CD19 on B cells with picomolar affinity, and to CD3ε on T cells with nanomolar affinity. A serum albumin binding domain was incorporated to extend serum half-life. In this setting, we biophysically characterize and report the activities of CLN-978 in cell co-culture assays, multiple mouse models and non-human primates. RESULTS: Human T cells redirected by CLN-978 could eliminate target cells expressing less than 300 copies of CD19 on their surface. The half-life extension and high affinity for CD19 led to significant antitumor activity in murine lymphoma models at very low doses of CLN-978. In primates, we observed a long serum half-life, deep and sustained depletion of normal B cells, and remarkable tolerability, in particular, reduced cytokine release when CLN-978 was administered subcutaneously. CONCLUSIONS: CLN-978 warrants further exploration. An ongoing clinical phase 1 trial is investigating safety, pharmacokinetics, pharmacodynamics, and the initial therapeutic potential of subcutaneously administered CLN-978 in patients with non-Hodgkin's lymphoma.


Sujet(s)
Lymphome malin non hodgkinien , Tumeurs , Humains , Animaux , Souris , Période , Protéines adaptatrices de la transduction du signal , Anticorps , Antigènes CD19
5.
MAbs ; 15(1): 2208697, 2023.
Article de Anglais | MEDLINE | ID: mdl-37165468

RÉSUMÉ

The field of immuno-oncology has revolutionized cancer patient care and improved survival and quality of life for patients. Much of the focus in the field has been on exploiting the power of the adaptive immune response through therapeutic targeting of T cells. While these approaches have markedly advanced the field, some challenges remain, and the clinical benefit of T cell therapies does not extend to all patients or tumor indications. Alternative strategies, such as engaging the innate immune system, have become an intense area of focus in the field. In particular, the engagement of natural killer (NK) cells as potent effectors of the innate immune response has emerged as a promising modality in immunotherapy. Here, we review therapeutic approaches for selective engagement of NK cells for cancer therapy, with a particular focus on targeting the key activating receptors NK Group 2D (NKG2D) and cluster of differentiation 16A (CD16A).


Sujet(s)
Sous-famille K des récepteurs de cellules NK de type lectine , Tumeurs , Humains , Qualité de vie , Cellules tueuses naturelles , Tumeurs/thérapie , Immunothérapie
6.
Oncoimmunology ; 12(1): 2182058, 2023.
Article de Anglais | MEDLINE | ID: mdl-36875551

RÉSUMÉ

T cell Receptor (TCR) Fusion Construct (TRuC®) T cells harness all signaling subunits of the TCR to activate T cells and eliminate tumor cells, with minimal release of cytokines. While adoptive cell therapy with chimeric antigen receptor (CAR)-T cells has shown unprecedented clinical efficacy against B-cell malignancies, monotherapy with CAR-T cells has suboptimal clinical efficacy against solid tumors, probably because of the artificial signaling properties of the CAR. TRuC-T cells may address the suboptimal efficacy of existing CAR-T therapies for solid tumors. Here, we report that mesothelin (MSLN)-specific TRuC-T cells (referred to as TC-210 T cells) potently kill MSLN+ tumor cells in vitro and efficiently eradicate MSLN+ mesothelioma, lung, and ovarian cancers in xenograft mouse tumor models. When benchmarked against MSLN-targeted BBζ CAR-T cells (MSLN-BBζ CAR-T cells), TC-210 T cells show an overall comparable level of efficacy; however, TC-210 T cells consistently show faster tumor rejection kinetics that are associated with earlier intratumoral accumulation and earlier signs of activation. Furthermore, in vitro and ex vivo metabolic profiling suggests TC-210 T cells have lower glycolytic activity and higher mitochondrial metabolism than MSLN-BBζ CAR-T cells. These data highlight TC-210 T cells as a promising cell therapy for treating MSLN-expressing cancers. The differentiated profile from CAR-T cells may translate into better efficacy and safety of TRuC-T cells for solid tumors.


Sujet(s)
Mésothéliome malin , Tumeurs de l'ovaire , Humains , Animaux , Souris , Femelle , Lymphocytes T , Mésothéline , Récepteurs aux antigènes des cellules T , Modèles animaux de maladie humaine
7.
Curr Opin Oncol ; 34(5): 552-558, 2022 09 01.
Article de Anglais | MEDLINE | ID: mdl-35880455

RÉSUMÉ

PURPOSE OF REVIEW: T-cell-engaging antibodies or T-cell engagers (TCEs) can connect a patient's cytotoxic T cells with cancer cells, leading to potent redirected lysis. Until very recently, only one TCE was approved, the CD19/CD3-bispecific blinatumomab. Many new TCEs in late-stage clinical development target various hematopoietic lineage markers like CD20, BCMA, or CD123. Although very compelling single-agent activity of TCEs was observed with various blood-borne cancers, therapy of solid tumor indications has thus far been less successful. RECENT FINDINGS: The approval in 2022 of the gp100 peptide-major histocompatibility complex (MHC)/CD3 bispecific TCE tebentafusp in uveal melanoma confirms that TCEs can also efficiently work against solid tumors. TCEs targeting peptide-MHC complexes will expand the target space for solid tumor therapy to intracellular targets. Likewise, early clinical trial data from TCEs targeting DLL3 in small cell lunger cancer showed promising antitumor activity. Various technologies for conditional activation of TCEs in the tumor microenvironment (TME) may expand the scope of conventional surface targets that suffer from a narrow therapeutic window. Finally, pharmacological enhancements for TCE therapies by engagement of certain costimulatory receptors and cytokines, or blockade of checkpoints, are showing promise. SUMMARY: Targeting peptide-MHC complexes, conditional TCE technologies, and concepts enhancing TCE-activated T cells are paving the way towards overcoming challenges associated with solid tumor therapy.


Sujet(s)
Anticorps bispécifiques , Mélanome , Anticorps bispécifiques/usage thérapeutique , Antigènes CD19 , Antigènes CD3 , Humains , Immunothérapie , Protéines et peptides de signalisation intracellulaire , Mélanome/traitement médicamenteux , Protéines membranaires , Protéines de fusion recombinantes , Lymphocytes T , Microenvironnement tumoral
8.
J Immunother Cancer ; 10(3)2022 03.
Article de Anglais | MEDLINE | ID: mdl-35288466

RÉSUMÉ

BACKGROUND: In lymphoid malignancies, the introduction of chimeric antigen receptor T (CAR-T) cells and bispecific antibodies (bsAbs) has achieved remarkable clinical success. However, such immunotherapeutic strategies are not yet established for acute myeloid leukemia (AML), the most common form of acute leukemia in adults. Common targets in AML such as CD33, CD123, and CLEC12A are highly expressed on both AML blasts and on normal myeloid cells and hematopoietic stem cells (HSCs), thereby raising toxicity concerns. In B-cell acute lymphoblastic leukemia (B-ALL), bsAbs and CAR-T therapy targeting CD19 and CD22 have demonstrated clinical success, but resistance via antigen loss is common, motivating the development of agents focused on alternative targets. An attractive emerging target is FLT3, a proto-oncogene expressed in both AML and B-ALL, with low and limited expression on myeloid dendritic cells and HSCs. METHODS: We developed and characterized CLN-049, a T cell-activating bsAb targeting CD3 and FLT3, constructed as an IgG heavy chain/scFv fusion. CLN-049 binds the membrane proximal extracellular domain of the FLT3 protein tyrosine kinase, which facilitates the targeting of leukemic blasts regardless of FLT3 mutational status. CLN-049 was evaluated for preclinical safety and efficacy in vitro and in vivo. RESULTS: CLN-049 induced target-restricted activation of CD4+ and CD8+ T cells. AML cell lines expressing a broad range of surface levels of FLT3 were efficiently lysed on treatment with subnanomolar concentrations of CLN-049, whereas FLT3-expressing hematopoietic progenitor cells and dendritic cells were not sensitive to CLN-049 killing. Treatment with CLN-049 also induced lysis of AML and B-ALL patient blasts by autologous T cells at the low effector-to-target ratios typically observed in patients with overt disease. Lysis of leukemic cells was not affected by supraphysiological levels of soluble FLT3 or FLT3 ligand. In mouse xenograft models, CLN-049 was highly active against human leukemic cell lines and patient-derived AML and B-ALL blasts. CONCLUSIONS: CLN-049 has a favorable efficacy and safety profile in preclinical models, warranting evaluation of its antileukemic activity in the clinic.


Sujet(s)
Leucémie aigüe myéloïde , Leucémie-lymphome lymphoblastique à précurseurs B et T , Animaux , Humains , Immunoglobuline G/usage thérapeutique , Immunothérapie adoptive , Sous-unité alpha du récepteur à l'interleukine-3 , Lectines de type C , Leucémie aigüe myéloïde/traitement médicamenteux , Souris , Récepteur mitogène
9.
Clin Cancer Res ; 27(5): 1452-1462, 2021 03 01.
Article de Anglais | MEDLINE | ID: mdl-33262134

RÉSUMÉ

PURPOSE: Mesothelin (MSLN) is a glycophosphatidylinositol-linked tumor antigen overexpressed in a variety of malignancies, including ovarian, pancreatic, lung, and triple-negative breast cancer. Early signs of clinical efficacy with MSLN-targeting agents have validated MSLN as a promising target for therapeutic intervention, but therapies with improved efficacy are still needed to address the significant unmet medical need posed by MSLN-expressing cancers. EXPERIMENTAL DESIGN: We designed HPN536, a 53-kDa, trispecific, T-cell-activating protein-based construct, which binds to MSLN-expressing tumor cells, CD3ε on T cells, and to serum albumin. Experiments were conducted to assess the potency, activity, and half-life of HPN536 in in vitro assays, rodent models, and in nonhuman primates (NHP). RESULTS: HPN536 binds to MSLN-expressing tumor cells and to CD3ε on T cells, leading to T-cell activation and potent redirected target cell lysis. A third domain of HPN536 binds to serum albumin for extension of plasma half-life. In cynomolgus monkeys, HPN536 at doses ranging from 0.1 to 10 mg/kg demonstrated MSLN-dependent pharmacologic activity, was well tolerated, and showed pharmacokinetics in support of weekly dosing in humans. CONCLUSIONS: HPN536 is potent, is well tolerated, and exhibits extended half-life in NHPs. It is currently in phase I clinical testing in patients with MSLN-expressing malignancies (NCT03872206).


Sujet(s)
Immunothérapie/méthodes , Activation des lymphocytes/immunologie , Mésothéline/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Anticorps à domaine unique/pharmacologie , Lymphocytes T/immunologie , Animaux , Antigènes néoplasiques/immunologie , Apoptose , Prolifération cellulaire , Femelle , Humains , Macaca fascicularis , Mâle , Souris , Souris de lignée NOD , Souris SCID , Tumeurs/immunologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Fragments peptidiques/immunologie , Anticorps à domaine unique/immunologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Mol Cancer Ther ; 20(1): 109-120, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33203731

RÉSUMÉ

T cells have a unique capability to eliminate cancer cells and fight malignancies. Cancer cells have adopted multiple immune evasion mechanisms aimed at inhibiting T cells. Dramatically improved patient outcomes have been achieved with therapies genetically reprogramming T cells, blocking T-cell inhibition by cancer cells, or transiently connecting T cells with cancer cells for redirected lysis. This last modality is based on antibody constructs that bind a surface antigen on cancer cells and an invariant component of the T-cell receptor. Although high response rates were observed with T-cell engagers specific for CD19, CD20, or BCMA in patients with hematologic cancers, the treatment of solid tumors has been less successful. Here, we developed and characterized a novel T-cell engager format, called TriTAC (for Trispecific T-cell Activating Construct). TriTACs are engineered with features to improve patient safety and solid tumor activity, including high stability, small size, flexible linkers, long serum half-life, and highly specific and potent redirected lysis. The present study establishes the structure/activity relationship of TriTACs and describes the development of HPN424, a PSMA- (FOLH1-) targeting TriTAC in clinical development for patients with metastatic castration-resistant prostate cancer.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs/traitement médicamenteux , Lymphocytes T/métabolisme , Albumines/pharmacologie , Animaux , Antinéoplasiques/sang , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/pharmacologie , Antigènes CD3/métabolisme , Mort cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Période , Humains , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Macaca fascicularis , Souris de lignée NOD , Souris SCID , Tumeurs/anatomopathologie , Antigène spécifique de la prostate/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques
11.
Cancer Metastasis Rev ; 39(3): 969-987, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32507912

RÉSUMÉ

EpCAM (epithelial cell adhesion molecule) was discovered four decades ago as a tumor antigen on colorectal carcinomas. Owing to its frequent and high expression on carcinomas and their metastases, EpCAM serves as a prognostic marker, a therapeutic target, and an anchor molecule on circulating and disseminated tumor cells (CTCs/DTCs), which are considered the major source for metastatic cancer cells. Today, EpCAM is reckoned as a multi-functional transmembrane protein involved in the regulation of cell adhesion, proliferation, migration, stemness, and epithelial-to-mesenchymal transition (EMT) of carcinoma cells. To fulfill these functions, EpCAM is instrumental in intra- and intercellular signaling as a full-length molecule and following regulated intramembrane proteolysis, generating functionally active extra- and intracellular fragments. Intact EpCAM and its proteolytic fragments interact with claudins, CD44, E-cadherin, epidermal growth factor receptor (EGFR), and intracellular signaling components of the WNT and Ras/Raf pathways, respectively. This plethora of functions contributes to shaping intratumor heterogeneity and partial EMT, which are major determinants of the clinical outcome of carcinoma patients. EpCAM represents a marker for the epithelial status of primary and systemic tumor cells and emerges as a measure for the metastatic capacity of CTCs. Consequentially, EpCAM has reclaimed potential as a prognostic marker and target on primary and systemic tumor cells.


Sujet(s)
Molécule d'adhérence des cellules épithéliales/métabolisme , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Animaux , Molécule d'adhérence des cellules épithéliales/biosynthèse , Transition épithélio-mésenchymateuse , Humains
12.
Immunotherapy ; 12(1): 89-103, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31902264

RÉSUMÉ

Recently, two chimeric antigen receptor (CAR) T cell therapies were approved based on their remarkable efficacy in patients with hematological malignancies. By contrast, CAR-T cell therapies results in solid tumors have been less promising. To develop the next generation of T cell therapies a better understanding of T cell receptor (TCR) biology and its implication for the design of synthetic receptors is critical. Here, we review current and newly developed forms of T cell therapies and how their utilization of different components of the TCR signaling machinery and their requirement for engagement (or not) of human leukocyte antigen impacts their design, efficacy and applicability as cancer drugs. Notably, we highlight the development of human leukocyte antigen-independent T cell platforms that utilize the full TCR complex as having promise to overcome some of the limitations of existing T cell therapies.


Sujet(s)
Vaccins anticancéreux/immunologie , Immunothérapie adoptive/méthodes , Tumeurs/thérapie , Récepteurs aux antigènes des cellules T/métabolisme , Lymphocytes T/métabolisme , Animaux , Humains , Thérapie moléculaire ciblée , Tumeurs/immunologie , Ingénierie des protéines , Récepteurs aux antigènes des cellules T/génétique , Transduction du signal , Lymphocytes T/immunologie
13.
Nat Commun ; 10(1): 2087, 2019 05 07.
Article de Anglais | MEDLINE | ID: mdl-31064990

RÉSUMÉ

T cells expressing CD19-targeting chimeric antigen receptors (CARs) reveal high efficacy in the treatment of B cell malignancies. Here, we report that T cell receptor fusion constructs (TRuCs) comprising an antibody-based binding domain fused to T cell receptor (TCR) subunits can effectively reprogram an intact TCR complex to recognize tumor surface antigens. Unlike CARs, TRuCs become a functional component of the TCR complex. TRuC-T cells kill tumor cells as potently as second-generation CAR-T cells, but at significant lower cytokine release and despite the absence of an extra co-stimulatory domain. TRuC-T cells demonstrate potent anti-tumor activity in both liquid and solid tumor xenograft models. In several models, TRuC-T cells are more efficacious than respective CAR-T cells. TRuC-T cells are shown to engage the signaling capacity of the entire TCR complex in an HLA-independent manner.


Sujet(s)
Immunothérapie adoptive/méthodes , Tumeurs/thérapie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs artificiels/immunologie , Anticorps à chaîne unique/immunologie , Lymphocytes T/immunologie , Animaux , Antigènes CD19/immunologie , Antigènes néoplasiques/immunologie , Lignée cellulaire tumorale , Femelle , Humains , Souris , Souris de lignée NOD , Tumeurs/immunologie , Culture de cellules primaires , Domaines protéiques , Récepteurs aux antigènes des cellules T/génétique , Récepteurs artificiels/génétique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie , Anticorps à chaîne unique/génétique , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
14.
J Virol ; 92(14)2018 07 15.
Article de Anglais | MEDLINE | ID: mdl-29720517

RÉSUMÉ

Today's gold standard in HIV therapy is combined antiretroviral therapy (cART). It requires strict adherence by patients and lifelong medication, which can lower the viral load below detection limits and prevent HIV-associated immunodeficiency but cannot cure patients. The bispecific T cell-engaging (BiTE) antibody technology has demonstrated long-term relapse-free outcomes in patients with relapsed and refractory acute lymphocytic leukemia. Here, we generated BiTE antibody constructs that target the HIV-1 envelope protein gp120 (HIV gp120) using either the scFv B12 or VRC01, the first two extracellular domains (1 + 2) of human CD4 alone or joined to the single chain variable fragment (scFv) of the antibody 17b fused to an anti-human CD3ε scFv. These engineered human BiTE antibody constructs showed engagement of T cells for redirected lysis of HIV gp120-transfected CHO cells. Furthermore, they substantially inhibited HIV-1 replication in peripheral blood mononuclear cells (PBMCs) as well as in macrophages cocultured with autologous CD8+ T cells, the most potent being the human CD4(1 + 2) BiTE [termed CD(1 + 2) h BiTE] antibody construct and the CD4(1 + 2)L17b BiTE antibody construct. The CD4(1 + 2) h BiTE antibody construct promoted HIV infection of human CD4-/CD8+ T cells. In contrast, the neutralizing B12 and the VRC01 BiTE antibody constructs, as well as the CD4(1 + 2)L17b BiTE antibody construct, did not. Thus, BiTE antibody constructs targeting HIV gp120 are very promising for constraining HIV and warrant further development as novel antiviral therapy with curative potential.IMPORTANCE HIV is a chronic infection well controlled with the current cART. However, we lack a cure for HIV, and the HIV pandemic goes on. Here, we showed in vitro and ex vivo that a BiTE antibody construct targeting HIV gp120 resulted in substantially reduced HIV replication. In addition, these BiTE antibody constructs display efficient killing of gp120-expressing cells and inhibited replication in ex vivo HIV-infected PBMCs or macrophages. We believe that BiTE antibody constructs recognizing HIV gp120 could be a very valuable strategy for a cure of HIV in combination with cART and compounds which reverse latency.


Sujet(s)
Anticorps bispécifiques/usage thérapeutique , Antiviraux/usage thérapeutique , Protéine d'enveloppe gp120 du VIH/immunologie , Infections à VIH/traitement médicamenteux , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Lymphocytes T/immunologie , Animaux , Anticorps bispécifiques/immunologie , Cellules CHO , Cricetinae , Cricetulus , Anticorps anti-VIH/immunologie , Infections à VIH/immunologie , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Humains , Immunothérapie , Agranulocytes/immunologie , Agranulocytes/virologie , Liaison aux protéines , Réplication virale/effets des médicaments et des substances chimiques , Réplication virale/immunologie
15.
Curr Clin Pharmacol ; 13(1): 55-64, 2018.
Article de Anglais | MEDLINE | ID: mdl-29773068

RÉSUMÉ

BACKGROUND: Blinatumomab is a bispecific T-cell engager (BiTE®) antibody construct targeting CD3ε on T cells and CD19 on B cells. We describe the relationship between pharmacokinetics (PK) of blinatumomab and pharmacodynamic (PD) changes in peripheral lymphocytes, serum cytokines, and tumor size in patients with non-Hodgkin lymphoma (NHL). METHODS: In a phase 1 study, 76 patients with relapsed/refractory NHL received blinatumomab by continuous intravenous infusion at various doses (0.5 to 90 µg/m2/day). PD changes were analyzed with respect to dose, blinatumomab concentration at steady state (Css), and cumulative area under the concentration-versus-time curve (AUCcum). RESULTS: B-cell depletion occurred within 48 hours at doses ≥5 µg/m2/day, followed first-order kinetics, and was blinatumomab exposure-dependent. Change in tumor size depended on systemic blinatumomab exposure and treatment duration and could be fitted to an Emax model, which predicted a 50% reduction in tumor size at AUCcum of ≥1,340 h×µg/L and Css of ≥1,830 pg/mL, corresponding to a blinatumomab dose of 47 µg/m2/day for 28 days. The magnitude of transient cytokine elevation, observed within 1-2 days of infusion start, was dose-dependent, with less pronounced elevation at low starting doses. CONCLUSION: B-lymphocyte depletion following blinatumomab infusion was exposure-dependent. Transient cytokine elevation increased with dose; it was less pronounced at low starting doses. Tumor response was a function of exposure, suggesting utility for the PK/PD relationship in dose selection for future studies, including NHL and other malignant settings.


Sujet(s)
Anticorps bispécifiques/administration et posologie , Antinéoplasiques/administration et posologie , Lymphocytes B/métabolisme , Lymphome malin non hodgkinien/traitement médicamenteux , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps bispécifiques/pharmacocinétique , Anticorps bispécifiques/pharmacologie , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/pharmacologie , Aire sous la courbe , Cytokines/sang , Relation dose-effet des médicaments , Femelle , Humains , Perfusions veineuses , Lymphome malin non hodgkinien/anatomopathologie , Mâle , Adulte d'âge moyen , Résultat thérapeutique , Jeune adulte
16.
PLoS One ; 12(8): e0183390, 2017.
Article de Anglais | MEDLINE | ID: mdl-28837681

RÉSUMÉ

For targets that are homogenously expressed, such as CD19 on cells of the B lymphocyte lineage, immunotherapies can be highly effective. Targeting CD19 with blinatumomab, a CD19/CD3 bispecific antibody construct (BiTE®), or with chimeric antigen receptor T cells (CAR-T) has shown great promise for treating certain CD19-positive hematological malignancies. In contrast, solid tumors with heterogeneous expression of the tumor-associated antigen (TAA) may present a challenge for targeted therapies. To prevent escape of TAA-negative cancer cells, immunotherapies with a local bystander effect would be beneficial. As a model to investigate BiTE®-mediated bystander killing in the solid tumor setting, we used epidermal growth factor receptor (EGFR) as a target. We measured lysis of EGFR-negative populations in vitro and in vivo when co-cultured with EGFR-positive cells, human T cells and an EGFR/CD3 BiTE® antibody construct. Bystander EGFR-negative cells were efficiently lysed by BiTE®-activated T cells only when proximal to EGFR-positive cells. Our mechanistic analysis suggests that cytokines released by BiTE®-activated T-cells induced upregulation of ICAM-1 and FAS on EGFR-negative bystander cells, contributing to T cell-induced bystander cell lysis.


Sujet(s)
Anticorps bispécifiques/immunologie , Effet bystander , Tumeurs/anatomopathologie , Lymphocytes T/immunologie , Animaux , Techniques de coculture , Cytokines/métabolisme , Cytotoxicité immunologique , Récepteurs ErbB/métabolisme , Femelle , Hétérogreffes , Humains , Activation des lymphocytes , Souris , Souris nude
17.
Exp Hematol Oncol ; 6: 14, 2017.
Article de Anglais | MEDLINE | ID: mdl-28533941

RÉSUMÉ

BACKGROUND: Blinatumomab has shown a remission rate of 69% in an exploratory single-arm, phase II dose-escalation study in adult patients with relapsed/refractory B-precursor acute lymphoblastic leukemia (ALL). We evaluated changes in laboratory parameters and immunopharmacodynamic markers in patients who received blinatumomab in the exploratory phase II study. METHODS: Data from 36 adults with relapsed/refractory ALL receiving blinatumomab as 4-week continuous IV infusions in various dose cohorts were analyzed for changes in liver enzymes, first-dose parameters, peripheral blood cell subpopulations, and cytokine/granzyme B release. Associations with clinical response were evaluated. RESULTS: Liver enzymes and inflammatory parameters transiently increased primarily during the first treatment week without clinical symptoms and reversed to baseline levels thereafter. B and T cells showed expected depletion and redistribution kinetics, respectively. Similarly, thrombocytes and T cells displayed an initial decline in cell counts, whereas neutrophils peaked during the first days after infusion start. T-cell redistribution coincided with upregulation of LFA-1 and CD69. Patients who responded to blinatumomab had more pronounced T-cell expansion, which was associated with proliferation of CD4+ and CD8+ T cells and memory subsets. Release of cytokines and granzyme B primarily occurred during the first week of cycle 1, except for IL-10, which was released in subsequent cycles. Blinatumomab step-dosing was associated with lower cytokine release and lower body temperature. CONCLUSIONS: In this study of relapsed/refractory ALL, blinatumomab-induced changes in laboratory parameters were transient and reversible. The evaluated PD markers demonstrated blinatumomab activity, and the analysis of cytokines supported the rationale for stepwise dosing. (ClinicalTrials.gov Identifier NCT01209286.).

18.
Antiviral Res ; 141: 155-164, 2017 05.
Article de Anglais | MEDLINE | ID: mdl-28257797

RÉSUMÉ

The ectodomain of the influenza A matrix protein 2 (M2e) is highly conserved amongst all influenza virus A subtypes. M2e is present on the surface of influenza A virus-infected cells, and therefore a suitable target for broadly protective therapies. We designed bispecific T cell engaging (BiTE®) antibody constructs specific for M2e by genetically fusing a single chain variable fragment (scFv) derived from an M2e-specific murine monoclonal antibody with a CD3ɛ-specific scFv. These so-called FLU BiTE® antibody constructs selectively mediate T cell dependent lysis of M2-expressing and influenza A virus infected cells and protect BALB/c mice against challenge with different influenza A virus subtypes. By humanizing the M2e-binding scFv, we generated human-like FLU BiTE® antibody constructs, with increased in vitro cytotoxic activity and in vivo protective capacity against influenza A virus infection. FLU BiTE® antibody constructs represent a promising new curative and prophylactic treatment option for influenza disease.


Sujet(s)
Anticorps bispécifiques/immunologie , Virus de la grippe A/composition chimique , Virus de la grippe A/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Lymphocytes T/immunologie , Protéines de la matrice virale/immunologie , Animaux , Anticorps bispécifiques/administration et posologie , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/immunologie , Anticorps antiviraux/sang , Tests de cytotoxicité immunologique , Mémoire immunologique , Vaccins antigrippaux/administration et posologie , Souris
19.
J Clin Oncol ; 34(10): 1104-11, 2016 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-26884582

RÉSUMÉ

PURPOSE: Blinatumomab is a CD19/CD3 BiTE (bispecific T-cell engager) antibody construct for the treatment of Philadelphia chromosome-negative acute B-lymphoblastic leukemia. We evaluated blinatumomab in relapsed/refractory B-cell non-Hodgkin lymphoma (NHL). PATIENTS AND METHODS: This 3 + 3 design, phase I dose-escalation study determined adverse events and the maximum tolerated dose (MTD) of continuous intravenous infusion blinatumomab in patients with relapsed/refractory NHL. Blinatumomab was administered over 4 or 8 weeks at seven different dose levels (0.5 to 90 µg/m(2)/day). End points were incidence of adverse events, pharmacokinetics, pharmacodynamics, and overall response rate. RESULTS: Between 2004 and 2011, 76 heavily pretreated patients with relapsed/refractory NHL, who included 14 with diffuse large B-cell lymphoma, were enrolled; 42 received treatment in the formal dose-escalation phase. Neurologic events were dose limiting, and 60 µg/m(2)/day was established as the MTD. Thirty-four additional patients were recruited to evaluate antilymphoma activity and strategies for mitigating neurologic events at a prespecified MTD. Stepwise dosing (5 to 60 µg/m(2)/day) plus pentosan polysulfate SP54 (n = 3) resulted in no treatment discontinuations; single-step (n = 5) and double-step (n = 24) dosing entailed two and seven treatment discontinuations due to neurologic events, respectively. Grade 3 neurologic events occurred in 22% of patients (no grade 4/5). Among patients treated at 60 µg/m(2)/day (target dose; n = 35), the overall response rate was 69% across NHL subtypes and 55% for diffuse large B-cell lymphoma (n = 11); median response duration was 404 days (95% CI, 207 to 1,129 days). CONCLUSION: In this phase I study of relapsed/refractory NHL, continuous infusion with CD19-targeted immunotherapy blinatumomab at various doses and schedules was feasible, with an MTD of 60 µg/m(2)/day. Single-agent blinatumomab showed antilymphoma activity.


Sujet(s)
Anticorps bispécifiques/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Immunothérapie/méthodes , Activation des lymphocytes/effets des médicaments et des substances chimiques , Lymphome malin non hodgkinien/traitement médicamenteux , Lymphome malin non hodgkinien/immunologie , Thérapie moléculaire ciblée/méthodes , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Adulte , Anticorps bispécifiques/administration et posologie , Anticorps bispécifiques/effets indésirables , Antigènes CD19/effets des médicaments et des substances chimiques , Antigènes CD19/immunologie , Antinéoplasiques/administration et posologie , Antinéoplasiques/effets indésirables , Antigènes CD3/effets des médicaments et des substances chimiques , Antigènes CD3/immunologie , Calendrier d'administration des médicaments , Femelle , Allemagne , Humains , Perfusions veineuses , Activation des lymphocytes/immunologie , Lymphome B/traitement médicamenteux , Lymphome B/immunologie , Lymphome folliculaire/traitement médicamenteux , Lymphome folliculaire/immunologie , Lymphome à cellules du manteau/traitement médicamenteux , Lymphome à cellules du manteau/immunologie , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Maladies du système nerveux/induit chimiquement , Récidive , Induction de rémission , Résultat thérapeutique
20.
PLoS One ; 10(10): e0141669, 2015.
Article de Anglais | MEDLINE | ID: mdl-26510188

RÉSUMÉ

BACKGROUND: Bispecific T cell engager (BiTE®) are single-chain bispecific antibody constructs with dual specificity for CD3 on T cells and a surface antigen on target cells. They can elicit a polyclonal cytotoxic T cell response that is not restricted by T cell receptor (TCR) specificity, and surface expression of MHC class I/peptide antigen complexes. Using human EpCAM/CD3-bispecific BiTE® antibody construct AMG 110, we here assessed to what extent surface expression of PD-L1, cytoplasmic expression of indoleamine-2,3-deoxygenase type 1, Bcl-2 and serpin PI-9, and the presence of transforming growth factor beta (TGF-ß), interleukin-10 (IL-10) and adenosine in culture medium can impact redirected lysis by AMG 110-engaged T cells. METHODS: The seven factors, which are all involved in inhibiting T cell functions by cancer cells, were tested with human EpCAM-expressing Chinese hamster ovary (CHO) target cells at levels that in most cases exceeded those observed in a number of human cancer cell lines. Co-culture experiments were used to determine the impact of the evasion mechanisms on EC50 values and amplitude of redirected lysis by AMG 110, and on BiTE®-induced proliferation of previously resting human peripheral T cells. FINDINGS: An inhibitory effect on redirected lysis by AMG 110-engaged T cells was seen upon overexpression of serpin PI-9, Bcl-2, TGF-ß and PD-L1. An inhibitory effect on induction of T cell proliferation was only seen with CHO cells overexpressing IDO. In no case, a single evasion mechanism rendered target cells completely resistant to BiTE®-induced lysis, and even various combinations could not. CONCLUSIONS: Our data suggest that diverse mechanisms employed by cancer cells to fend off T cells cannot inactivate AMG 110-engaged T cells, and that inhibitory effects observed in vitro may be overcome by increased concentrations of the BiTE® antibody construct.


Sujet(s)
Anticorps bispécifiques/immunologie , Antigènes néoplasiques/immunologie , Antigènes CD3/immunologie , Molécules d'adhérence cellulaire/immunologie , Échappement immunitaire , Tumeurs/immunologie , Lymphocytes T/immunologie , Animaux , Anticorps bispécifiques/génétique , Antigène CD274/génétique , Antigène CD274/métabolisme , Cellules CHO , Lignée cellulaire tumorale , Cricetulus , Cytotoxicité immunologique/génétique , Cytotoxicité immunologique/immunologie , Molécule d'adhérence des cellules épithéliales , Expression des gènes , Vecteurs génétiques/génétique , Humains , Échappement immunitaire/génétique , Activation des lymphocytes/génétique , Activation des lymphocytes/immunologie , Tumeurs/génétique , Tumeurs/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Serpines/génétique , Serpines/métabolisme , Lymphocytes T/métabolisme , Transfection , Facteur de croissance transformant bêta/génétique , Facteur de croissance transformant bêta/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE