Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(31): e2403002121, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39047033

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) represents a challenge in oncology, with limited treatment options for advanced-stage patients. Chimeric antigen receptor T cell (CAR T) therapy targeting mesothelin (MSLN) shows promise, but challenges such as the hostile immunosuppressive tumor microenvironment (TME) hinder its efficacy. This study explores the synergistic potential of combining proton radiation therapy (RT) with MSLN-targeting CAR T therapy in a syngeneic PDAC model. Proton RT significantly increased MSLN expression in tumor cells and caused a significant increase in CAR T cell infiltration into tumors. The combination therapy reshaped the immunosuppressive TME, promoting antitumorigenic M1 polarized macrophages and reducing myeloid-derived suppressor cells (MDSC). In a flank PDAC model, the combination therapy demonstrated superior attenuation of tumor growth and improved survival compared to individual treatments alone. In an orthotopic PDAC model treated with image-guided proton RT, tumor growth was significantly reduced in the combination group compared to the RT treatment alone. Further, the combination therapy induced an abscopal effect in a dual-flank tumor model, with increased serum interferon-γ levels and enhanced proliferation of extratumoral CAR T cells. In conclusion, combining proton RT with MSLN-targeting CAR T therapy proves effective in modulating the TME, enhancing CAR T cell trafficking, and exerting systemic antitumor effects. Thus, this combinatorial approach could present a promising strategy for improving outcomes in unresectable PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , GPI-Linked Proteins , Immunotherapy, Adoptive , Mesothelin , Pancreatic Neoplasms , Receptors, Chimeric Antigen , Tumor Microenvironment , Animals , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/radiotherapy , Pancreatic Neoplasms/pathology , Mice , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Immunotherapy, Adoptive/methods , Tumor Microenvironment/immunology , Humans , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/radiotherapy , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Proton Therapy/methods , Combined Modality Therapy , T-Lymphocytes/immunology , Female
2.
J Biochem Mol Toxicol ; 37(10): e23467, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37466109

ABSTRACT

Multidrug resistance (MDR) causes difficulties in the treatment of infections and cancer. Research and development studies have become increasingly important for the strategy of preventing MDR. There is a need for new multitarget drug research and advancement to reduce the development of drug resistance in drug-drug interactions and reduce cost and toxic effects. This study aimed to determine the effects of multi-target triazene compounds on antibacterial, antifungal, antiviral, cytotoxic, and larvicidal activities were investigated in vitro. A series of 12 novel of 1,3-diaryltriazene-substituted sulfadiazine (SDZ) derivatives were synthesized, and the obtained pure products characterized in detail by spectroscopic and analytic methods (FT-IR, 1 H-NMR, 13 C-NMR, and melting points). The antibacterial and antifungal activities of these derivatives (AH1-12) were determined by broth microdilution method. All derivatives have been evaluated in cell-based assays for cytotoxic and antiviral activities against Modified Vaccinia Virus Ankara. The larvicidal efficacy of these chemical compounds was also investigated by using Lucilia sericata (L. sericata) larvae. Twelve 1,3-diaryltriazene-substituted SDZ derivatives (AH1-12) were designed and developed as potent multitargeted compounds. Among them, the AH1 derivative showed the most antibacterial and antifungal activity. Besides, synthesized derivatives AH2, AH3, AH5, and AH7 showed higher antiviral activity than SDZ. All synthesized derivatives showed higher cytotoxic activity than SDZ. Also, they showed larvicidal activity at 72 h of the experiment. As a result, these compounds might be great leads for the development of next-generation multitargeted agents.


Subject(s)
Antineoplastic Agents , Sulfadiazine , Antifungal Agents/pharmacology , Triazenes/chemistry , Triazenes/pharmacology , Spectroscopy, Fourier Transform Infrared , Antineoplastic Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Antiviral Agents/pharmacology , Microbial Sensitivity Tests , Structure-Activity Relationship
3.
J Biochem Mol Toxicol ; 37(8): e23375, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37129082

ABSTRACT

Schiff bases (imines or azomethines) are versatile ligands synthesized from the condensation of amino compounds with active carbonyl groups and used for many pharmaceutical and medicinal applications. In our study, we aimed to determine the cytotoxic, antifungal and larvicidal activities of biologically potent bis-sulfonamide Schiff base derivatives that were re-synthesized by us. For this aim, 16 compounds were re-synthesized and tested for their cytotoxic, antifungal and larvicidal properties. Among this series, compounds A1B2, A1B4, A4B2, A4B3, and A4B4 were shown to have cytotoxic activity against tested cancer lung cell line (A549). The most potent antifungal activity was observed in compounds A2B1 and A2B2 against all fungi. A1B1 showed the strongest larvicidal effect at all concentrations at the 72nd h (100% mortality). These obtained results demonstrate that these type of bis-substituted compounds might be used as biologically potent pharmacophores against different types of diseases.


Subject(s)
Antifungal Agents , Schiff Bases , Antifungal Agents/pharmacology , Schiff Bases/pharmacology , Fungi , Sulfanilamide , Cell Line , Microbial Sensitivity Tests
4.
Int Ophthalmol ; 43(4): 1249-1259, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36255613

ABSTRACT

PURPOSE: Demodex folliculorum and Demodex brevis are common ectoparasites on skin that also can lead to blepharitis and chalazion. The aim of our study is to determine the prevalence of Demodex spp. in eyelashes of patients diagnosed with chronic blepharitis and chalazion. METHODS: This study included 330 patients diagnosed with chronic blepharitis, 70 patients diagnosed with chalazion and 130 volunteers without any ocular problems. Patient eyelashes were examined under a light microscope at magnifications of × 40, × 100 and × 400. Demodex spp. were determined. RESULTS: Parasite prevalence was significantly higher in blepharitis (75.5%) and chalazion groups (70%) compared to the control group (16.2%) (p < 0.001). The prevalence of D. folliculorum in the blepharitis group and D. brevis in the chalazion group was found to be significantly higher compared to other groups (p < 0.05). The average number of mites per eyelash was found to be significantly higher in patients with Demodex positive blepharitis (p = 0.001) and in chalazion patients (p = 0.047) than in the control group. It has been determined that mite positivity increases with age in blepharitis and control groups (p < 0.05). In the group with blepharitis, it was found that mite positivity was significant in the presence of symptoms (p = 0.0001) and Demodex positivity decreased as the education level of individuals increased (p = 0.039). CONCLUSION: The results of the study show that Demodex spp infestations should be considered in chronic blepharitis and chalazion.


Subject(s)
Ascomycota , Blepharitis , Chalazion , Eye Infections, Parasitic , Eyelashes , Mite Infestations , Mites , Animals , Humans , Chalazion/epidemiology , Mite Infestations/epidemiology , Mite Infestations/parasitology , Prevalence , Blepharitis/epidemiology , Blepharitis/parasitology , Eyelashes/parasitology , Chronic Disease , Eye Infections, Parasitic/diagnosis , Eye Infections, Parasitic/epidemiology
5.
J Dtsch Dermatol Ges ; 19(3): 359-362, 2021 03.
Article in English | MEDLINE | ID: mdl-33591642

ABSTRACT

Whereas approximately half of metastatic melanoma patients benefit from combined immune checkpoint inhibition targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and Programmed cell death protein 1 (PD-1), for those who do not respond, further strategies and treatment options need to be developed. Thus, focus is turning to the use of chimeric antigen receptor (CAR) T cells, a novel therapy that has not yet achieved a major breakthrough in solid tumors despite the impressive response rates reported for their use in hematologic malignancies. In melanoma and other solid tumor entities, different problems still need to be addressed to improve this therapy, with mechanisms to counteract tumor escape being one of them. In this context, we could show the feasibility of combining two different transfection methods - lentiviral transduction and RNA-electroporation - for equipping the same T lymphocyte with two different tumor antigen-specific receptors. While further analysis is required to transfer this novel strategy from bench to bedside, appropriate target antigens that avoid on-target/off-tumor toxicities and additional optimization to increase CAR T cell power are also needed to maximize their potential use in dermatologic oncology.


Subject(s)
Melanoma , Neoplasms , Antigens, Neoplasm , Humans , Immunotherapy, Adoptive , Melanoma/therapy , T-Lymphocytes
6.
Exp Dermatol ; 29(11): 1039-1045, 2020 11.
Article in English | MEDLINE | ID: mdl-32627228

ABSTRACT

After the recent success and approvals of chimeric antigen receptor (CAR) T cells in haematological malignancies, its efficacy is currently evaluated in a broad spectrum of tumor entities including melanoma. However, severe and potentially life-threatening side effects like cytokine release syndrome, neurologic toxicities, and the competing risk of morbidity and mortality from the treatment itself are still a major limiting factor in the current CAR T-cell landscape. In addition, especially in solid tumors, the lack of ideal target antigens to avoid on-target/off-tumor toxicities also restricts its use. While various groups are working on strategies to boost CAR T-cell efficacy, mechanisms to increase engineered T-cell safety should not move out of focus. Thus, the aim of this article is to summarize and to discuss current and potential future strategies and mechanisms to increase CAR T-cell safety in order to enable the wide use of this promising approach in melanoma and other tumor entities.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy, Adoptive/adverse effects , Melanoma/immunology , Melanoma/therapy , Skin Neoplasms/immunology , Skin Neoplasms/therapy , T-Lymphocytes/immunology , Transfection/methods , Humans
7.
Cytotherapy ; 21(11): 1166-1178, 2019 11.
Article in English | MEDLINE | ID: mdl-31668486

ABSTRACT

BACKGROUND: Although dendritic cell (DC)-based cancer vaccines represent a promising treatment strategy, its exploration in the clinic is hampered due to the need for Good Manufacturing Practice (GMP) facilities and associated trained staff for the generation of large numbers of DCs. The Quantum bioreactor system offered by Terumo BCT represents a hollow-fiber platform integrating GMP-compliant manufacturing steps in a closed system for automated cultivation of cellular products. In the respective established protocols, the hollow fibers are coated with fibronectin and trypsin is used to harvest the final cell product, which in the case of DCs allows processing of only one tenth of an apheresis product. MATERIALS AND RESULTS: We successfully developed a new protocol that circumvents the need for fibronectin coating and trypsin digestion, and makes the Quantum bioreactor system now suitable for generating large numbers of mature human monocyte-derived DCs (Mo-DCs) by processing a complete apheresis product at once. To achieve that, it needed a step-by-step optimization of DC-differentiation, e.g., the varying of media exchange rates and cytokine concentration until the total yield (% of input CD14+ monocytes), as well as the phenotype and functionality of mature Mo-DCs, became equivalent to those generated by our established standard production of Mo-DCs in cell culture bags. CONCLUSIONS: By using this new protocol for the Food and Drug Administration-approved Quantum system, it is now possible for the first time to process one complete apheresis to automatically generate large numbers of human Mo-DCs, making it much more feasible to exploit the potential of individualized DC-based immunotherapy.


Subject(s)
Bioreactors , Blood Component Removal , Cancer Vaccines , Cell Culture Techniques , Dendritic Cells/cytology , Dendritic Cells/physiology , Monocytes/physiology , Automation, Laboratory/standards , Bioreactors/standards , Blood Component Removal/instrumentation , Blood Component Removal/methods , Blood Component Removal/standards , Cancer Vaccines/standards , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Differentiation , Drug Industry/instrumentation , Drug Industry/standards , Guideline Adherence , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Leukapheresis/instrumentation , Leukapheresis/methods , Leukapheresis/standards , Manufactured Materials/standards , Monocytes/cytology
8.
Acta Derm Venereol ; 99(10): 889-893, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31141157

ABSTRACT

Mycobacterium chelonae is a rapidly growing non-tuberculous mycobacterium, which causes infections of the human skin and soft tissue. Despite an increasing incidence of such infections, patients are often misdiagnosed. We report here 5 patients with cutaneous and/or soft tissue infection due to M. chelonae who were diagnosed and treated at our centre. Two of the 5 patients were on immunosuppressive treatment. While clinical presentations differed in each patient, all had a long history of skin lesions. In addition to careful history-taking, tissue biopsies were obtained for mycobacterial culture and histopathological examination. Culture-directed antibiotic therapy was initiated, which resulted in a slow, but continuous, healing of the lesions. In summary, M. chelonae infections are still relatively rare, but should be considered in both immunocompromised and immunocompetent patients with prolonged skin lesions resistant to standard antibiotic treatment. For diagnosis, tissue analysis for mycobacterial culture and histopathological examination, and once diagnosed, adequate antibiotic treatment, is needed.


Subject(s)
Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium chelonae/isolation & purification , Opportunistic Infections/microbiology , Skin Diseases, Bacterial/microbiology , Soft Tissue Infections/microbiology , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/therapeutic use , Female , Humans , Immunocompromised Host , Immunosuppressive Agents/adverse effects , Male , Microbial Sensitivity Tests , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium chelonae/drug effects , Opportunistic Infections/diagnosis , Opportunistic Infections/drug therapy , Opportunistic Infections/immunology , Skin Diseases, Bacterial/diagnosis , Skin Diseases, Bacterial/drug therapy , Skin Diseases, Bacterial/immunology , Soft Tissue Infections/diagnosis , Soft Tissue Infections/drug therapy , Soft Tissue Infections/immunology , Treatment Outcome
9.
Exp Dermatol ; 27(12): 1315-1321, 2018 12.
Article in English | MEDLINE | ID: mdl-30288790

ABSTRACT

Chimeric antigen receptor (CAR)-T cells are one of the impressive recent success stories of anti-cancer immunotherapy. Especially in haematological malignancies, this treatment strategy has shown promising results leading to the recent approval of two CAR-T cell constructs targeting CD19 in the United States and the European Union. After the huge success in haematological cancers, the next step will be the evaluation of its efficacy in different solid tumors, which is currently investigated in preclinical as well as clinical settings. A commonly examined tumor model in the context of immunotherapy is melanoma, since it is known for its immunogenic features. However, the first results of CAR-T cell therapy in solid tumors did not reveal the same impressive outcomes that were observed in haematological malignancies, as engineered cells need to cope with several challenges. Obstacles include the lack of migration of CAR-T cells from blood vessels to the tumor site as well as the immunosuppressive tumor microenvironment within solid tumors. Another hurdle is posed by the identification of an ideal target antigen to avoid on-target/off-tumor toxicities. Regarding immune escape mechanisms, which can be developed by tumor cells to bypass immune recognition, the observation of antigen loss should also be considered. This article gives an overview of the challenges displayed in CAR-T cell therapy for the use in solid tumors and discusses different new strategies and approaches that deal with these problems in order to improve CAR-T cell therapy, particularly for its use in melanoma.


Subject(s)
Immunotherapy, Adoptive/trends , Melanoma/therapy , Receptors, Chimeric Antigen/therapeutic use , Skin Neoplasms/therapy , Antigens, CD19/chemistry , Antigens, Neoplasm/chemistry , Humans , Immune System , Immunosuppression Therapy , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/immunology , Treatment Outcome , Tumor Microenvironment/immunology
10.
Exp Dermatol ; 27(7): 769-778, 2018 07.
Article in English | MEDLINE | ID: mdl-29704887

ABSTRACT

Chimeric antigen receptor (CAR)-T cells have been used successfully for cancer immunotherapy. While substantial tumor regression was observed in leukaemia and lymphoma, CAR therapy of solid tumors needs further improvement. A major obstacle to the efficiency of engineered T cells is posed by triggering of inhibitory receptors, for example programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4), leading to an impaired antitumor activity. To boost CAR-T-cell function, we co-electroporated T cells with both, mRNA encoding a CAR specific for chondroitin sulphate proteoglycan 4 (CSPG4) and small-interfering RNAs (siRNAs) to downregulate PD-1 (siPD-1) and CTLA-4 (siCTLA-4). Flow cytometry revealed that activation-induced upregulation of both PD-1 and CTLA-4 was suppressed when compared to CAR-T cells electroporated with negative control siRNA. The siRNA transfection showed no influence on CAR expression of engineered T cells. Functionality assays were performed using PD-L1- and CD80-transfected melanoma cells endogenously expressing CSPG4. CAR-T cells transfected with siPD-1 alone showed improvement in cytokine secretion. Additionally, CAR-T cells transfected with either siPD-1 alone or together with siCTLA-4 exhibited a significantly increased cytotoxicity. No or only little effects were observed when CAR-T cells were co-transfected with siCTLA-4 only. Taken together, it is feasible to optimize CAR-T cells by co-transfection of CAR-encoding mRNA and siRNAs to downregulate inhibitory receptors. Our in vitro data indicate an improvement of the functionality of these CAR-T cells, suggesting that this strategy could represent a novel method to enhance CAR-T-cell immunotherapy of cancer.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy, Adoptive/methods , Melanoma/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/therapy , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/genetics , Cytokines/metabolism , Cytotoxicity, Immunologic , Down-Regulation , Electroporation , Humans , Melanoma/genetics , Melanoma/immunology , Programmed Cell Death 1 Receptor/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Transfection
11.
Int J Mol Sci ; 19(8)2018 Aug 11.
Article in English | MEDLINE | ID: mdl-30103488

ABSTRACT

Natural killer T (NKT) cells represent a cell subpopulation that combines characteristics of natural killer (NK) cells and T cells. Through their endogenous T-cell receptors (TCRs), they reveal a pronounced intrinsic anti-tumor activity. Thus, a NKT cell transfected with a chimeric antigen receptor (CAR), which recognizes a tumor-specific surface antigen, could attack tumor cells antigen-specifically via the CAR and additionally through its endogenous TCR. NKT cells were isolated from peripheral blood mononuclear cells (PBMCs), expanded, and electroporated with mRNA encoding a chondroitin sulfate proteoglycan 4 (CSPG4)-specific CAR. The CAR expression on NKT cells and their in vitro functionality were analyzed. A transfection efficiency of more than 80% was achieved. Upon stimulation with melanoma cells, CAR-NKT cells produced cytokines antigen-specifically. Compared with conventional CAR-T cells, cytokine secretion of CAR-NKT cells was generally lower. Specific cytotoxicity, however, was similar with CAR-NKT cells showing a trend towards improved cytotoxicity. Additionally, CAR-NKT cells could kill target cells through their endogenous TCRs. In summary, it is feasible to generate CAR-NKT cells by using mRNA electroporation. Their CAR-mediated cytotoxicity is at least equal to that of conventional CAR-T cells, while their intrinsic cytotoxic activity is maintained. Thus, CAR-NKT cells may represent a valuable alternative to conventional CAR-T cells for cancer immunotherapy.


Subject(s)
Immunotherapy/methods , Melanoma/therapy , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell , Humans , Jurkat Cells , Melanoma/genetics , Melanoma/immunology , Natural Killer T-Cells/pathology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
12.
BMC Cancer ; 17(1): 551, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28818060

ABSTRACT

BACKGROUND: Adoptive T-cell therapy relying on conventional T cells transduced with T-cell receptors (TCRs) or chimeric antigen receptors (CARs) has caused substantial tumor regression in several clinical trials. However, genetically engineered T cells have been associated with serious side-effects due to off-target toxicities and massive cytokine release. To obviate these concerns, we established a protocol adaptable to GMP to expand and transiently transfect γ/δ T cells with mRNA. METHODS: PBMC from healthy donors were stimulated using zoledronic-acid or OKT3 to expand γ/δ T cells and bulk T cells, respectively. Additionally, CD8+ T cells and γ/δ T cells were MACS-isolated from PBMC and expanded with OKT3. Next, these four populations were electroporated with RNA encoding a gp100/HLA-A2-specific TCR or a CAR specific for MCSP. Thereafter, receptor expression, antigen-specific cytokine secretion, specific cytotoxicity, and killing of the endogenous γ/δ T cell-target Daudi were analyzed. RESULTS: Using zoledronic-acid in average 6 million of γ/δ T cells with a purity of 85% were generated from one million PBMC. MACS-isolation and OKT3-mediated expansion of γ/δ T cells yielded approximately ten times less cells. OKT3-expanded and CD8+ MACS-isolated conventional T cells behaved correspondingly similar. All employed T cells were efficiently transfected with the TCR or the CAR. Upon respective stimulation, γ/δ T cells produced IFNγ and TNF, but little IL-2 and the zoledronic-acid expanded T cells exceeded MACS-γ/δ T cells in antigen-specific cytokine secretion. While the cytokine production of γ/δ T cells was in general lower than that of conventional T cells, specific cytotoxicity against melanoma cell lines was similar. In contrast to OKT3-expanded and MACS-CD8+ T cells, mock-electroporated γ/δ T cells also lysed tumor cells reflecting the γ/δ T cell-intrinsic anti-tumor activity. After transfection, γ/δ T cells were still able to kill MHC-deficient Daudi cells. CONCLUSION: We present a protocol adaptable to GMP for the expansion of γ/δ T cells and their subsequent RNA-transfection with tumor-specific TCRs or CARs. Given the transient receptor expression, the reduced cytokine release, and the equivalent cytotoxicity, these γ/δ T cells may represent a safer complementation to genetically engineered conventional T cells in the immunotherapy of melanoma (Exper Dermatol 26: 157, 2017, J Investig Dermatol 136: A173, 2016).


Subject(s)
RNA , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Adult , Cell Culture Techniques , Cytokines/metabolism , Cytotoxicity, Immunologic , Electroporation , Genetic Engineering , HLA-A2 Antigen/immunology , Healthy Volunteers , Humans , Immunomagnetic Separation , Immunophenotyping , Immunotherapy, Adoptive , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Melanoma/therapy , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Cell Antigen Receptor Specificity , Transfection , Young Adult , gp100 Melanoma Antigen/immunology
13.
Exp Dermatol ; 25(11): 872-879, 2016 11.
Article in English | MEDLINE | ID: mdl-27246630

ABSTRACT

The adoptive transfer of engineered T cells represents an important approach in immunotherapy of melanoma. However, relapse of the tumor can occur due to immune-escape mechanisms developed by the tumor cells, for example antigen loss, downregulation of the major histocompatibility complex presentation machinery and defects in antigen processing. To counteract these mechanisms, we combined a T-cell receptor and a chimeric antigen receptor, specific for different common melanoma antigens, gp100 (PMEL) and MCSP (HMW-MAA), to generate functional CD8+ T cells expressing two additional receptors (TETARs) by electroporation of receptor-encoding mRNA. These TETARs produced cytokines and were lytic upon recognition of each of their cognate antigens, while no reciprocal inhibition of the receptors occurred. When stimulated with target cells, which express both antigens, an enhanced effect was suggested. The confirmation that chimeric antigen receptors and T-cell receptors can be functionally combined opens up new avenues in cancer immunotherapy, and the generation of TETARs helps by-passing major mechanisms by which tumor cells escape immune recognition.


Subject(s)
Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes/transplantation , Immunotherapy, Adoptive/methods , Melanoma/therapy , Receptors, Antigen, T-Cell/genetics , gp100 Melanoma Antigen/genetics , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Healthy Volunteers , Humans , Receptors, Antigen, T-Cell/metabolism , Transfection , gp100 Melanoma Antigen/metabolism
14.
15.
Ann Surg Oncol ; 22(12): 4073-80, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25712801

ABSTRACT

BACKGROUND: Sentinel lymph node biopsy (SLNB) plays an important role in the prognostic classification of melanoma and is now a standard staging procedure. However, due to the complex drainage pattern and the risk of site associated morbidity, the potential survival benefit of SLNB is controversial in head and neck (H&N) melanoma. METHODS: Patients with primary H&N melanoma with a tumor thickness ≥1.00 mm diagnosed in the Department of Dermatology, University of Tuebingen, Germany between 1991 and 2010 were included in this study. Regarding patterns of metastases, disease-free, and overall-survival, 259 patients with SLNB were compared retrospectively to 218 patients without SLNB. RESULTS: The detection of micrometastasis in SLN proved to be a significant prognostic factor in H&N patients [hazard ratio (HR) 3.69, p < 0.0001]. A significant improvement of recurrence-free survival (RFS, p = 0.011), regional lymph node metastasis-free survival (LFS, p = 0.007), and distant metastasis-free survival (DMSF, p = 0.015) was observed for patients with SLNB versus non-SLNB. Furthermore, a trend towards better overall survival (OS) was found (p = 0.053) for the SLNB group. CONCLUSIONS: SLNB improved prognostic outcome in H&N melanoma in terms of disease-free and distant metastases survival, reduced subsequent regional lymph node metastases, and showed a trend towards a better OS.


Subject(s)
Facial Neoplasms/pathology , Lymph Nodes/pathology , Melanoma/secondary , Neoplasm Recurrence, Local/pathology , Scalp , Sentinel Lymph Node Biopsy , Skin Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Disease-Free Survival , Dissection , Facial Neoplasms/surgery , Female , Humans , Lymph Nodes/surgery , Lymphatic Metastasis , Male , Melanoma/surgery , Middle Aged , Neck , Neoplasm Micrometastasis/pathology , Retrospective Studies , Skin Neoplasms/surgery , Survival Rate , Young Adult
17.
Cancer Cell ; 42(8): 1319-1325, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39059390

ABSTRACT

Chimeric antigen receptor (CAR) T cells are effectively used in certain hematological malignancies, though tumor relapse and limited success in solid tumors persist. Recent efforts focus on developing combination treatments to enhance outcomes and safety. Here, we provide a comprehensive overview of such combinatorial approaches and a consideration of ongoing clinical trials.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Neoplasms/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Combined Modality Therapy , Clinical Trials as Topic , Receptors, Antigen, T-Cell/immunology , Animals
18.
Nat Commun ; 15(1): 3933, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730243

ABSTRACT

As a strategy to improve the therapeutic success of chimeric antigen receptor T cells (CART) directed against solid tumors, we here test the combinatorial use of CART and IMSA101, a newly developed stimulator of interferon genes (STING) agonist. In two syngeneic tumor models, improved overall survival is observed when mice are treated with intratumorally administered IMSA101 in addition to intravenous CART infusion. Transcriptomic analyses of CART isolated from tumors show elevated T cell activation, as well as upregulated cytokine pathway signatures, in particular IL-18, in the combination treatment group. Also, higher levels of IL-18 in serum and tumor are detected with IMSA101 treatment. Consistent with this, the use of IL-18 receptor negative CART impair anti-tumor responses in mice receiving combination treatment. In summary, we find that IMSA101 enhances CART function which is facilitated through STING agonist-induced IL-18 secretion.


Subject(s)
Interleukin-18 , Membrane Proteins , Receptors, Chimeric Antigen , Animals , Interleukin-18/metabolism , Membrane Proteins/agonists , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Humans , Cell Line, Tumor , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Lymphocyte Activation/drug effects , Immunotherapy, Adoptive/methods , Female , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy
20.
Cancer Immunol Res ; 11(7): 865, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37347991

ABSTRACT

In this issue, Tsimberidou and colleagues report the results of a first-in-human clinical trial using a personalized, multi-target, endogenous T-cell therapy in patients with metastatic solid tumors. This, and results of other recently published clinical trials, confirms the rationale of multi-target approaches that can increase tumor responses and counteract tumor heterogeneity and mechanisms of immune evasion. See related article by Tsimberidou et al., p. 925 (4).


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Humans , Feasibility Studies , Neoplasms/therapy , Neoplasms/immunology , Cell- and Tissue-Based Therapy , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL