1
|
Hirigoyen U, Guilbaud C, Krejbich M, Fouet M, Fresquet J, Arnaud B, Com E, Pineau C, Cadiou G, Burlaud-Gaillard J, Erbs P, Fradin D, Labarrière N, Fonteneau JF, Petithomme T, Boisgerault N. Oncolytic viruses alter the biogenesis of tumor extracellular vesicles and influence their immunogenicity. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200887. [PMID: 39492948 PMCID: PMC11530755 DOI: 10.1016/j.omton.2024.200887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 01/19/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024]
Abstract
Extracellular vesicles (EVs) are mediators of intercellular communication in the tumor microenvironment. Tumor EVs are commonly associated with metastasis, immunosuppression or drug resistance. Viral infections usually increase EV secretion, but little is known about the effect of oncolytic viruses (OVs) on tumor EVs. Here, we investigated the impact of oncolytic vesicular stomatitis virus (VSV) and vaccinia virus on EVs secreted by human melanoma and thoracic cancer cells. We found that OV infection increases the production of EVs by tumor cells. These EVs contain proteins of viral origin, such as VSV-G, thus creating a continuum of particles sharing markers of both canonical EVs and viruses. As such, the presence of VSV-G on EVs improves the transfer of their protein content to cell types commonly found in the tumor microenvironment. A proteomic analysis also revealed that EVs-OV secreted during VSV infection are enriched in immunity-related proteins. Finally, CD8+ T cells incubated with EVs-OV from infected cells display slightly enhanced cytotoxic functions. Taken together, these data suggest that OVs enhance the communication mediated by tumor EVs, which could participate in the therapeutic efficacy of OVs. These results also provide rationale for engineering OVs to exploit EVs and disseminate therapeutic proteins within the tumor microenvironment.
Collapse
Affiliation(s)
- Ugo Hirigoyen
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, 44000 Nantes, France
- LabEx IGO, Nantes Université, 44000 Nantes, France
| | - Coraly Guilbaud
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, 44000 Nantes, France
- LabEx IGO, Nantes Université, 44000 Nantes, France
| | - Morgane Krejbich
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, 44000 Nantes, France
- LabEx IGO, Nantes Université, 44000 Nantes, France
| | - Morgane Fouet
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, 44000 Nantes, France
- LabEx IGO, Nantes Université, 44000 Nantes, France
| | - Judith Fresquet
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, 44000 Nantes, France
- LabEx IGO, Nantes Université, 44000 Nantes, France
| | - Bastien Arnaud
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) – UMR_S 1085, 35000 Rennes, France
- University Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim core facility, 35000 Rennes, France
| | - Emmanuelle Com
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) – UMR_S 1085, 35000 Rennes, France
- University Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim core facility, 35000 Rennes, France
| | - Charles Pineau
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) – UMR_S 1085, 35000 Rennes, France
- University Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim core facility, 35000 Rennes, France
| | - Gwenann Cadiou
- LabEx IGO, Nantes Université, 44000 Nantes, France
- Nantes Université, Inserm UMR 1302, CNRS EMR 6001, Université d’Angers, INCIT, 44000 Nantes, France
| | - Julien Burlaud-Gaillard
- Plate-Forme IBiSA de Microscopie Electronique, Université de Tours and CHRU de Tours, 37000 Tours, France
| | | | - Delphine Fradin
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, 44000 Nantes, France
- LabEx IGO, Nantes Université, 44000 Nantes, France
| | - Nathalie Labarrière
- LabEx IGO, Nantes Université, 44000 Nantes, France
- Nantes Université, Inserm UMR 1302, CNRS EMR 6001, Université d’Angers, INCIT, 44000 Nantes, France
| | - Jean-François Fonteneau
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, 44000 Nantes, France
- LabEx IGO, Nantes Université, 44000 Nantes, France
| | - Tacien Petithomme
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, 44000 Nantes, France
- LabEx IGO, Nantes Université, 44000 Nantes, France
- Nantes Université, CHU Nantes, 44000 Nantes, France
| | - Nicolas Boisgerault
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, 44000 Nantes, France
- LabEx IGO, Nantes Université, 44000 Nantes, France
| |
Collapse
|
2
|
Autin P, Deshayes S, Lea J, Boisgerault N, Dupré E, Labarrière N, Leguevel R, Fonteneau JF, Blanquart C, Fradin D. The DCMU Herbicide Shapes T-cell Functions By Modulating Micro-RNA Expression Profiles. Front Immunol 2022; 13:925241. [PMID: 35967413 PMCID: PMC9366666 DOI: 10.3389/fimmu.2022.925241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
DCMU [N-(3,4-dichlorophenyl)-N-dimethylurea] or diuron is a widely used herbicide, which can cause adverse effects on human, especially on immune cells, due to their intrinsic properties and wide distribution. These cells are important for fighting not only against virus or bacteria but also against neoplastic cell development. We developed an approach that combines functional studies and miRNA and RNA sequencing data to evaluate the effects of DCMU on the human immune response against cancer, particularly the one carried out by CD8+ T cells. We found that DCMU modulates the expression of miRNA in a dose-dependent manner, leading to a specific pattern of gene expression and consequently to a diminished cytokine and granzyme B secretions. Using mimics or anti-miRs, we identified several miRNA, such as hsa-miR-3135b and hsa-miR-21-5p, that regulate these secretions. All these changes reduce the CD8+ T cells’ cytotoxic activity directed against cancer cells, in vitro and in vivo in a zebrafish model. To conclude, our study suggests that DCMU reduces T-cell abilities, participating thus to the establishment of an environment conducive to cancer development.
Collapse
Affiliation(s)
- Pierre Autin
- Nantes Université, INSERM UMR1307, CNRS UMR6075, Université d’Angers, CRCI2NA, Nantes, France
| | - Sophie Deshayes
- Nantes Université, INSERM UMR1307, CNRS UMR6075, Université d’Angers, CRCI2NA, Nantes, France
| | - Juliette Lea
- Université de Rennes, ImPACcell Plateform, BIOSIT, Rennes, France
| | - Nicolas Boisgerault
- Nantes Université, INSERM UMR1307, CNRS UMR6075, Université d’Angers, CRCI2NA, Nantes, France
| | - Emilie Dupré
- Nantes Université, Univ Angers, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, Nantes, France
| | - Nathalie Labarrière
- Nantes Université, Univ Angers, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, Nantes, France
| | - Rémy Leguevel
- Université de Rennes, ImPACcell Plateform, BIOSIT, Rennes, France
| | - Jean-François Fonteneau
- Nantes Université, INSERM UMR1307, CNRS UMR6075, Université d’Angers, CRCI2NA, Nantes, France
| | - Christophe Blanquart
- Nantes Université, INSERM UMR1307, CNRS UMR6075, Université d’Angers, CRCI2NA, Nantes, France
| | - Delphine Fradin
- Nantes Université, INSERM UMR1307, CNRS UMR6075, Université d’Angers, CRCI2NA, Nantes, France
- *Correspondence: Delphine Fradin,
| |
Collapse
|
3
|
Dehmani S, Nerrière-Daguin V, Néel M, Elain-Duret N, Heslan JM, Belarif L, Mary C, Thepenier V, Biteau K, Poirier N, Blancho G, Haspot F. SIRPγ-CD47 Interaction Positively Regulates the Activation of Human T Cells in Situation of Chronic Stimulation. Front Immunol 2021; 12:732530. [PMID: 34925315 PMCID: PMC8671138 DOI: 10.3389/fimmu.2021.732530] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/05/2021] [Indexed: 11/16/2022] Open
Abstract
A numerous number of positive and negative signals via various molecules modulate T-cell activation. Within the various transmembrane proteins, SIRPγ is of interest since it is not expressed in rodents. SIRPγ interaction with CD47 is reevaluated in this study. Indeed, we show that the anti-SIRPγ mAb clone LSB2.20 previously used by others has not been appropriately characterized. We reveal that the anti-SIRPα clone KWAR23 is a Pan anti-SIRP mAb which efficiently blocks SIRPα and SIRPγ interactions with CD47. We show that SIRPγ expression on T cells varies with their differentiation and while being expressed on Tregs, is not implicated in their suppressive functions. SIRPγ spatial reorganization at the immune synapse is independent of its interaction with CD47. In vitro SIRPα-γ/CD47 blockade with KWAR23 impairs IFN-γ secretion by chronically activated T cells. In vivo in a xeno-GvHD model in NSG mice, the SIRPγ/CD47 blockade with the KWAR23 significantly delays the onset of the xeno-GvHD and deeply impairs human chimerism. In conclusion, we have shown that T-cell interaction with CD47 is of importance notably in chronic stimulation.
Collapse
Affiliation(s)
- Safa Dehmani
- OSE Immunotherapeutics, Nantes, France.,Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Urologie-Néphrologie (ITUN), Nantes, France
| | - Véronique Nerrière-Daguin
- Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Urologie-Néphrologie (ITUN), Nantes, France
| | - Mélanie Néel
- Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Urologie-Néphrologie (ITUN), Nantes, France
| | - Nathan Elain-Duret
- Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Urologie-Néphrologie (ITUN), Nantes, France
| | - Jean-Marie Heslan
- Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Urologie-Néphrologie (ITUN), Nantes, France
| | | | | | | | | | | | - Gilles Blancho
- Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Urologie-Néphrologie (ITUN), Nantes, France
| | - Fabienne Haspot
- Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Urologie-Néphrologie (ITUN), Nantes, France
| |
Collapse
|
4
|
Hamieh M, Chatillon JF, Dupel E, Bayeux F, Fauquembergue E, Maby P, Drouet A, Duval-Modeste AB, Adriouch S, Boyer O, Latouche JB. Generation of Pure Highly Functional Human Anti-Tumor Specific Cytotoxic T Lymphocytes With Stem Cell-Like Memory Features for Melanoma Immunotherapy. Front Immunol 2021; 12:674276. [PMID: 34566953 PMCID: PMC8456028 DOI: 10.3389/fimmu.2021.674276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/13/2021] [Indexed: 02/03/2023] Open
Abstract
Adoptive immunotherapy based on the transfer of anti-tumor cytotoxic T lymphocytes (CTLs) is a promising strategy to cure cancers. However, rapid expansion of numerous highly functional CTLs with long-lived features remains a challenge. Here, we constructed NIH/3T3 mouse fibroblast-based artificial antigen presenting cells (AAPCs) and precisely evaluated their ability to circumvent this difficulty. These AAPCs stably express the essential molecules involved in CTL activation in the HLA-A*0201 context and an immunogenic HLA-A*0201 restricted analogue peptide derived from MART-1, an auto-antigen overexpressed in melanoma. Using these AAPCs and pentamer-based magnetic bead-sorting, we defined, in a preclinical setting, the optimal conditions to expand pure MART-1-specific CTLs. Numerous highly purified MART-1-specific CTLs were rapidly obtained from healthy donors and melanoma patients. Both TCR repertoire and CDR3 sequence analyses revealed that MART-1-specific CTL responses were similar to those reported in the literature and obtained with autologous or allogeneic presenting cells. These MART-1-specific CTLs were highly cytotoxic against HLA-A*0201+ MART-1+ tumor cells. Moreover, they harbored a suitable phenotype for immunotherapy, with effector memory, central memory and, most importantly, stem cell-like memory T cell features. Notably, the cells harboring stem cell-like memory phenotype features were capable of self-renewal and of differentiation into potent effector anti-tumor T cells. These "off-the-shelf" AAPCs represent a unique tool to rapidly and easily expand large numbers of long-lived highly functional pure specific CTLs with stem cell-like memory T cell properties, for the development of efficient adoptive immunotherapy strategies against cancers.
Collapse
Affiliation(s)
- Mohamad Hamieh
- Normandie University, UNIROUEN, Inserm U1245, Institute for Research and Innovation in Biomedecine (IRIB), Rouen, France
| | - Jean-François Chatillon
- Normandie University, UNIROUEN, Inserm U1234 - Pathophysiology, Autoimmunity, Neuromuscular diseases and regenerative THERapies (PANTHER), IRIB, Rouen, France
| | - Estelle Dupel
- Normandie University, UNIROUEN, Inserm U1245, Institute for Research and Innovation in Biomedecine (IRIB), Rouen, France
| | - Florence Bayeux
- Normandie University, UNIROUEN, Inserm U1234 - Pathophysiology, Autoimmunity, Neuromuscular diseases and regenerative THERapies (PANTHER), IRIB, Rouen, France
| | - Emilie Fauquembergue
- Normandie University, UNIROUEN, Inserm U1245, Institute for Research and Innovation in Biomedecine (IRIB), Rouen, France
| | - Pauline Maby
- Normandie University, UNIROUEN, Inserm U1245, Institute for Research and Innovation in Biomedecine (IRIB), Rouen, France
| | - Aurelie Drouet
- Normandie University, UNIROUEN, Inserm U1245, Institute for Research and Innovation in Biomedecine (IRIB), Rouen, France
| | | | - Sahil Adriouch
- Normandie University, UNIROUEN, Inserm U1234 - Pathophysiology, Autoimmunity, Neuromuscular diseases and regenerative THERapies (PANTHER), IRIB, Rouen, France
| | - Olivier Boyer
- Normandie University, UNIROUEN, Inserm U1234 - Pathophysiology, Autoimmunity, Neuromuscular diseases and regenerative THERapies (PANTHER), IRIB, Rouen, France.,Department of Immunology and Biotherapy, Rouen University Hospital, Rouen, France
| | - Jean-Baptiste Latouche
- Normandie University, UNIROUEN, Inserm U1245, Institute for Research and Innovation in Biomedecine (IRIB), Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| |
Collapse
|
5
|
Marotte L, Simon S, Vignard V, Dupre E, Gantier M, Cruard J, Alberge JB, Hussong M, Deleine C, Heslan JM, Shaffer J, Beauvais T, Gaschet J, Scotet E, Fradin D, Jarry A, Nguyen T, Labarriere N. Increased antitumor efficacy of PD-1-deficient melanoma-specific human lymphocytes. J Immunother Cancer 2021; 8:jitc-2019-000311. [PMID: 32001504 PMCID: PMC7057432 DOI: 10.1136/jitc-2019-000311] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2020] [Indexed: 01/08/2023] Open
Abstract
Background Genome editing offers unique perspectives for optimizing the functional properties of T cells for adoptive cell transfer purposes. So far, PDCD1 editing has been successfully tested mainly in chimeric antigen receptor T (CAR-T) cells and human primary T cells. Nonetheless, for patients with solid tumors, the adoptive transfer of effector memory T cells specific for tumor antigens remains a relevant option, and the use of high avidity T cells deficient for programmed cell death-1 (PD-1) expression is susceptible to improve the therapeutic benefit of these treatments. Methods Here we used the transfection of CAS9/sgRNA ribonucleoproteic complexes to edit PDCD1 gene in human effector memory CD8+ T cells specific for the melanoma antigen Melan-A. We cloned edited T cell populations and validated PDCD1 editing through sequencing and cytometry in each T cell clone, together with T-cell receptor (TCR) chain’s sequencing. We also performed whole transcriptomic analyses on wild-type (WT) and edited T cell clones. Finally, we documented in vitro and in vivo through adoptive transfer in NOD scid gamma (NSG) mice, the antitumor properties of WT and PD-1KO T cell clones, expressing the same TCR. Results Here we demonstrated the feasibility to edit PDCD1 gene in human effector memory melanoma-specific T lymphocytes. We showed that PD-1 expression was dramatically reduced or totally absent on PDCD1-edited T cell clones. Extensive characterization of a panel of T cell clones expressing the same TCR and exhibiting similar functional avidity demonstrated superior antitumor reactivity against a PD-L1 expressing melanoma cell line. Transcriptomic analysis revealed a downregulation of genes involved in proliferation and DNA replication in PD-1-deficient T cell clones, whereas genes involved in metabolism and cell signaling were upregulated. Finally, we documented the superior ability of PD-1-deficient T cells to significantly delay the growth of a PD-L1 expressing human melanoma tumor in an NSG mouse model. Conclusion The use of such lymphocytes for adoptive cell transfer purposes, associated with other approaches modulating the tumor microenvironment, would be a promising alternative to improve immunotherapy efficacy in solid tumors.
Collapse
Affiliation(s)
- Lucine Marotte
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Sylvain Simon
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Virginie Vignard
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Emilie Dupre
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Malika Gantier
- LabEx IGO, Université de Nantes, Nantes, France.,Université de Nantes, Inserm, CRTI, F-44000 Nantes, France
| | - Jonathan Cruard
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | | | - Melanie Hussong
- NGS Assay Research & Development, Qiagen Sciences, Frederick, Maryland, United States
| | - Cecile Deleine
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Jean-Marie Heslan
- LabEx IGO, Université de Nantes, Nantes, France.,Université de Nantes, Inserm, CRTI, F-44000 Nantes, France
| | - Jonathan Shaffer
- NGS Assay Research & Development, Qiagen Sciences, Frederick, Maryland, United States
| | - Tiffany Beauvais
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Joelle Gaschet
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Emmanuel Scotet
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Delphine Fradin
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Anne Jarry
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France.,LabEx IGO, Université de Nantes, Nantes, France
| | - Tuan Nguyen
- Université de Nantes, Inserm, CRTI, F-44000 Nantes, France
| | - Nathalie Labarriere
- Université de Nantes, Inserm, CRCINA, F-44000 Nantes, France .,LabEx IGO, Université de Nantes, Nantes, France
| |
Collapse
|
6
|
Dréno B, Khammari A, Fortun A, Vignard V, Saiagh S, Beauvais T, Jouand N, Bercegay S, Simon S, Lang F, Labarrière N. Phase I/II clinical trial of adoptive cell transfer of sorted specific T cells for metastatic melanoma patients. Cancer Immunol Immunother 2021; 70:3015-3030. [PMID: 34120214 PMCID: PMC8423703 DOI: 10.1007/s00262-021-02961-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Adoptive cell transfer (ACT) of tumor-specific T lymphocytes represents a relevant therapeutic strategy to treat metastatic melanoma patients. Ideal T-cells should combine tumor specificity and reactivity with survival in vivo, while avoiding autoimmune side effects. Here we report results from a Phase I/II clinical trial (NCT02424916, performed between 2015 and 2018) in which 6 metastatic HLA-A2 melanoma patients received autologous antigen-specific T-cells produced from PBMC, after peptide stimulation in vitro, followed by sorting with HLA-peptide multimers and amplification. Each patient received a combination of Melan-A and MELOE-1 polyclonal specific T-cells, whose specificity and anti-tumor reactivity were checked prior to injection, with subcutaneous IL-2. Transferred T-cells were also characterized in terms of functional avidity, diversity and phenotype and their blood persistence was evaluated. An increase in specific T-cells was detected in the blood of all patients at day 1 and progressively disappeared from day 7 onwards. No serious adverse events occurred after this ACT. Clinically, five patients progressed and one patient experienced a partial response following therapy. Melan-A and MELOE-1 specific T-cells infused to this patient were diverse, of high avidity, with a high proportion of T lymphocytes co-expressing PD-1 and TIGIT but few other exhaustion markers. In conclusion, we demonstrated the feasibility and safety of ACT with multimer-sorted Melan-A and MELOE-1 specific T cells to metastatic melanoma patients. The clinical efficacy of such therapeutic strategy could be further enhanced by the selection of highly reactive T-cells, based on PD-1 and TIGIT co-expression, and a combination with ICI, such as anti-PD-1.
Collapse
Affiliation(s)
- Brigitte Dréno
- Dermato-Cancerology Department, CIC 1413, CHU Nantes, Nantes, France.,UTCG, CHU Nantes, Nantes, France.,CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes, France
| | - Amir Khammari
- Dermato-Cancerology Department, CIC 1413, CHU Nantes, Nantes, France.,CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes, France
| | - Agnès Fortun
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Virginie Vignard
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes, France
| | | | - Tiffany Beauvais
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes, France
| | - Nicolas Jouand
- LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,SFR Santé, CNRS, Inserm, Inserm UMS 016, CNRS UMS 3556, Université de Nantes, CHU Nantes, 44000, Nantes, France
| | | | - Sylvain Simon
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - François Lang
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France. .,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.
| | - Nathalie Labarrière
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France. .,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France. .,SFR Santé, CNRS, Inserm, Inserm UMS 016, CNRS UMS 3556, Université de Nantes, CHU Nantes, 44000, Nantes, France.
| |
Collapse
|
7
|
Stringhini M, Spadafora I, Catalano M, Mock J, Probst P, Spörri R, Neri D. Cancer therapy in mice using a pure population of CD8 + T cell specific to the AH1 tumor rejection antigen. Cancer Immunol Immunother 2021; 70:3183-3197. [PMID: 33796916 PMCID: PMC8505334 DOI: 10.1007/s00262-021-02912-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/08/2021] [Indexed: 11/25/2022]
Abstract
There is a growing interest in the use of patient-derived T cells for the treatment of various types of malignancies. The expansion of a polyclonal and polyspecific population of tumor-reactive T cells, with a subsequent infusion into the same donor patient, has been implemented, sometimes with positive results. It is not known, however, whether a set of T cells with a single antigen specificity may be sufficient for an effective therapy. To gain more insights in this matter, we used naturally occurring T cells recognizing a retroviral peptide (AH1), which is endogenous in many tumor cell lines of BALB/c origin and which serves as potent tumor rejection antigen. We were able to isolate and expand this rare population of T cells to numbers suitable for therapy experiments in mice (i.e., up to 30 × 106 cells/mouse). After the expansion process, T cells efficiently killed antigen-positive tumor cells in vitro and demonstrated tumor growth inhibition in two syngeneic murine models of cancer. However, AH1-specific T cells failed to induce complete regressions of established tumors. The incomplete activity was associated with a failure of injected T cells to survive in vivo, as only a very limited amount of T cells was found in tumor or secondary lymphoid organs 72 h after injection. These data suggest that future therapeutic strategies based on autologous T cells may require the potentiation of tumor-homing and survival properties of cancer-specific T cells.
Collapse
Affiliation(s)
- Marco Stringhini
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Ilaria Spadafora
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Marco Catalano
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Jacqueline Mock
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Philipp Probst
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Roman Spörri
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland.
| |
Collapse
|
8
|
Gauttier V, Pengam S, Durand J, Biteau K, Mary C, Morello A, Néel M, Porto G, Teppaz G, Thepenier V, Danger R, Vince N, Wilhelm E, Girault I, Abes R, Ruiz C, Trilleaud C, Ralph K, Trombetta ES, Garcia A, Vignard V, Martinet B, Glémain A, Bruneau S, Haspot F, Dehmani S, Duplouye P, Miyasaka M, Labarrière N, Laplaud D, Le Bas-Bernardet S, Blanquart C, Catros V, Gouraud PA, Archambeaud I, Aublé H, Metairie S, Mosnier JF, Costantini D, Blancho G, Conchon S, Vanhove B, Poirier N. Selective SIRPα blockade reverses tumor T cell exclusion and overcomes cancer immunotherapy resistance. J Clin Invest 2021; 130:6109-6123. [PMID: 33074246 DOI: 10.1172/jci135528] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
T cell exclusion causes resistance to cancer immunotherapies via immune checkpoint blockade (ICB). Myeloid cells contribute to resistance by expressing signal regulatory protein-α (SIRPα), an inhibitory membrane receptor that interacts with ubiquitous receptor CD47 to control macrophage phagocytosis in the tumor microenvironment. Although CD47/SIRPα-targeting drugs have been assessed in preclinical models, the therapeutic benefit of selectively blocking SIRPα, and not SIRPγ/CD47, in humans remains unknown. We report a potent synergy between selective SIRPα blockade and ICB in increasing memory T cell responses and reverting exclusion in syngeneic and orthotopic tumor models. Selective SIRPα blockade stimulated tumor nest T cell recruitment by restoring murine and human macrophage chemokine secretion and increased anti-tumor T cell responses by promoting tumor-antigen crosspresentation by dendritic cells. However, nonselective SIRPα/SIRPγ blockade targeting CD47 impaired human T cell activation, proliferation, and endothelial transmigration. Selective SIRPα inhibition opens an attractive avenue to overcoming ICB resistance in patients with elevated myeloid cell infiltration in solid tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mélanie Néel
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France.,CHU Nantes, Nantes, France
| | - Georgia Porto
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | | | | | - Richard Danger
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France.,CHU Nantes, Nantes, France
| | - Nicolas Vince
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | | | | | - Riad Abes
- OSE Immunotherapeutics, Nantes, France
| | | | - Charlène Trilleaud
- OSE Immunotherapeutics, Nantes, France.,Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | - Kerry Ralph
- Cancer Immunology & Immune Modulation, Boehringer Ingelheim, Ridgefield, Connecticut, USA
| | - E Sergio Trombetta
- Cancer Immunology & Immune Modulation, Boehringer Ingelheim, Ridgefield, Connecticut, USA
| | - Alexandra Garcia
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France.,CHU Nantes, Nantes, France
| | - Virginie Vignard
- CHU Nantes, Nantes, France.,Université de Nantes, CNRS, INSERM, Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), F-44000 Nantes, France
| | - Bernard Martinet
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | - Alexandre Glémain
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | - Sarah Bruneau
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | - Fabienne Haspot
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | - Safa Dehmani
- OSE Immunotherapeutics, Nantes, France.,Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | - Pierre Duplouye
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | - Masayuki Miyasaka
- Immunology Frontier Research Center, Osaka University, Yamada-oka, Suita, Japan
| | - Nathalie Labarrière
- Université de Nantes, CNRS, INSERM, Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), F-44000 Nantes, France
| | - David Laplaud
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France.,CHU Nantes, Nantes, France
| | - Stéphanie Le Bas-Bernardet
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | - Christophe Blanquart
- Université de Nantes, CNRS, INSERM, Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), F-44000 Nantes, France
| | - Véronique Catros
- Université de Rennes, INSERM, CHU Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer), UMR_S 1241, CRB Santé Rennes, Rennes, France
| | - Pierre-Antoine Gouraud
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | - Isabelle Archambeaud
- CHU Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif (IMAD), Service d'Hépato-Gastro-Entérologie et Chirurgie Digestive
| | - Hélène Aublé
- CHU Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif (IMAD), Service d'Hépato-Gastro-Entérologie et Chirurgie Digestive.,Centre d'investigation Clinique and
| | - Sylvie Metairie
- CHU Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif (IMAD), Service d'Hépato-Gastro-Entérologie et Chirurgie Digestive
| | - Jean-François Mosnier
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France.,Service d'Anatomie et Cytologie Pathologiques, CHU Nantes, Nantes, France
| | | | - Gilles Blancho
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France.,CHU Nantes, Nantes, France
| | - Sophie Conchon
- Université de Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064.,Institut de Transplantation Urologie Néphrologie (ITUN), F-44000 Nantes, France
| | | | | |
Collapse
|
9
|
Towards new horizons: characterization, classification and implications of the tumour antigenic repertoire. Nat Rev Clin Oncol 2020; 17:595-610. [PMID: 32572208 PMCID: PMC7306938 DOI: 10.1038/s41571-020-0387-x] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2020] [Indexed: 12/21/2022]
Abstract
Immune-checkpoint inhibition provides an unmatched level of durable clinical efficacy in various malignancies. Such therapies promote the activation of antigen-specific T cells, although the precise targets of these T cells remain unknown. Exploiting these targets holds great potential to amplify responses to treatment, such as by combining immune-checkpoint inhibition with therapeutic vaccination or other antigen-directed treatments. In this scenario, the pivotal hurdle remains the definition of valid HLA-restricted tumour antigens, which requires several levels of evidence before targets can be established with sufficient confidence. Suitable antigens might include tumour-specific antigens with alternative or wild-type sequences, tumour-associated antigens and cryptic antigens that exceed exome boundaries. Comprehensive antigen classification is required to enable future clinical development and the definition of innovative treatment strategies. Furthermore, clinical development remains challenging with regard to drug manufacturing and regulation, as well as treatment feasibility. Despite these challenges, treatments based on diligently curated antigens combined with a suitable therapeutic platform have the potential to enable optimal antitumour efficacy in patients, either as monotherapies or in combination with other established immunotherapies. In this Review, we summarize the current state-of-the-art approaches for the identification of candidate tumour antigens and provide a structured terminology based on their underlying characteristics. Immune-checkpoint inhibition has transformed the treatment of patients with advanced-stage cancers. Nonetheless, the specific antigens targeted by T cells that are activated or reactivated by these agents remain largely unknown. In this Review, the authors describe the characterization and classification of tumour antigens including descriptions of the most appropriate detection methods, and discuss potential regulatory issues regarding the use of tumour antigen-based therapeutics. Immune-checkpoint inhibition has profoundly changed the paradigm for the care of several malignancies. Although these therapies activate antigen-specific T cells, the precise mechanisms of action and their specific targets remain largely unknown. Anticancer immunotherapies encompass two fundamentally different therapeutic principles based on knowledge of their therapeutic targets, that either have been characterized (antigen-aware) or have remained elusive (antigen-unaware). HLA-presented tumour antigens of potential therapeutic relevance can comprise alternative or wild-type amino acid sequences and can be subdivided into different categories based on their mechanisms of formation. The available methods for the detection of HLA-presented antigens come with intrinsic challenges and limitations and, therefore, warrant multiple lines of evidence of robust tumour specificity before being considered for clinical use. Knowledge obtained using various antigen-detection strategies can be combined with different therapeutic platforms to create individualized therapies that hold great promise, including when combined with already established immunotherapies. Tailoring immunotherapies while taking into account the substantial heterogeneity of malignancies as well as that of HLA loci not only requires innovative science, but also demands innovative approaches to trial design and drug regulation.
Collapse
|
10
|
Tumor infiltrating lymphocytes as adjuvant treatment in stage III melanoma patients with only one invaded lymph node after complete resection: results from a multicentre, randomized clinical phase III trial. Cancer Immunol Immunother 2020; 69:1663-1672. [PMID: 32306076 DOI: 10.1007/s00262-020-02572-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 04/07/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Adoptive tumor-infiltrating lymphocytes (TIL) therapy and interleukin-2 (IL-2) have been investigated in melanoma. AIM To confirm previously observed preventive effects of TIL + IL2 in a subgroup of patients with relapsing metastatic stage III melanoma. METHODOLOGY Open-label, randomized two-group, multicenter five-year trial in adult stage III melanoma patients with only one invaded lymph node after complete resection. Patients received TIL + IL2 or abstention. TIL + IL2 was administered within 8 weeks after lymph node resection and 4 weeks after. Disease-free survival was assessed every 2 months up to month 18, every 3 months up to month 36 and every 4 months up to 5 years. A once-a-year follow-up was scheduled beyond the five-year follow-up. Safety was assessed throughout the trial. RESULTS Overall, 49 patients accounted for the modified intent-to-treat and 47 for the PP. Slightly more male than female patients participated; mean age was 57.7 ± 11.4 years in the TIL + IL2 group and 53.5 ± 13.0 years in the abstention group. After 5 years of follow-up, 11/26 patients in the TIL + IL2 group and 13/23 in the abstention group had relapsed. There was no statistical difference between the groups (HR: 0.63 CI 95% [0.28-1.41], p = 0.258), nine patients in the TIL + IL2 and 11 in the abstention group died with no significant difference between the two groups (HR: 0.65 CI95% [0.27 - 1.59], p = 0.34). Safety was good. CONCLUSION We did not confirm results of a previous trial. However, ulceration of the primary melanoma may be considered predictive of the efficacy of TIL in melanoma in adjuvant setting, in a manner similar to interferon α.
Collapse
|
11
|
Gerber HP, Sibener LV, Lee LJ, Gee MH. Identification of Antigenic Targets. Trends Cancer 2020; 6:299-318. [PMID: 32209445 DOI: 10.1016/j.trecan.2020.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022]
Abstract
The ideal cancer target antigen (Ag) is expressed at high copy numbers on neoplastic cells, absent on normal tissues, and contributes to the survival of cancer cells. Despite significant investments in the identification of cell surface Ags, there is a paucity of targets that meet such ideal cancer target criteria. Recent clinical trials in patients with cancer treated with immune checkpoint inhibitors (ICIs) indicate that cluster of differentiation (CD)8+ T cells, by means of their T cell receptors (TCRs) recognizing intracellular targets presented as peptides in the context of human leukocyte antigen (peptide-human leukocyte antigen complex; pHLA) molecules on tumor cells, can mediate deep and long-lasting antitumor responses in patients with solid tumors. Therefore, pHLA-target Ags may represent the long sought-after, ideal targets for solid tumor targeting by high-potency oncology compounds.
Collapse
Affiliation(s)
| | - Leah V Sibener
- 3T Biosciences, 1455 Adams Drive, Menlo Park, CA 94025, USA
| | - Luke J Lee
- 3T Biosciences, 1455 Adams Drive, Menlo Park, CA 94025, USA
| | - Marvin H Gee
- 3T Biosciences, 1455 Adams Drive, Menlo Park, CA 94025, USA
| |
Collapse
|
12
|
Benveniste PM, Nakatsugawa M, Nguyen L, Ohashi PS, Hirano N, Zúñiga-Pflücker JC. In vitro-generated MART-1-specific CD8 T cells display a broader T-cell receptor repertoire than ex vivo naïve and tumor-infiltrating lymphocytes. Immunol Cell Biol 2019; 97:427-434. [PMID: 30633397 DOI: 10.1111/imcb.12231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 12/07/2018] [Accepted: 12/09/2018] [Indexed: 12/01/2022]
Abstract
The differentiation of human hematopoietic stem cells into CD8 T cells can be achieved in vitro with the OP9-DL4 system. We considered that in the absence of medullary thymic epithelial cells, which serve to restrict the breath of the T-cell receptor (TCR) repertoire by expressing tissue-restricted antigens, a distinct repertoire would be generated in vitro. To test this notion, we compared the TCR-Vα/Vβ (TRAV/TRBV) gene usage of major histocompatibility complex-restricted antigen (MART-1)-specific T cells generated in vitro to that from ex vivo naïve T cells and tumor-infiltrating lymphocytes (TILs) using high-throughput DNA sequencing. In contrast to naïve T cells and TILs, which showed the expected narrow TRAV repertoire, in vitro-generated MART-1-specific T cells used almost all TRAV gene families and displayed unique CDR3 lengths. Our work demonstrates that the OP9-DL4 system supports the creation of a broad antigen-specific TCR repertoire, suggesting that T cells generated in vitro may undergo a different set of selection events that otherwise constrains the TCR repertoire of thymus-derived T cells.
Collapse
Affiliation(s)
| | | | - Linh Nguyen
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Pamela S Ohashi
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Naoto Hirano
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Simon S, Wu Z, Cruard J, Vignard V, Fortun A, Khammari A, Dreno B, Lang F, Rulli SJ, Labarriere N. TCR Analyses of Two Vast and Shared Melanoma Antigen-Specific T Cell Repertoires: Common and Specific Features. Front Immunol 2018; 9:1962. [PMID: 30214446 PMCID: PMC6125394 DOI: 10.3389/fimmu.2018.01962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022] Open
Abstract
Among Immunotherapeutic approaches for cancer treatment, the adoptive transfer of antigen specific T cells is still a relevant approach, that could have higher efficacy when further combined with immune check-point blockade. A high number of adoptive transfer trials have been performed in metastatic melanoma, due to its high immunogenic potential, either with polyclonal TIL or antigen-specific polyclonal populations. In this setting, the extensive characterization of T cell functions and receptor diversity of infused polyclonal T cells is required, notably for monitoring purposes. We developed a clinical grade procedure for the selection and amplification of polyclonal CD8 T cells, specific for two shared and widely expressed melanoma antigens: Melan-A and MELOE-1. This procedure is currently used in a clinical trial for HLA-A2 metastatic melanoma patients. In this study, we characterized the T-cell diversity (T-cell repertoire) of such T cell populations using a new RNAseq strategy. We first assessed the added-value of TCR receptor sequencing, in terms of sensitivity and specificity, by direct comparison with cytometry analysis of the T cell populations labeled with anti-Vß-specific antibodies. Results from these analyzes also confirmed specific features already reported for Melan-A and MELOE-1 specific T cell repertoires in terms of V-alpha recurrence usage, on a very high number of T cell clonotypes. Furthermore, these analyses also revealed undescribed features, such as the recurrence of a specific motif in the CDR3α region for MELOE-1 specific T cell repertoire. Finally, the analysis of a large number of T cell clonotypes originating from various patients revealed the existence of public CDR3α and ß clonotypes for Melan-A and MELOE-1 specific T cells. In conclusion, this method of high throughput TCR sequencing is a reliable and powerful approach to deeply characterize polyclonal T cell repertoires, and to reveal specific features of a given TCR repertoire, that would be useful for immune follow-up of cancer patients treated by immunotherapeutic approaches.
Collapse
Affiliation(s)
- Sylvain Simon
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - Zhong Wu
- Qiagen Sciences, Frederick, MD, United States
| | - J Cruard
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - Virginie Vignard
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France.,Centre Hospitalier Universitaire Nantes, Nantes, France
| | - Agnes Fortun
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - Amir Khammari
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France.,Department of Dermato-Cancerology of Nantes Hospital, Nantes, France
| | - Brigitte Dreno
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France.,Department of Dermato-Cancerology of Nantes Hospital, Nantes, France
| | - Francois Lang
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | | | - Nathalie Labarriere
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France.,Centre Hospitalier Universitaire Nantes, Nantes, France
| |
Collapse
|
14
|
Tsuji T, Yoneda A, Matsuzaki J, Miliotto A, Ryan C, Koya RC, Odunsi K. Rapid Construction of Antitumor T-cell Receptor Vectors from Frozen Tumors for Engineered T-cell Therapy. Cancer Immunol Res 2018; 6:594-604. [PMID: 29588318 DOI: 10.1158/2326-6066.cir-17-0434] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/26/2017] [Accepted: 03/12/2018] [Indexed: 12/31/2022]
Abstract
T cells genetically engineered with tumor antigen-specific T-cell receptor (TCR) genes have demonstrated therapeutic potential in patients with solid tumors. In order to achieve broader application, an efficient method to identify TCR genes for an array of tumor antigens and HLA restriction elements is required. Here, we have developed a method to construct a TCR-expression library from specimens, including frozen tumor biopsies, that contain antigen-specific T cells. TCR-expressing cassettes were constructed and cloned in a retroviral plasmid vector within 24 hours by unbiased PCR amplification of TCR α and β chain variable regions assembled with TCR constant regions. The method was validated by constructing TCR-expressing vectors from tumor antigen-specific T-cell clones and functionally assessing TCR gene-transduced T cells. We applied this method to frozen ovarian tumor specimens that were infiltrated by tumor antigen-specific T cells. The tumor-derived TCR libraries were expressed in peripheral T cells from healthy volunteers and screened for tumor antigen-specific TCR pairs with the use of an MHC/peptide tetramer reagent. Tumor antigen-specific TCR-expressing transgenes were recovered from isolated tetramer-positive T cells. Peripheral T cells that were engineered with library-derived TCR gene showed potent therapeutic antitumor effect in a tumor xenograft model. Our method can efficiently and rapidly provide tumor-specific TCR-expressing viral vectors for the manufacture of therapeutic and personalized antitumor T-cell products. Cancer Immunol Res; 6(5); 594-604. ©2018 AACR.
Collapse
Affiliation(s)
- Takemasa Tsuji
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York.,Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | - Akira Yoneda
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York
| | - Junko Matsuzaki
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York.,Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | - Anthony Miliotto
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York
| | - Courtney Ryan
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York
| | - Richard C Koya
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York.,Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York. .,Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York.,Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
15
|
Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes in Advanced Melanoma Patients. J Immunol Res 2018; 2018:3530148. [PMID: 29750176 PMCID: PMC5883986 DOI: 10.1155/2018/3530148] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 12/02/2022] Open
Abstract
Immunotherapy for melanoma includes adoptive cell therapy with autologous tumor-infiltrating lymphocytes (TILs). This monocenter retrospective study was undertaken to evaluate the efficacy and safety of this treatment of patients with advanced melanoma. All advanced melanoma patients treated with TILs using the same TIL expansion methodology and same treatment interleukin-2 (IL-2) regimen between 2009 and 2012 were included. After sterile intralesional excision of a cutaneous or subcutaneous metastasis, TILs were produced according to a previously described method and then infused into the patient who also received a complementary subcutaneous IL-2 regimen. Nine women and 1 man were treated for unresectable stage IIIC (n = 4) or IV (n = 6) melanoma. All but 1 patient with unresectable stage III melanoma (1st line) had received at least 2 previous treatments, including anti-CTLA-4 antibody for 4. The number of TILs infused ranged from 0.23 × 109 to 22.9 × 109. Regarding safety, no serious adverse effect was reported. Therapeutic responses included a complete remission, a partial remission, 2 stabilizations, and 6 progressions. Among these 4 patients with clinical benefit, 1 is still alive with 9 years of follow-up and 1 died from another cause after 8 years of follow-up. Notably, patients treated with high percentages of CD4 + CD25 + CD127lowFoxp3+ T cells among their TILs had significantly shorter OS. The therapeutic effect of combining TILs with new immunotherapies needs further investigation.
Collapse
|
16
|
Choi BK, Kim SH, Kim YH, Kwon BS. Cancer immunotherapy using tumor antigen-reactive T cells. Immunotherapy 2018; 10:235-245. [DOI: 10.2217/imt-2017-0130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Studies over the last 30 years have shown the promise of cancer immunotherapy using T cells. In particular, since the report by Rosenberg and colleagues in 2002 that adoptive T-cell therapy (ACT) under lymphopenic conditions substantially increased response rates in melanoma patients, ACT has become a promising immunotherapeutic route to cancer treatment. Here we provide a brief history of ACT and review the characteristics of T-cell therapeutics that are specific to this approach. Since every T-cell treatment has its own unique properties in terms of number and type of target antigens, and number of epitopes and type of T cells, we review the main strategies for designing ACT: how Ag specificity is determined, how is it standardized and the need for lymphodepletion to induce epitope spreading. We also briefly consider the next generation of ACT.
Collapse
Affiliation(s)
- Beom K. Choi
- Biomedicine Production Branch, National Cancer Center, Goyang, Korea
| | - Seon-Hee Kim
- Immunotherapeutics Branch, Division of Convergence Technology, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Korea 10408
| | - Young H. Kim
- Biomedicine Production Branch, National Cancer Center, Goyang, Korea
- Eutilex, Suite 1401 Daeryung Technotown 17, Gasan Digital 1-ro 25, Geumcheon-gu, Seoul, Korea 08594
| | - Byoung S. Kwon
- Eutilex, Suite 1401 Daeryung Technotown 17, Gasan Digital 1-ro 25, Geumcheon-gu, Seoul, Korea 08594
| |
Collapse
|
17
|
Simon S, Vignard V, Varey E, Parrot T, Knol AC, Khammari A, Gervois N, Lang F, Dreno B, Labarriere N. Emergence of High-Avidity Melan-A–Specific Clonotypes as a Reflection of Anti–PD-1 Clinical Efficacy. Cancer Res 2017; 77:7083-7093. [DOI: 10.1158/0008-5472.can-17-1856] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/29/2017] [Accepted: 09/19/2017] [Indexed: 11/16/2022]
Abstract
Abstract
Therapeutic strategies using anti–PD-1–blocking antibodies reported unparalleled effectiveness for melanoma immunotherapy, but deciphering immune responses modulated by anti–PD-1 treatment remains a crucial issue. Here, we analyzed the composition and functions of the large Melan-A–specific T-cell repertoire in the peripheral blood of 9 melanoma patients before and after 2 months of treatment with anti–PD-1. We observed amplification of Melan-A–specific Vß subfamilies undetectable before therapy (thereafter called emerging Vß subfamilies) in responding patients, with a predominant expansion in patients with a complete response. These emerging Vß subfamilies displayed a higher functional avidity for their cognate antigen than Vß subfamilies not amplified upon anti–PD-1 therapy and could be identified by a sustained coexpression of PD-1 and TIGIT receptors. Thus, in addition to the emergence of neoantigen-specific T cells previously documented upon anti–PD-1 therapy, our work describes the emergence of high-avidity Melan-A–specific clonotypes as a surrogate marker of treatment efficacy. Cancer Res; 77(24); 7083–93. ©2017 AACR.
Collapse
Affiliation(s)
- Sylvain Simon
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
| | - Virginie Vignard
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 3CHU Nantes, Nantes, France
| | - Emilie Varey
- 4Department of Dermato-cancerology of Nantes Hospital, Nantes, France
| | - Tiphaine Parrot
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
| | - Anne-Chantal Knol
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 4Department of Dermato-cancerology of Nantes Hospital, Nantes, France
| | - Amir Khammari
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 4Department of Dermato-cancerology of Nantes Hospital, Nantes, France
| | - Nadine Gervois
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
| | - Francois Lang
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
| | - Brigitte Dreno
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 4Department of Dermato-cancerology of Nantes Hospital, Nantes, France
| | - Nathalie Labarriere
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 3CHU Nantes, Nantes, France
| |
Collapse
|
18
|
Spontaneous regression of malignant melanoma - is it based on the interplay between host immune system and melanoma antigens? Anticancer Drugs 2017; 28:819-830. [DOI: 10.1097/cad.0000000000000526] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
19
|
Chen S, Gu F, Li K, Zhang K, Liu Y, Liang J, Gao W, Wu G, Liu L. A hybrid of B and T lymphoblastic cell line could potentially substitute dendritic cells to efficiently expand out Her-2/neu-specific cytotoxic T lymphocytes from advanced breast cancer patients in vitro. J Hematol Oncol 2017; 10:63. [PMID: 28245833 PMCID: PMC5331710 DOI: 10.1186/s13045-017-0429-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 02/23/2017] [Indexed: 01/01/2023] Open
Abstract
Adoptive transfer of cytotoxic T lymphocytes (CTLs) holds promises to cure cancer. However, this treatment is hindered by lacking a robust way to specifically expand out CTLs. Here, we developed a hybrid of B lymphoblastic cell line and T lymphoblastic cell line (T2 cells) as a substitute of dendritic cells, together with irradiated autologous peripheral blood mononuclear cell (PBMC) as feeder cells and rhIL-2, to activate and expand Her-2/neu-specific CD8+ T cells from human epidermal growth factor receptor 2 (Her-2/neu) and human leukocyte antigen (HLA)-A2 double positive advanced breast cancer patients in vitro. These Her-2/neu-loaded T2 cells reproducibly activated and expanded out Her-2/neu-specific CD8+ T cells to 107 in 8 weeks. Furthermore, these Her-2/neu-specific CD8+ T cells had good sensitivity of recognition and killing Her-2/neu-overexpressed breast cancer cell line SK.BR.3. This technique gives us another insight on how to rapidly obtain sufficient CTLs for adoptive cancer immunotherapy.
Collapse
Affiliation(s)
- Sheng Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Feifei Gu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kang Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kai Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yangyang Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinyan Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Gao
- Traumatology Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
20
|
Phase I Clinical Trial of 4-1BB-based Adoptive T-Cell Therapy for Epstein-Barr Virus (EBV)-positive Tumors. J Immunother 2016; 39:140-8. [PMID: 26938947 PMCID: PMC4834812 DOI: 10.1097/cji.0000000000000113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Although adoptive cell therapy using Ag-specific T cells has been tested successfully in the clinic, the production of these T cells has been challenging. By applying our simple and practical 4-1BB-based method for the generation of Ag-specific CD8 T cells, here we determined the maximum tolerated dose, toxicity profile, immunologic responses, and clinical efficacy of autologous Epstein-Barr virus (EBV)/LMP2A-specific CD8 T cells (EBV-induced Natural T cell; EBViNT) in patients with relapsed/refractory EBV-positive tumors. This was a single-center, phase I, dose-escalation trial study evaluating 4 escalating dosing schedules of single injected EBViNT. CD8 T-cell responses against different LMP2A peptides in each patient were determined, and the most effective peptides were used to produce EBViNT. The produced autologous EBViNTs were single infused to patients with EBV-associated malignancy who had failed to standard treatments and were of HLA-A02 or A24 type. Of 11 patients enrolled, 8 patients received a single infusion of EBViNT: 4 with nasopharyngeal carcinomas, 1 with Hodgkin lymphoma, 2 with extranodal NK/T lymphomas, and 1 with diffuse large B-cell lymphoma. Single infusion of EBViNT was well tolerated by all the patients and generated objective antitumor responses in 3 of them. EBViNT infusion induced 2 waves of interferon-γ response: 1 approximately 1 week and the other 4-8 weeks after the treatment. The strength of the second wave was related to the efficacy of the treatment. The current trial shows that EBViNT therapy is safe and may provide a new option for treating EBV-positive recurrent cancer patients resistant to conventional therapy.
Collapse
|
21
|
Dutoit V, Migliorini D, Dietrich PY, Walker PR. Immunotherapy of Malignant Tumors in the Brain: How Different from Other Sites? Front Oncol 2016; 6:256. [PMID: 28003994 PMCID: PMC5141244 DOI: 10.3389/fonc.2016.00256] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/24/2016] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy is now advancing at remarkable pace for tumors located in various tissues, including the brain. Strategies launched decades ago, such as tumor antigen-specific therapeutic vaccines and adoptive transfer of tumor-infiltrating lymphocytes are being complemented by molecular engineering approaches allowing the development of tumor-specific TCR transgenic and chimeric antigen receptor T cells. In addition, the spectacular results obtained in the last years with immune checkpoint inhibitors are transfiguring immunotherapy, these agents being used both as single molecules, but also in combination with other immunotherapeutic modalities. Implementation of these various strategies is ongoing for more and more malignancies, including tumors located in the brain, raising the question of the immunological particularities of this site. This may necessitate cautious selection of tumor antigens, minimizing the immunosuppressive environment and promoting efficient T cell trafficking to the tumor. Once these aspects are taken into account, we might efficiently design immunotherapy for patients suffering from tumors located in the brain, with beneficial clinical outcome.
Collapse
Affiliation(s)
- Valérie Dutoit
- Laboratory of Tumor Immunology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| | - Denis Migliorini
- Oncology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| | - Pierre-Yves Dietrich
- Oncology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| | - Paul R Walker
- Laboratory of Tumor Immunology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| |
Collapse
|
22
|
Santegoets SJAM, Welters MJP, van der Burg SH. Monitoring of the Immune Dysfunction in Cancer Patients. Vaccines (Basel) 2016; 4:vaccines4030029. [PMID: 27598210 PMCID: PMC5041023 DOI: 10.3390/vaccines4030029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/25/2016] [Accepted: 08/27/2016] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy shows promising clinical results in patients with different types of cancer, but its full potential is not reached due to immune dysfunction as a result of several suppressive mechanisms that play a role in cancer development and progression. Monitoring of immune dysfunction is a prerequisite for the development of strategies aiming to alleviate cancer-induced immune suppression. At this point, the level at which immune dysfunction occurs has to be established, the underlying mechanism(s) need to be known, as well as the techniques to assess this. While it is relatively easy to measure general signs of immune suppression, it turns out that accurate monitoring of the frequency and function of immune-suppressive cells is still difficult. A lack of truly specific markers, the phenotypic complexity among suppressive cells of the same lineage, but potentially with different functions and functional assays that may not cover every mechanistic aspect of immune suppression are among the reasons complicating proper assessments. Technical innovations in flow and mass cytometry will allow for more complete sets of markers to precisely determine phenotype and associated function. There is, however, a clear need for functional assays that recapitulate more of the mechanisms employed to suppress the immune system.
Collapse
Affiliation(s)
- Saskia J A M Santegoets
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Marij J P Welters
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
23
|
Spear TT, Nagato K, Nishimura MI. Strategies to genetically engineer T cells for cancer immunotherapy. Cancer Immunol Immunother 2016; 65:631-49. [PMID: 27138532 DOI: 10.1007/s00262-016-1842-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/25/2016] [Indexed: 12/15/2022]
Abstract
Immunotherapy is one of the most promising and innovative approaches to treat cancer, viral infections, and other immune-modulated diseases. Adoptive immunotherapy using gene-modified T cells is an exciting and rapidly evolving field. Exploiting knowledge of basic T cell biology and immune cell receptor function has fostered innovative approaches to modify immune cell function. Highly translatable clinical technologies have been developed to redirect T cell specificity by introducing designed receptors. The ability to engineer T cells to manifest desired phenotypes and functions is now a thrilling reality. In this review, we focus on outlining different varieties of genetically engineered T cells, their respective advantages and disadvantages as tools for immunotherapy, and their promise and drawbacks in the clinic.
Collapse
Affiliation(s)
- Timothy T Spear
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Loyola University Chicago, 2160 S. 1st Ave, Bldg 112, Room 308, Maywood, IL, 60153, USA.
| | - Kaoru Nagato
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Loyola University Chicago, 2160 S. 1st Ave, Bldg 112, Room 308, Maywood, IL, 60153, USA
- Department of General Thoracic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Michael I Nishimura
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Loyola University Chicago, 2160 S. 1st Ave, Bldg 112, Room 308, Maywood, IL, 60153, USA
| |
Collapse
|
24
|
Favorable alteration of tumor microenvironment by immunomodulatory cytokines for efficient T-cell therapy in solid tumors. PLoS One 2015; 10:e0131242. [PMID: 26107883 PMCID: PMC4479879 DOI: 10.1371/journal.pone.0131242] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/29/2015] [Indexed: 01/12/2023] Open
Abstract
Unfavorable ratios between the number and activation status of effector and suppressor immune cells infiltrating the tumor contribute to resistance of solid tumors to T-cell based therapies. Here, we studied the capacity of FDA and EMA approved recombinant cytokines to manipulate this balance in favor of efficient anti-tumor responses in B16.OVA melanoma bearing C57BL/6 mice. Intratumoral administration of IFN-α2, IFN-γ, TNF-α, and IL-2 significantly enhanced the anti-tumor effect of ovalbumin-specific CD8+ T-cell (OT-I) therapy, whereas GM-CSF increased tumor growth in association with an increase in immunosuppressive cell populations. None of the cytokines augmented tumor trafficking of OT-I cells significantly, but injections of IFN-α2, IFN-γ and IL-2 increased intratumoral cytokine secretion and recruitment of endogenous immune cells capable of stimulating T-cells, such as natural killer and maturated CD11c+ antigen-presenting cells. Moreover, IFN-α2 and IL-2 increased the levels of activated tumor-infiltrating CD8+ T-cells concomitant with reduction in the CD8+ T-cell expression of anergy markers CTLA-4 and PD-1. In conclusion, intratumoral administration of IFN-α2, IFN-γ and IL-2 can lead to immune sensitization of the established tumor, whereas GM-CSF may contribute to tumor-associated immunosuppression. The results described here provide rationale for including local administration of immunostimulatory cytokines into T-cell therapy regimens. One appealing embodiment of this would be vectored delivery which could be advantageous over direct injection of recombinant molecules with regard to efficacy, cost, persistence and convenience.
Collapse
|
25
|
Tähtinen S, Grönberg-Vähä-Koskela S, Lumen D, Merisalo-Soikkeli M, Siurala M, Airaksinen AJ, Vähä-Koskela M, Hemminki A. Adenovirus Improves the Efficacy of Adoptive T-cell Therapy by Recruiting Immune Cells to and Promoting Their Activity at the Tumor. Cancer Immunol Res 2015; 3:915-25. [PMID: 25977260 DOI: 10.1158/2326-6066.cir-14-0220-t] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 05/07/2015] [Indexed: 11/16/2022]
Abstract
Despite the rapid progress in the development of novel adoptive T-cell therapies, the clinical benefits in treatment of established tumors have remained modest. Several immune evasion mechanisms hinder T-cell entry into tumors and their activity within the tumor. Of note, oncolytic adenoviruses are intrinsically immunogenic due to inherent pathogen-associated molecular patterns. Here, we studied the capacity of adenovirus to overcome resistance of chicken ovalbumin-expressing B16.OVA murine melanoma tumors to adoptive ovalbumin-specific CD8(+) T-cell (OT-I) therapy. Following intraperitoneal transfer of polyclonally activated OT-I lymphocytes, control of tumor growth was superior in mice given intratumoral adenovirus compared with control mice, even in the absence of oncolytic virus replication. Preexisting antiviral immunity against serotype 5 did not hinder the therapeutic efficacy of the combination treatment. Intratumoral adenovirus injection was associated with an increase in proinflammatory cytokines, CD45(+) leukocytes, CD8(+) lymphocytes, and F4/80(+) macrophages, suggesting enhanced tumor immunogenicity. The proinflammatory effects of adenovirus on the tumor microenvironment led to expression of costimulatory signals on CD11c(+) antigen-presenting cells and subsequent activation of T cells, thus breaking the tumor-induced peripheral tolerance. An increased number of CD8(+) T cells specific for endogenous tumor antigens TRP-2 and gp100 was detected in combination-treated mice, indicating epitope spreading. Moreover, the majority of virus/T-cell-treated mice rejected the challenge of parental B16.F10 tumors, suggesting that systemic antitumor immunity was induced. In summary, we provide proof-of-mechanism data on combining adoptive T-cell therapy and adenovirotherapy for the treatment of cancer.
Collapse
Affiliation(s)
- Siri Tähtinen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Susanna Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Dave Lumen
- Laboratory of Radiochemistry, Department of Chemistry, University of Helsinki, Helsinki, Finland
| | - Maiju Merisalo-Soikkeli
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Mikko Siurala
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland. TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Anu J Airaksinen
- Laboratory of Radiochemistry, Department of Chemistry, University of Helsinki, Helsinki, Finland. Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland. TILT Biotherapeutics Ltd, Helsinki, Finland. Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
| |
Collapse
|
26
|
Abstract
Harnessing the immune system to recognize and destroy tumor cells has been the central goal of anti-cancer immunotherapy. In recent years, there has been an increased interest in optimizing this technology in order to make it a clinically feasible treatment. One of the main treatment modalities within cancer immunotherapy has been adoptive T cell therapy (ACT). Using this approach, tumor-specific cytotoxic T cells are infused into cancer patients with the goal of recognizing, targeting, and destroying tumor cells. In the current review, we revisit some of the major successes of ACT, the major hurdles that have been overcome to optimize ACT, the remaining challenges, and future approaches to make ACT widely available.
Collapse
Affiliation(s)
- Karlo Perica
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Juan Carlos Varela
- Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mathias Oelke
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan Schneck
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
27
|
Miamen AG, Gustafson MP, Roberts LR. Rethinking cancer immunotherapy: Using advanced cancer genetics in immune-mediated eradication of gastrointestinal cancers. Hepatology 2014; 60:2121-4. [PMID: 25220571 DOI: 10.1002/hep.27442] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Alexander G Miamen
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| | | | | |
Collapse
|
28
|
Chandran SS, Paria BC, Srivastava AK, Rothermel LD, Stephens DJ, Dudley ME, Somerville R, Wunderlich JR, Sherry RM, Yang JC, Rosenberg SA, Kammula US. Persistence of CTL clones targeting melanocyte differentiation antigens was insufficient to mediate significant melanoma regression in humans. Clin Cancer Res 2014; 21:534-43. [PMID: 25424856 DOI: 10.1158/1078-0432.ccr-14-2208] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Adoptive transfer of autologous tumor infiltrating lymphocytes (TIL) can mediate durable cancer regression in selected patients with metastatic melanoma. However, the tumor antigens associated with these favorable responses remain unclear. We hypothesized that a clinical strategy involving the iterative adoptive transfer of selected autologous antigen-specific T-cell clones could help systematically define immunologic targets associated with successful cancer therapy, without the interpretative ambiguity of transferring polyclonal populations. Here, we evaluated the clinical efficacy of CD8(+) T-cell clones specific for the melanocyte differentiation antigens (MDA), gp100 and MART-1, respectively. EXPERIMENTAL DESIGN We conducted two consecutive phase II clinical trials involving the adoptive transfer of highly selected autologous antigen-specific CD8(+) T-cell clones against gp100 and MART-1, respectively. Fifteen patients with HLA-A2(+) treatment-refractory metastatic melanoma received highly avid MDA-specific CD8(+) T-cell clones specific for either gp100 (n = 10) or MART-1 (n = 5) with or without intravenous interleukin-2 (IL2) after a lymphodepleting myeloablative preparative regimen. RESULTS Of the 15 treated patients, we observed immune-mediated targeting of skin melanocytes in 11 patients (73%) and clonal engraftment in eight patients (53%) after cell transfer. There were only transient minor tumor regressions observed, but no objective tumor responses based on Response Evaluation Criteria in Solid Tumor (RECIST) criteria. CONCLUSIONS Despite successful clonal repopulation and evidence of in vivo antigen targeting, the poor therapeutic efficacy after the adoptive transfer of autologous MDA-specific T cells raises significant concerns regarding future immunotherapy efforts targeting this class of tumor antigens.
Collapse
Affiliation(s)
- Smita S Chandran
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Biman C Paria
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Abhishek K Srivastava
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Luke D Rothermel
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Daniel J Stephens
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Mark E Dudley
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Robert Somerville
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - John R Wunderlich
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Richard M Sherry
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - James C Yang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Steven A Rosenberg
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Udai S Kammula
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
29
|
Prediction and identification of HLA-A*0201-restricted epitopes from leukemia-associated protein MLAA-22 which elicit cytotoxic T lymphocytes. Med Oncol 2014; 31:293. [PMID: 25355639 DOI: 10.1007/s12032-014-0293-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/13/2014] [Indexed: 10/24/2022]
Abstract
Cytotoxic T lymphocytes (CTLs) play a critical role in the control of leukemia. However, few effective CTL epitopes have been identified to date yet. We previously reported that MLAA-22, a protein composed of 631 amino acid residues, is a novel acute myeloid leukemia (AML)-associated antigen. In the present study, ten high-score 9-mer peptides, which were selected from MLAA-22 by using ProPred1 and SYFPEITHI bioinformatics tools, were screened to identify HLA-A*0201-restricted-specific CTL epitopes. Monocyte-derived dendritic cells were generated in vitro to be used as antigen-presenting cells for the induction of CTLs. We found that peptide MLAA-22(379-387) (LLPNAIYKV) exhibited the highest binding affinity to HLA-A*0201 among all peptide candidates in the peptide-T2 binding assay. The percentage of positive T2 cells treated with MLAA-22(379-387) was about 96.3%, which is even higher than that of the positive control peptide CML28(173-181) (95.1%). MLAA-22(379-387)-induced CTLs showed the most significant cytotoxic activity and apparent killing effects on the cell lines including THP-1 (human acute monocytic leukemia), A549, T2, U937, and MCF-7, and the specific lysis ratios were 83.8, 32.6, 64.4, 64.4, and 32.6%, respectively, when the effector to target ratio (E/T) was 20:1. Specific lysis (%) of MLAA1 was significantly increased (P < 0.05, P < 0.001, respectively) in THP-1 cell than those in other cancer cell lines and were 28.5, 67.8, and 83.8% at ratio 5:1, 10:1, and 20:1, respectively. Hence, MLAA-22(379-387) is a potential tumor-associated antigen target for AML immunotherapy.
Collapse
|
30
|
Butler MO, Hirano N. Human cell-based artificial antigen-presenting cells for cancer immunotherapy. Immunol Rev 2014; 257:191-209. [PMID: 24329798 DOI: 10.1111/imr.12129] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adoptive T-cell therapy, where anti-tumor T cells are first prepared in vitro, is attractive since it facilitates the delivery of essential signals to selected subsets of anti-tumor T cells without unfavorable immunoregulatory issues that exist in tumor-bearing hosts. Recent clinical trials have demonstrated that anti-tumor adoptive T-cell therapy, i.e. infusion of tumor-specific T cells, can induce clinically relevant and sustained responses in patients with advanced cancer. The goal of adoptive cell therapy is to establish anti-tumor immunologic memory, which can result in life-long rejection of tumor cells in patients. To achieve this goal, during the process of in vitro expansion, T-cell grafts used in adoptive T-cell therapy must be appropriately educated and equipped with the capacity to accomplish multiple, essential tasks. Adoptively transferred T cells must be endowed, prior to infusion, with the ability to efficiently engraft, expand, persist, and traffic to tumor in vivo. As a strategy to consistently generate T-cell grafts with these capabilities, artificial antigen-presenting cells have been developed to deliver the proper signals necessary to T cells to enable optimal adoptive cell therapy.
Collapse
Affiliation(s)
- Marcus O Butler
- Immune Therapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
31
|
Strioga MM, Darinskas A, Pasukoniene V, Mlynska A, Ostapenko V, Schijns V. Xenogeneic therapeutic cancer vaccines as breakers of immune tolerance for clinical application: to use or not to use? Vaccine 2014; 32:4015-24. [PMID: 24837511 DOI: 10.1016/j.vaccine.2014.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 02/06/2023]
Abstract
Accumulation of firm evidence that clinically apparent cancer develops only when malignant cells manage to escape immunosurveillance led to the introduction of tumor immunotherapy strategies aiming to reprogramm the cancer-dysbalanced antitumor immunity and restore its capacity to control tumor growth. There are several immunotherapeutical strategies, among which specific active immunotherapy or therapeutic cancer vaccination is one of the most promising. It targets dendritic cells (DCs) which have a unique ability of inducing naive and central memory T cell-mediated immune response in the most efficient manner. DCs can be therapeutically targeted either in vivo/in situ or by ex vivo manipulations followed by their re-injection back into the same patient. The majority of current DC targeting strategies are based on autologous or allogeneic tumor-associated antigens (TAAs) which possess various degrees of inherent tolerogenic potential. Therefore still limited efficacy of various tumor immunotherapy approaches may be attributed, among various other mechanisms, to the insufficient immunogenicity of self-protein-derived TAAs. Based on such an idea, the use of homologous xenogeneic antigens, derived from different species was suggested to overcome the natural immune tolerance to self TAAs. Xenoantigens are supposed to differ sufficiently from self antigens to a degree that renders them immunogenic, but at the same time preserves an optimal homology range with self proteins still allowing xenoantigens to induce cross-reactive T cells. Here we discuss the concept of xenogeneic vaccination, describe the cons and pros of autologous/allogeneic versus xenogeneic therapeutic cancer vaccines, present the results of various pre-clinical and several clinical studies and highlight the future perspectives of integrating xenovaccination into rapidly developing tumor immunotherapy regimens.
Collapse
Affiliation(s)
- Marius M Strioga
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Adas Darinskas
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Vita Pasukoniene
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Agata Mlynska
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Valerijus Ostapenko
- Section of Breast Surgery, 3(rd) Department of Surgery, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Virgil Schijns
- Immune Intervention, Cell Biology & Immunology group, Wageningen University, Wageningen, the Netherlands; Epitopoietic Research Corporation (ERC), Namur, Belgium.
| |
Collapse
|
32
|
Ménager J, Ebstein F, Oger R, Hulin P, Nedellec S, Duverger E, Lehmann A, Kloetzel PM, Jotereau F, Guilloux Y. Cross-presentation of synthetic long peptides by human dendritic cells: a process dependent on ERAD component p97/VCP but Not sec61 and/or Derlin-1. PLoS One 2014; 9:e89897. [PMID: 24587108 PMCID: PMC3937416 DOI: 10.1371/journal.pone.0089897] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/25/2014] [Indexed: 12/22/2022] Open
Abstract
Antitumor vaccination using synthetic long peptides (SLP) is an additional therapeutic strategy currently under development. It aims to activate tumor-specific CD8+ CTL by professional APCs such as DCs. DCs can activate T lymphocytes by MHC class I presentation of exogenous antigens - a process referred to as “cross-presentation”. Until recently, the intracellular mechanisms involved in cross-presentation of soluble antigens have been unclear. Here, we characterize the cross-presentation pathway of SLP Melan-A16–40 containing the HLA-A2-restricted epitope26–35 (A27L) in human DCs. Using confocal microscopy and specific inhibitors, we show that SLP16–40 is rapidly taken up by DC and follows a classical TAP- and proteasome-dependent cross-presentation pathway. Our data support a role for the ER-associated degradation machinery (ERAD)-related protein p97/VCP in the transport of SLP16–40 from early endosomes to the cytoplasm but formally exclude both sec61 and Derlin-1 as possible retro-translocation channels for cross-presentation. In addition, we show that generation of the Melan-A26–35 peptide from the SLP16–40 was absolutely not influenced by the proteasome subunit composition in DC. Altogether, our findings propose a model for cross-presentation of SLP which tends to enlarge the repertoire of potential candidates for retro-translocation of exogenous antigens to the cytosol.
Collapse
Affiliation(s)
- Jérémie Ménager
- INSERM U892, Nantes, France
- Université de Nantes, Nantes, France
- CNRS, UMR 6299, Nantes, France
| | - Frédéric Ebstein
- Institut of Biochemistry, Charité University Hospital, Humboldt University, Berlin, Germany
| | - Romain Oger
- INSERM U892, Nantes, France
- Université de Nantes, Nantes, France
- CNRS, UMR 6299, Nantes, France
| | - Philippe Hulin
- INSERM U892, Nantes, France
- CNRS, UMR 6299, Nantes, France
| | | | - Eric Duverger
- Glycobiochimie, ICOA, Université d’Orléans, Orléans, France
| | - Andrea Lehmann
- Institut of Biochemistry, Charité University Hospital, Humboldt University, Berlin, Germany
| | - Peter-Michael Kloetzel
- Institut of Biochemistry, Charité University Hospital, Humboldt University, Berlin, Germany
| | - Francine Jotereau
- INSERM U892, Nantes, France
- Université de Nantes, Nantes, France
- CNRS, UMR 6299, Nantes, France
| | - Yannick Guilloux
- INSERM U892, Nantes, France
- Université de Nantes, Nantes, France
- CNRS, UMR 6299, Nantes, France
- * E-mail:
| |
Collapse
|
33
|
A full GMP process to select and amplify epitope-specific T lymphocytes for adoptive immunotherapy of metastatic melanoma. Clin Dev Immunol 2013; 2013:932318. [PMID: 24194775 PMCID: PMC3806119 DOI: 10.1155/2013/932318] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 08/27/2013] [Accepted: 08/27/2013] [Indexed: 12/31/2022]
Abstract
A number of trials of adoptive transfer of tumor-specific T lymphocytes have been performed in the last 20 years in metastatic melanoma, with increasingly encouraging results as the relevant melanoma antigens were identified and the purity/specificity of injected T cells improved. We have previously described a sorting method of epitope-specific T lymphocytes that uses magnetic beads coated with HLA/peptide complexes and we suggested that this method could be applied to a clinical setting. In the present work, we provide a detailed description of the whole GMP process of sorting and amplification of clinical grade T cells specific for the melanoma antigens Melan-A and MELOE-1. All the reagents used in this process including the sorting reagent were produced in GMP conditions and we document the optimization of the different steps of the process such as peptide stimulation, sorting, and amplification. The optimized procedure, validated in 3 blank runs in a clinical setting, allowed the production of at least 108 pure (>90%) Melan-A- and MELOE-1-specific T cells within 28 days starting with 100 mL of blood from metastatic melanoma patients. This GMP process is thus ready to be used in an upcoming phase I/II clinical trial on metastatic melanoma patients.
Collapse
|
34
|
Wang A, Chandran S, Shah SA, Chiu Y, Paria BC, Aghamolla T, Alvarez-Downing MM, Lee CCR, Singh S, Li T, Dudley ME, Restifo NP, Rosenberg SA, Kammula US. The stoichiometric production of IL-2 and IFN-γ mRNA defines memory T cells that can self-renew after adoptive transfer in humans. Sci Transl Med 2013; 4:149ra120. [PMID: 22932225 DOI: 10.1126/scitranslmed.3004306] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adoptive immunotherapy using ex vivo-expanded tumor-reactive lymphocytes can mediate durable cancer regression in selected melanoma patients. Analyses of these trials have associated the in vivo engraftment ability of the transferred cells with their antitumor efficacy. Thus, there is intensive clinical interest in the prospective isolation of tumor-specific T cells that can reliably persist after transfer. Animal studies have suggested that central memory CD8(+) T cells (T(CM)) have divergent capabilities including effector differentiation to target antigen and stem cell-like self-renewal that enable long-term survival after adoptive transfer. We sought to isolate human melanoma-specific T(CM) to define their in vivo fate and function after autologous therapeutic transfer to metastatic patients. To facilitate the high-throughput identification of these rare cells from patients, we report that T(CM) have a defined stoichiometric production of interleukin-2 (IL-2) and interferon-γ (IFN-γ) mRNA after antigen stimulation. Melanoma-specific T cells screened for high relative IL-2 production had a T(CM) phenotype and superior in vitro proliferative capacity compared to cells with low IL-2 production. To investigate in vivo effector function and self-renewal capability, we allowed melanoma-specific T(CM) to undergo in vitro expansion and differentiation into lytic effector clones and then adoptively transferred them back into their hosts. These clones targeted skin melanocytes in all five patients and persisted long term and reacquired parental T(CM) attributes in four patients after transfer. These findings demonstrate the favorable engraftment fitness for human T(CM)-derived clones, but further efforts to improve their antitumor efficacy are still necessary.
Collapse
Affiliation(s)
- Anran Wang
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20817, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology 2012; 1:306-315. [PMID: 22737606 PMCID: PMC3382856 DOI: 10.4161/onci.19549] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
During the last two decades, several approaches for the activation of the immune system against cancer have been developed. These include rather unselective maneuvers such as the systemic administration of immunostimulatory agents (e.g., interleukin-2) as well as targeted interventions, encompassing highly specific monoclonal antibodies, vaccines and cell-based therapies. Among the latter, adoptive cell transfer (ACT) involves the selection of autologous lymphocytes with antitumor activity, their expansion/activation ex vivo, and their reinfusion into the patient, often in the context of lymphodepleting regimens (to minimize endogenous immunosuppression). Such autologous cells can be isolated from tumor-infiltrating lymphocytes or generated by manipulating circulating lymphocytes for the expression of tumor-specific T-cell receptors. In addition, autologous lymphocytes can be genetically engineered to prolong their in vivo persistence, to boost antitumor responses and/or to minimize side effects. ACT has recently been shown to be associated with a consistent rate of durable regressions in melanoma and renal cell carcinoma patients and holds great promises in several other oncological settings. In this Trial Watch, we will briefly review the scientific rationale behind ACT and discuss the progress of recent clinical trials evaluating the safety and effectiveness of adoptive cell transfer as an anticancer therapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- INSERM; U848; Villejuif, France
- Institut Gustave Roussy; Villejuif, France
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
| | - Erika Vacchelli
- INSERM; U848; Villejuif, France
- Institut Gustave Roussy; Villejuif, France
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
| | | | - Wolf Herve´ Fridman
- INSERM; U872; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Jerome Galon
- INSERM; U872; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
| | - Catherine Sautès-Fridman
- INSERM; U872; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Université Paris Descartes; Sorbonne Paris Cité; Paris, France
| | - Eric Tartour
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
- Université Paris Descartes; Sorbonne Paris Cité; Paris, France
- INSERM; U970; Paris, France
| | - Laurence Zitvogel
- Institut Gustave Roussy; Villejuif, France
- INSERM; U1015; Villejuif, France
| | - Guido Kroemer
- INSERM; U848; Villejuif, France
- Institut Gustave Roussy; Villejuif, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
- Université Paris Descartes; Sorbonne Paris Cité; Paris, France
- Metabolomics Platform; Institut Gustave Roussy; Villejuif, France
| |
Collapse
|
36
|
Ochsenreither S, Fusi A, Geikowski A, Stather D, Busse A, Stroux A, Letsch A, Keilholz U. Wilms' tumor protein 1 (WT1) peptide vaccination in AML patients: predominant TCR CDR3β sequence associated with remission in one patient is detectable in other vaccinated patients. Cancer Immunol Immunother 2012; 61:313-22. [PMID: 21898091 PMCID: PMC11029123 DOI: 10.1007/s00262-011-1099-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 08/13/2011] [Indexed: 12/01/2022]
Abstract
BACKGROUND Clinically effective T-cell responses can be elicited by single peptide vaccination with Wilms' tumor 1 (WT1) epitope 126-134 in patients with acute myeloid leukemia (AML). We recently showed that a predominant T-cell receptor (TCR) β chain was associated with vaccine-induced complete remission in an AML patient (patient 1). In this study, we address the question of whether this predominant clone or the accompanying Vβ11 restriction could be found in other AML patients vaccinated with the same WT1 peptide. MATERIALS AND METHODS For assessment of Vβ usage, cytotoxic T lymphocytes (CTLs) from four vaccinated patients were divided into specific and non-specific by epitope-specific enrichment. Vβ families were quantified in both fractions using reverse transcribed quantitative PCR. Vβ11-positive 'complementary determining region 3' (CDR3) sequences were amplified from these samples, from bone marrow samples of 17 other vaccination patients, and from peripheral blood of six healthy controls, cloned and sequenced. RESULTS We observed a clear bias towards Vβ11 usage of the WT1-specific CTL populations in all four patients. The predominant CDR3β amino acid (AA) sequence of patient 1 was detected in two other patients. CDR3β loops with closely related AA sequences were only found in patient 1. There were no CDR3β AA sequences with side chains of identical chemical properties detected in any patient. CONCLUSION We provide the first data addressing TCR Vβ chain usage in WT1-specific T-cell populations after HLA A*0201-restricted single peptide vaccination. We demonstrate both shared Vβ restriction and the sharing of a TCR β transcript with proven clinical impact in one patient.
Collapse
MESH Headings
- Acute Disease
- Adult
- Aged
- Aged, 80 and over
- Amino Acid Sequence
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Clone Cells/immunology
- Clone Cells/metabolism
- Complementarity Determining Regions/genetics
- Complementarity Determining Regions/immunology
- Epitopes/immunology
- Female
- HLA-A2 Antigen/immunology
- Humans
- Leukemia, Myeloid/immunology
- Leukemia, Myeloid/therapy
- Male
- Middle Aged
- Molecular Sequence Data
- Peptide Fragments/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Remission Induction
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Vaccination
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- WT1 Proteins/chemistry
- WT1 Proteins/immunology
- Young Adult
Collapse
Affiliation(s)
- Sebastian Ochsenreither
- Department of Hematology and Oncology, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Alberto Fusi
- Department of Hematology and Oncology, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Anne Geikowski
- Department of Hematology and Oncology, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - David Stather
- Department of Hematology and Oncology, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Antonia Busse
- Department of Hematology and Oncology, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Andrea Stroux
- Institute for Biometry and Clinical Epidemiology, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Anne Letsch
- Department of Hematology and Oncology, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Ulrich Keilholz
- Department of Hematology and Oncology, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
- Department of Medicine III, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| |
Collapse
|
37
|
Jotereau F, Gervois N, Labarrière N. Adoptive transfer with high-affinity TCR to treat human solid tumors: how to improve the feasibility? Target Oncol 2012; 7:3-14. [PMID: 22350487 DOI: 10.1007/s11523-012-0207-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 01/12/2012] [Indexed: 01/05/2023]
Abstract
The adoptive transfer of tumor antigen-specific T cells recently achieved clinical efficacy for a fraction of melanoma patients refractory to other therapies. Unfortunately, the application of this strategy to the remaining melanoma and most other cancer patients is hampered by the difficulty to generate high-affinity tumor-reactive T cells. Two strategies are currently developed to extend the feasibility of this therapeutic approach: clinical grade tool production for MHC-peptide multimer-driven sorting of antigen-specific T cells from the endogenous peripheral T cell repertoire and de novo engineering of the missing repertoire by genetic transfer of cloned specific T cell receptor (TCR) into T cells. The expected multiplication of adoptive transfer treatments, by these strategies, and their careful evaluation should enable the cure of a number of otherwise compromised cancer patients and to gain insight into the characteristics of transferred T cells best fitted to eradicate tumor cells, in terms of antigen specificities, phenotype, and functions. In particular, identification of tumor-rejection antigens by this approach would improve the design and efficacy of all immunotherapeutic approaches.
Collapse
|
38
|
Chauvin JM, Larrieu P, Sarrabayrouse G, Prévost-Blondel A, Lengagne R, Desfrançois J, Labarrière N, Jotereau F. HLA anchor optimization of the melan-A-HLA-A2 epitope within a long peptide is required for efficient cross-priming of human tumor-reactive T cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:2102-10. [PMID: 22291187 DOI: 10.4049/jimmunol.1101807] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The uptake and long-term cross-presentation of tumor Ag long peptides (LP) by dendritic cells (DC) make them attractive cancer vaccine candidates. However, it remains to be established whether LP can prime long-lived tumor-reactive CTL and whether other cell types are able to cross-present them. Using HLA-A2 healthy donor and melanoma patient-derived PBMC, we studied the in vitro cross-priming potential of Melan-A 16-40 LP bearing the HLA-A2-restricted epitope 26-35 or its analog 26-35(A27L) and compared it to the priming capacity of the short analog. We then addressed LP priming capacity in vivo using HLA-A2 mice. We also studied LP cross-presentation by monocyte-derived DC, plasmacytoid DC, monocytes, and B cells. We showed that the modified LP gave rise to high and sustained cross-presentation by monocyte-derived DC. This led to cross priming in vitro and in vivo and to expansion of long-lived tumor-reactive cytotoxic T cells. In contrast, the LP containing the natural 26-35 epitope primed specific T cells poorly, despite its long-lived cross-presentation, and T cells primed against the short analog were short-lived. We further showed that LP cross-presentation is restricted to monocytes and conventional DC. These results document for the first time, to our knowledge, the strong immunogenicity of a human tumor Ag LP. Of note, they underscore that this property is critically dependent on sufficient HLA binding affinity and/or TCR ligand potency of the cross-presented epitope. We conclude that LP fulfilling this requirement should be used as tumor vaccines, together with DC maturating agents, especially the Melan-A 16-40(A27L) LP, for the treatment of HLA-A2(+) melanoma patients.
Collapse
|
39
|
Abstract
Historically, sharing T cell receptors (TCRs) between individuals has been speculated to be impossible, considering the dramatic discrepancy between the potential enormity of the TCR repertoire and the limited number of T cells generated in each individual. However, public T cell response, in which multiple individuals share identical TCRs in responding to a same antigenic epitope, has been extensively observed in a variety of immune responses across many species. Public T cell responses enable individuals within a population to generate similar antigen-specific TCRs against certain ubiquitous pathogens, leading to favorable biological outcomes. However, the relatively concentrated feature of TCR repertoire may limit T cell response in a population to some other pathogens. It could be a great benefit for human health if public T cell responses can be manipulated. Therefore, the mechanistic insight of public TCR generation is important to know. Recently, high-throughput DNA sequencing has revolutionized the study of immune receptor repertoires, which allows a much better understanding of the factors that determine the overlap of TCR repertoire among individuals. Here, we summarize the current knowledge on public T-cell response and discuss future challenges in this field.
Collapse
Affiliation(s)
- Hanjie Li
- State Key Laboratory of Cellular Stress Biology and School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | | | | | | |
Collapse
|
40
|
Wieërs G, Demotte N, Godelaine D, van der Bruggen P. Immune suppression in tumors as a surmountable obstacle to clinical efficacy of cancer vaccines. Cancers (Basel) 2011; 3:2904-54. [PMID: 24212939 PMCID: PMC3759179 DOI: 10.3390/cancers3032904] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 07/01/2011] [Accepted: 07/07/2011] [Indexed: 02/07/2023] Open
Abstract
Human tumors are usually not spontaneously eliminated by the immune system and therapeutic vaccination of cancer patients with defined antigens is followed by tumor regressions only in a small minority of the patients. The poor vaccination effectiveness could be explained by an immunosuppressive tumor microenvironment. Because T cells that infiltrate tumor metastases have an impaired ability to lyse target cells or to secrete cytokine, many researchers are trying to decipher the underlying immunosuppressive mechanisms. We will review these here, in particular those considered as potential therapeutic targets. A special attention will be given to galectins, a family of carbohydrate binding proteins. These lectins have often been implicated in inflammation and cancer and may be useful targets for the development of new anti-cancer therapies.
Collapse
Affiliation(s)
- Grégoire Wieërs
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| | - Nathalie Demotte
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| | - Danièle Godelaine
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| | - Pierre van der Bruggen
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| |
Collapse
|
41
|
Labarriere N, Khammari A, Lang F, Dreno B. Is antigen specificity the key to efficient adoptive T-cell therapy? Immunotherapy 2011; 3:495-505. [DOI: 10.2217/imt.11.16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Adoptive transfer of T cells remains a promising approach in melanoma. Initial clinical trials performed with polyclonal tumor-infiltrating lymphocyte gave limited clinical results. Nonetheless, encouraging results have been reported in adjuvant setting (stage III melanoma), and when tumor-infiltrating lymphocytes were associated with lymphodepleting regimens. Specificity of adoptive cell therapy has been achieved with the infusion of antigen specific cytotoxic T-lymphocyte clones, associated with some clinical responses. Antigen specificity can also be obtained by the allogeneic transfer of high-avidity T-cell receptors into autologous T cells. We propose an alternative strategy based on the selection of antigen-specific T cells with magnetic beads coated with HLA–peptide multimers. Future improvements of adoptive melanoma immunotherapy may be achieved by its association with other therapeutic strategies such as targeted therapy against signaling pathways.
Collapse
Affiliation(s)
- Nathalie Labarriere
- Unite Mixte de Recherche Institut National de la Sante et de la Recherche Medicale, Unite 892, Centre de Recherche en Canerologie Nantes-Angers, F-44007 Nantes, France
| | - Amir Khammari
- Unite Mixte de Recherche Institut National de la Sante et de la Recherche Medicale, Unite 892, Centre de Recherche en Canerologie Nantes-Angers, F-44007 Nantes, France
- Centre Hospitalo-Universitaire de Nantes, Unit of Skin Cancer, F-44093 Nantes, France
| | - Francois Lang
- Unite Mixte de Recherche Institut National de la Sante et de la Recherche Medicale, Unite 892, Centre de Recherche en Canerologie Nantes-Angers, F-44007 Nantes, France
- Université de Nantes, Unite de Formation et de Recherche des Sciences Pharmaceutiques, F-44093 Nantes, France
| | | |
Collapse
|
42
|
Miles JJ, Douek DC, Price DA. Bias in the αβ T-cell repertoire: implications for disease pathogenesis and vaccination. Immunol Cell Biol 2011; 89:375-87. [PMID: 21301479 DOI: 10.1038/icb.2010.139] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The naïve T-cell repertoire is vast, containing millions of unique T-cell receptor (TCR) structures. Faced with such diversity, the mobilization of TCR structures from this enormous pool was once thought to be a stochastic, even chaotic, process. However, steady and systematic dissection over the last 20 years has revealed that this is not the case. Instead, the TCR repertoire deployed against individual antigens is routinely ordered and biased. Often, identical and near-identical TCR repertoires can be observed across different individuals, suggesting that the system encompasses an element of predictability. This review provides a catalog of αβ TCR bias by disease and by species, and discusses the mechanisms that govern this inherent and widespread phenomenon.
Collapse
Affiliation(s)
- John J Miles
- T Cell Modulation Laboratory, Department of Infection, Immunity and Biochemistry, Cardiff University School of Medicine, Cardiff, UK.
| | | | | |
Collapse
|
43
|
Corbière V, Chapiro J, Stroobant V, Ma W, Lurquin C, Lethé B, van Baren N, Van den Eynde BJ, Boon T, Coulie PG. Antigen spreading contributes to MAGE vaccination-induced regression of melanoma metastases. Cancer Res 2011; 71:1253-62. [PMID: 21216894 DOI: 10.1158/0008-5472.can-10-2693] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A core challenge in cancer immunotherapy is to understand the basis for efficacious vaccine responses in human patients. In previous work we identified a melanoma patient who displayed a low-level antivaccine cytolytic T-cell (CTL) response in blood with tumor regression after vaccination with melanoma antigens (MAGE). Using a genetic approach including T-cell receptor β (TCRβ) cDNA libraries, we found very few antivaccine CTLs in regressing metastases. However, a far greater number of TCRβ sequences were found with several of these corresponding to CTL clones specific for nonvaccine tumor antigens, suggesting that antigen spreading was occurring in regressing metastases. In this study, we found another TCR belonging to tumor-specific CTL enriched in regressing metastases and detectable in blood only after vaccination. We used the TCRβ sequence to detect and clone the desired T cells from tumor-infiltrating lymphocytes isolated from the patient. This CD8 clone specifically lysed autologous melanoma cells and displayed HLA-A2 restriction. Its target antigen was identified as the mitochondrial enzyme caseinolytic protease. The target antigen gene was mutated in the tumor, resulting in production of a neoantigen. Melanoma cell lysis by the CTL was increased by IFN-γ treatment due to preferential processing of the antigenic peptide by the immunoproteasome. These results argue that tumor rejection effectors in the patient were indeed CTL responding to nonvaccine tumor-specific antigens, further supporting our hypothesis. Among such antigens, the mutated antigen we found is the only antigen against which no T cells could be detected before vaccination. We propose that antigen spreading of an antitumor T-cell response to truly tumor-specific antigens contributes decisively to tumor regression.
Collapse
Affiliation(s)
- Véronique Corbière
- de Duve Institute, Université Catholique de Louvain and Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
T-cell therapy involves the ex vivo isolation and expansion of antigen-specific T cells for adoptive transfer. The use of T-cell clones represents one embodiment of this approach and provides a uniform population of effector cells, so that parameters contributing to an effective response can be rigorously evaluated. T cells of defined specificity, phenotype, and function are isolated and expanded; when infused into patients, these intrinsic factors can be considered in light of extrinsic factors such as the type of conditioning regimen, cytokine support, and immunomodulatory reagents. In this chapter, 2 topics related to the use of antigen-specific T-cell clones are discussed: first, advances enabling the isolation and expansion of antigen-specific T-cell clones for human trials of adoptive therapy, and second, a contextual framework of advantages and limitations in which the use of adoptively transferred T-cell clones can be judiciously applied as a means to dissect the requirements for effective therapy.
Collapse
|
45
|
Nguyen LT, Yen PH, Nie J, Liadis N, Ghazarian D, Al-Habeeb A, Easson A, Leong W, Lipa J, McCready D, Reedijk M, Hogg D, Joshua AM, Quirt I, Messner H, Shaw P, Crump M, Sharon E, Ohashi PS. Expansion and characterization of human melanoma tumor-infiltrating lymphocytes (TILs). PLoS One 2010; 5:e13940. [PMID: 21085676 PMCID: PMC2978109 DOI: 10.1371/journal.pone.0013940] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 10/19/2010] [Indexed: 12/26/2022] Open
Abstract
Background Various immunotherapeutic strategies for cancer are aimed at augmenting the T cell response against tumor cells. Adoptive cell therapy (ACT), where T cells are manipulated ex vivo and subsequently re-infused in an autologous manner, has been performed using T cells from various sources. Some of the highest clinical response rates for metastatic melanoma have been reported in trials using tumor-infiltrating lymphocytes (TILs). These protocols still have room for improvement and furthermore are currently only performed at a limited number of institutions. The goal of this work was to develop TILs as a therapeutic product at our institution. Principal Findings TILs from 40 melanoma tissue specimens were expanded and characterized. Under optimized culture conditions, 72% of specimens yielded rapidly proliferating TILs as defined as at least one culture reaching ≥3×107 TILs within 4 weeks. Flow cytometric analyses showed that cultures were predominantly CD3+ T cells, with highly variable CD4+:CD8+ T cell ratios. In total, 148 independent bulk TIL cultures were assayed for tumor reactivity. Thirty-four percent (50/148) exhibited tumor reactivity based on IFN-γ production and/or cytotoxic activity. Thirteen percent (19/148) showed specific cytotoxic activity but not IFN-γ production and only 1% (2/148) showed specific IFN-γ production but not cytotoxic activity. Further expansion of TILs using a 14-day “rapid expansion protocol” (REP) is required to induce a 500- to 2000-fold expansion of TILs in order to generate sufficient numbers of cells for current ACT protocols. Thirty-eight consecutive test REPs were performed with an average 1865-fold expansion (+/− 1034-fold) after 14 days. Conclusions TILs generally expanded efficiently and tumor reactivity could be detected in vitro. These preclinical data from melanoma TILs lay the groundwork for clinical trials of ACT.
Collapse
Affiliation(s)
- Linh T. Nguyen
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Pei Hua Yen
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Jessica Nie
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Nicole Liadis
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Danny Ghazarian
- Department of Pathology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Ayman Al-Habeeb
- Department of Pathology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Alexandra Easson
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Wey Leong
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Joan Lipa
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - David McCready
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Michael Reedijk
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - David Hogg
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Anthony M. Joshua
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Ian Quirt
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Hans Messner
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Patricia Shaw
- Department of Pathology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Michael Crump
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Eran Sharon
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Pamela S. Ohashi
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
- Departments of Medical Biophysics and Immunology, University of Toronto, Toronto, Canada
- * E-mail:
| |
Collapse
|
46
|
Enrichment of CD8+ cells from melanoma tumor-infiltrating lymphocyte cultures reveals tumor reactivity for use in adoptive cell therapy. J Immunother 2010; 33:547-56. [PMID: 20463593 DOI: 10.1097/cji.0b013e3181d367bd] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TIL) for metastatic melanoma has shown objective response rates as high as 72%. The successful application of this therapy requires the selection of unique tumor-reactive lymphocyte cultures for each patient. This is a technically and logistically difficult undertaking, and patients who do not have tumor-reactive TIL are not considered eligible for treatment. To simplify the methods of TIL generation and extend TIL-based immunotherapy to additional patients, methods were developed to use unselected, minimally cultured ("young") TIL. Young TIL cultures contain a variable number of CD8(+), CD4(+), and CD3(-)CD56(+) natural killer cells. In this study we retrospectively investigated a role for these subsets in the clinical outcome of patients treated with TIL derived from selected microcultures. This analysis demonstrated a suggestive but nonsignificant association between the number of CD8(+) cells administered and tumor regression. We therefore investigated the feasibility of selecting CD8(+) cells from young TIL cultures for ACT therapy. The available methods for clinical scale CD8(+) enrichment proved inadequate for TIL, so an optimized CD8(+) enrichment method was developed and is reported here. We observed that CD8 (+)enrichment of some TIL cultures revealed in vitro tumor recognition that was not evident in bulk culture, and an improved in vitro recognition of tumor in other TIL cultures. In addition, the enriched CD8(+) young TIL expanded more reliably and predictably in rapid expansions than the bulk TIL. Thus, optimized CD8(+) selection combines the benefits of antigen-selected TIL and young TIL for generating lymphocyte cultures for ACT, and should be evaluated in cell transfer therapy protocols.
Collapse
|
47
|
Cole DK, Edwards ESJ, Wynn KK, Clement M, Miles JJ, Ladell K, Ekeruche J, Gostick E, Adams KJ, Skowera A, Peakman M, Wooldridge L, Price DA, Sewell AK. Modification of MHC anchor residues generates heteroclitic peptides that alter TCR binding and T cell recognition. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:2600-10. [PMID: 20639478 PMCID: PMC3024538 DOI: 10.4049/jimmunol.1000629] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Improving T cell Ags by altering MHC anchor residues is a common strategy used to enhance peptide vaccines, but there has been little assessment of how such modifications affect TCR binding and T cell recognition. In this study, we use surface plasmon resonance and peptide-MHC tetramer binding at the cell surface to demonstrate that changes in primary peptide anchor residues can substantially and unpredictably alter TCR binding. We also demonstrate that the ability of TCRs to differentiate between natural and anchor-modified heteroclitic peptides distinguishes T cells that exhibit a strong preference for either type of Ag. Furthermore, we show that anchor-modified heteroclitic peptides prime T cells with different TCRs compared with those primed with natural Ag. Thus, vaccination with heteroclitic peptides may elicit T cells that exhibit suboptimal recognition of the intended natural Ag and, consequently, impaired functional attributes in vivo. Heteroclitic peptide-based immune interventions therefore require careful evaluation to ensure efficacy in the clinic.
Collapse
MESH Headings
- Amino Acid Sequence
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Cytokines/immunology
- Cytokines/metabolism
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Flow Cytometry
- HLA-A Antigens/genetics
- HLA-A Antigens/immunology
- HLA-A Antigens/metabolism
- HLA-A2 Antigen
- Humans
- Mutation
- Oligopeptides/genetics
- Oligopeptides/immunology
- Oligopeptides/metabolism
- Peptide Library
- Protein Binding/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Surface Plasmon Resonance
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- David K Cole
- Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Godet Y, Desfrançois J, Vignard V, Schadendorf D, Khammari A, Dreno B, Jotereau F, Labarrière N. Frequent occurrence of high affinity T cells against MELOE-1 makes this antigen an attractive target for melanoma immunotherapy. Eur J Immunol 2010; 40:1786-94. [PMID: 20217862 DOI: 10.1002/eji.200940132] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We recently showed that the infusion of tumor infiltrating lymphocytes specific for the MELOE-1 antigen was associated with a prolonged relapse-free survival for HLA-A2(+) melanoma patients who received tumor infiltrating lymphocytes therapy. Here, we characterized the MELOE-1/A2-specific T-cell repertoire in healthy donors and melanoma patients to further support an immunotherapy targeting this epitope. Using tetramer enrichment followed by multicolor staining, we found that MELOE-1-specific T cells were present in the blood of healthy donors and patients at similar frequencies (around 1 in 1x10(5) CD8(+) cells). These cells mainly displayed a naïve phenotype in 4/6 healthy donors and 3/6 patients, whereas high proportions of memory cells were observed in the remaining individuals of both groups. There was a recurrent usage of the Valpha12.1 chain for 17/18 MELOE-1-specific T-cell clones derived from healthy donors or patients, associated with diverse Vbeta chains and V(D)J junctional sequences. All clones derived from melanoma patients (9/9) were reactive against the MELOE-1(36-44) peptide and against HLA-A2(+) melanoma cell lines. This study documents the existence of a large TCR repertoire specific for the MELOE-1/A2 epitope and its capacity to give rise to antitumor CTL that supports the development of immunotherapies targeting this epitope.
Collapse
|
49
|
Godet Y, Moreau-Aubry A, Mompelat D, Vignard V, Khammari A, Dreno B, Lang F, Jotereau F, Labarriere N. An additional ORF on meloe cDNA encodes a new melanoma antigen, MELOE-2, recognized by melanoma-specific T cells in the HLA-A2 context. Cancer Immunol Immunother 2010; 59:431-9. [PMID: 19730858 PMCID: PMC11029903 DOI: 10.1007/s00262-009-0762-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Accepted: 08/20/2009] [Indexed: 11/25/2022]
Abstract
We characterized a new melanoma antigen derived from one of the multiple open reading frames (ORFs) of the meloe transcript. The meloe gene is overexpressed in melanomas as compared to other cancer cell lines and normal tissues. The corresponding transcript is rather unusual, in that it does not contain a long unique ORF but multiple short ORFs. We recently characterized a tumor epitope derived from a polypeptide (MELOE-1) encoded by the ORF(1230-1370) and involved in relapse prevention of melanoma patients treated with autologous tumor infiltrating lymphocytes (TIL). Here we show that the ORF(285-404) encodes a polypeptide called MELOE-2 that also generated a HLA-A2 epitope recognized by a melanoma-specific T cell clone derived from the same TIL population from which we derived the MELOE-1-specific T cell clone. We also showed that HLA-A2 melanoma cells were spontaneously recognized by the MELOE-2-specific T cell clone, and we detected the presence of MELOE-2 reactive T cells in another TIL population infused to a patient who remained relapse-free after TIL treatment. These results demonstrate that translation of meloe transcript in melanoma cells can produce at least two immunogenic polypeptides, MELOE-1 and MELOE-2, from two distinct ORFs that could be relevant target for melanoma immunotherapy.
Collapse
Affiliation(s)
- Yann Godet
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
| | - Agnès Moreau-Aubry
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Faculté des Sciences, Université de Nantes, 44322 Nantes, France
| | - Dimitri Mompelat
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
| | - Virginie Vignard
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
| | - Amir Khammari
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Unit of Skin Cancer, Centre Hospitalo-Universitaire de Nantes, 44093 Nantes, France
| | - Brigitte Dreno
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Unit of Skin Cancer, Centre Hospitalo-Universitaire de Nantes, 44093 Nantes, France
| | - Francois Lang
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Université de Nantes, UFR des Sciences Pharmaceutiques, 44322 Nantes, France
| | - Francine Jotereau
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Faculté des Sciences, Université de Nantes, 44322 Nantes, France
| | - Nathalie Labarriere
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
| |
Collapse
|
50
|
Treatment of Metastatic Melanoma with Autologous Melan-A/Mart-1-Specific Cytotoxic T Lymphocyte Clones. J Invest Dermatol 2009; 129:2835-42. [DOI: 10.1038/jid.2009.144] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|