1
|
García-Ferreras R, Osuna-Pérez J, Ramírez-Santiago G, Méndez-Pérez A, Acosta-Moreno AM, Del Campo L, Gómez-Sánchez MJ, Iborra M, Herrero-Fernández B, González-Granado JM, Sánchez-Madrid F, Carrasco YR, Boya P, Martínez-Martín N, Veiga E. Bacteria-instructed B cells cross-prime naïve CD8 + T cells triggering effective cytotoxic responses. EMBO Rep 2023:e56131. [PMID: 37184882 DOI: 10.15252/embr.202256131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/16/2023] Open
Abstract
In addition to triggering humoral responses, conventional B cells have been described in vitro to cross-present exogenous antigens activating naïve CD8+ T cells. Nevertheless, the way B cells capture these exogenous antigens and the physiological roles of B cell-mediated cross-presentation remain poorly explored. Here, we show that B cells capture bacteria by trans-phagocytosis from previously infected dendritic cells (DC) when they are in close contact. Bacterial encounter "instructs" the B cells to acquire antigen cross-presentation abilities, in a process that involves autophagy. Bacteria-instructed B cells, henceforth referred to as BacB cells, rapidly degrade phagocytosed bacteria, process bacterial antigens and cross-prime naïve CD8+ T cells which differentiate into specific cytotoxic cells that efficiently control bacterial infections. Moreover, a proof-of-concept experiment shows that BacB cells that have captured bacteria expressing tumor antigens could be useful as novel cellular immunotherapies against cancer.
Collapse
Affiliation(s)
- Raquel García-Ferreras
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Jesús Osuna-Pérez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Guillermo Ramírez-Santiago
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Almudena Méndez-Pérez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Andrés M Acosta-Moreno
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Lara Del Campo
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
- Departamento de Biología Celular, Facultad de Odontología, Universidad Complutense de Madrid, Madrid, Spain
| | - María J Gómez-Sánchez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
- Department of Immunology, School of Medicine, Complutense University of Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Marta Iborra
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Beatriz Herrero-Fernández
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - José M González-Granado
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Sánchez-Madrid
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, Madrid, Spain
| | - Yolanda R Carrasco
- Department of Immunology & Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Patricia Boya
- Department of Neuroscience, University of Fribourg, Fribourg, Switzerland
| | | | - Esteban Veiga
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| |
Collapse
|
2
|
The Evolution and Future of Targeted Cancer Therapy: From Nanoparticles, Oncolytic Viruses, and Oncolytic Bacteria to the Treatment of Solid Tumors. NANOMATERIALS 2021; 11:nano11113018. [PMID: 34835785 PMCID: PMC8623458 DOI: 10.3390/nano11113018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 02/07/2023]
Abstract
While many classes of chemotherapeutic agents exist to treat solid tumors, few can generate a lasting response without substantial off-target toxicity despite significant scientific advancements and investments. In this review, the paths of development for nanoparticles, oncolytic viruses, and oncolytic bacteria over the last 20 years of research towards clinical translation and acceptance as novel cancer therapeutics are compared. Novel nanoparticle, oncolytic virus, and oncolytic bacteria therapies all start with a common goal of accomplishing therapeutic drug activity or delivery to a specific site while avoiding off-target effects, with overlapping methodology between all three modalities. Indeed, the degree of overlap is substantial enough that breakthroughs in one therapeutic could have considerable implications on the progression of the other two. Each oncotherapeutic modality has accomplished clinical translation, successfully overcoming the potential pitfalls promising therapeutics face. However, once studies enter clinical trials, the data all but disappears, leaving pre-clinical researchers largely in the dark. Overall, the creativity, flexibility, and innovation of these modalities for solid tumor treatments are greatly encouraging, and usher in a new age of pharmaceutical development.
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW Novel therapies are needed for the treatment of recurrent cervical cancer. The best chemotherapy regimen to date has a response rate of 48% with an overall survival of 17 months, with limited options for second-line chemotherapy. Immunotherapy can induce a strong immune response in cervical cancer due to retained viral antigens and is reviewed in this article. RECENT FINDINGS Current clinical trials include treatment with Listeria that elicits an immune response against the E7 oncoprotein and active vaccines against the E7 oncoprotein. Although the response rates to programmed cell death 1 (PD-1) inhibition alone have been modest, the landmark survival reported in these trials suggests the activity of these agents may not be measured by RECIST criteria. The KEYNOTE-158 trial has led to the approval of pembrolizumab in recurrent programmed cell death ligand 1 (PD-L1) positive cervical cancer. Combinations of programmed cell death 1 and anticytotoxic T-lymphocyte-associated protein 4 inhibitors (CTLA4) inhibitors have shown promising and durable activity. There is active research with new combinations of checkpoint inhibitors, as well as combinations of these drugs with chemotherapy and radiation, and other novel approaches. SUMMARY Immune therapy has broad activity in cervical cancer. Responses to immunotherapy can be dramatic and durable. Continued work to find the optimal combination and setting for immunotherapy is ongoing.
Collapse
|
4
|
D'Orazio SEF. Innate and Adaptive Immune Responses during Listeria monocytogenes Infection. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0065-2019. [PMID: 31124430 PMCID: PMC11086964 DOI: 10.1128/microbiolspec.gpp3-0065-2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Indexed: 12/15/2022] Open
Abstract
It could be argued that we understand the immune response to infection with Listeria monocytogenes better than the immunity elicited by any other bacteria. L. monocytogenes are Gram-positive bacteria that are genetically tractable and easy to cultivate in vitro, and the mouse model of intravenous (i.v.) inoculation is highly reproducible. For these reasons, immunologists frequently use the mouse model of systemic listeriosis to dissect the mechanisms used by mammalian hosts to recognize and respond to infection. This article provides an overview of what we have learned over the past few decades and is divided into three sections: "Innate Immunity" describes how the host initially detects the presence of L. monocytogenes and characterizes the soluble and cellular responses that occur during the first few days postinfection; "Adaptive Immunity" discusses the exquisitely specific T cell response that mediates complete clearance of infection and immunological memory; "Use of Attenuated Listeria as a Vaccine Vector" highlights the ways that investigators have exploited our extensive knowledge of anti-Listeria immunity to develop cancer therapeutics.
Collapse
Affiliation(s)
- Sarah E F D'Orazio
- University of Kentucky, Microbiology, Immunology & Molecular Genetics, Lexington, KY 40536-0298
| |
Collapse
|
5
|
Tintelnot J, Ufer F, Engler JB, Winkler H, Lücke K, Mittrücker HW, Friese MA. Arc/Arg3.1 defines dendritic cells and Langerhans cells with superior migratory ability independent of phenotype and ontogeny in mice. Eur J Immunol 2019; 49:724-736. [PMID: 30786014 DOI: 10.1002/eji.201847797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 12/14/2018] [Accepted: 02/18/2019] [Indexed: 01/22/2023]
Abstract
The key function of migratory dendritic cells (migDCs) is to take up antigens in peripheral tissues and migrate to draining lymph nodes (dLN) to initiate immune responses. Recently, we discovered that in the mouse immune system activity-regulated cytoskeleton associated protein/activity-regulated gene 3.1 (Arc/Arg3.1) is exclusively expressed by migDCs and is a central driver of fast inflammatory migration. However, the frequency of Arc/Arg3.1-expressing cells in different migDC subsets and Langerhans cells (LCs), their phylogenetic origin, transcription factor dependency, and functional role remain unclear. Here, we found that Arc/Arg3.1+ migDCs derived from common DC precursors and radio-resistant LCs. We detected Arc/Arg3.1+ migDCs in varying frequencies within each migDC subset and LCs. Consistently, they showed superiority in inflammatory migration. Arc/Arg3.1 expression was independent of the transcription factors Irf4 or Batf3 in vivo. In intradermal Staphylococcus aureus infection that relies on inflammatory antigen transport, Arc/Arg3.1 deletion reduced T-cell responses. By contrast, Arc/Arg3.1 deficiency did not hamper the immune response to systemic Listeria monocytogenes infection, which does not require antigen transport. Thus, Arc/Arg3.1 expression is independent of ontogeny and phenotype and although it is restricted to a small fraction within each migDC subset and LCs, Arc/Arg3.1+ migDCs are important to facilitate infectious migration.
Collapse
Affiliation(s)
- Joseph Tintelnot
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Ufer
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Hana Winkler
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Karsten Lücke
- Institut für Immunologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | | | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
6
|
Lifelong CMV infection improves immune defense in old mice by broadening the mobilized TCR repertoire against third-party infection. Proc Natl Acad Sci U S A 2018; 115:E6817-E6825. [PMID: 29967140 DOI: 10.1073/pnas.1719451115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lifelong interactions between host and the ubiquitous and persistent cytomegalovirus (CMV) have been proposed to contribute to the age-related decline in immunity. Prior work from us and others found some support for that idea, yet evidence that this led to increased vulnerability to other infections was not obtained. Moreover, evidence has accumulated that CMV infection can be beneficial to immune defense in young/adult mice and humans, dominantly via enhanced innate immunity. Here, we describe an unexpected impact of murine CMV (MCMV) upon the T cell response of old mice to Listeria monocytogenes expressing the model antigen, OVA (Lm-OVA). Single-cell sequencing of the OVA-specific CD8 T cell receptor β (TCRβ) repertoire of old mice demonstrated that old MCMV-infected mice recruited many diverse clonotypes that afforded broad and often more efficient recognition of antigenic peptide variants. This stood in contrast to old control mice, which exhibited strong narrowing and homogenization of the elicited repertoire. High-throughput sequencing of the total naïve CD8 TCRβ repertoire showed that many of these diverse OVA-specific clonotypes were present in the naïve CD8 repertoire of mice in all groups (adult, old control, and old MCMV+) yet were only recruited into the Lm-OVA response in MCMV+ old mice. These results have profound implications for our understanding of T cell immunity over a life span and suggest that our coevolution with CMV may include surprising, potentially positive impacts on adaptive heterologous immunity in late life.
Collapse
|
7
|
Basu P, Mehta A, Jain M, Gupta S, Nagarkar RV, John S, Petit R. A Randomized Phase 2 Study of ADXS11-001 Listeria monocytogenes-Listeriolysin O Immunotherapy With or Without Cisplatin in Treatment of Advanced Cervical Cancer. Int J Gynecol Cancer 2018; 28:764-772. [PMID: 29538258 PMCID: PMC5929492 DOI: 10.1097/igc.0000000000001235] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/30/2018] [Accepted: 02/07/2018] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES A global unmet medical need exists for effective treatments for persistent, recurrent, or metastatic cervical cancer, as patients have a short life expectancy. Recently, immunotherapies have shown promising survival benefits for patients with advanced forms of cancer. Axalimogene filolisbac (ADXS11-001), a Listeria monocytogenes immunotherapy with a broad effect on the immune system, is under investigation for treatment of human papillomavirus-associated cancers including cervical cancer. METHODS This phase 2 study evaluated the safety and efficacy of ADXS11-001, administered with or without cisplatin, in patients with recurrent/refractory cervical cancer following prior chemotherapy and/or radiotherapy. A total of 109 patients were treated, and 69 were evaluable for tumor response at equal to or more than 3 months postbaseline. RESULTS Median overall survival (OS) was comparable between treatment groups (ADXS11-001: 8.28 months; 95% confidence interval [CI], 5.85-10.5 months; ADXS11-001 + cisplatin: 8.78 months; 95% CI, 7.4-13.3 months). The 12- and 18-month milestone OS rates were 30.9% versus 38.9%, and 23.6% versus 25.9% for each group, respectively (34.9% and 24.8% combined). Median progression-free survival (6.10 vs 6.08 months) and the overall response rate (17.1% vs 14.7%) were similar for both groups. ADXS11-001 was generally well tolerated; adverse events were predominantly mild to moderate in severity and not related to treatment. More adverse events were reported in the combination group (429 vs 275). CONCLUSIONS These promising safety and efficacy results, including the encouraging 12-month 34.9% combined OS rate, warrant further investigation of ADXS11-001 for treatment of recurrent/refractory cervical cancer.
Collapse
Affiliation(s)
- Partha Basu
- *Chittaranjan National Cancer Institute, Kolkata; †Central India Cancer Research Institute, Nagpur; ‡Ruby Hall Clinic, Pune; §Tata Memorial Hospital, Mumbai; ∥Curie Manavata Cancer Centre, Nashik; and ¶Christian Medical College Vellore, Vellore, India; and #Advaxis, Inc, Princeton, NJ
| | - Ajay Mehta
- *Chittaranjan National Cancer Institute, Kolkata; †Central India Cancer Research Institute, Nagpur; ‡Ruby Hall Clinic, Pune; §Tata Memorial Hospital, Mumbai; ∥Curie Manavata Cancer Centre, Nashik; and ¶Christian Medical College Vellore, Vellore, India; and #Advaxis, Inc, Princeton, NJ
| | - Minish Jain
- *Chittaranjan National Cancer Institute, Kolkata; †Central India Cancer Research Institute, Nagpur; ‡Ruby Hall Clinic, Pune; §Tata Memorial Hospital, Mumbai; ∥Curie Manavata Cancer Centre, Nashik; and ¶Christian Medical College Vellore, Vellore, India; and #Advaxis, Inc, Princeton, NJ
| | - Sudeep Gupta
- *Chittaranjan National Cancer Institute, Kolkata; †Central India Cancer Research Institute, Nagpur; ‡Ruby Hall Clinic, Pune; §Tata Memorial Hospital, Mumbai; ∥Curie Manavata Cancer Centre, Nashik; and ¶Christian Medical College Vellore, Vellore, India; and #Advaxis, Inc, Princeton, NJ
| | - Rajnish V. Nagarkar
- *Chittaranjan National Cancer Institute, Kolkata; †Central India Cancer Research Institute, Nagpur; ‡Ruby Hall Clinic, Pune; §Tata Memorial Hospital, Mumbai; ∥Curie Manavata Cancer Centre, Nashik; and ¶Christian Medical College Vellore, Vellore, India; and #Advaxis, Inc, Princeton, NJ
| | - Subhashini John
- *Chittaranjan National Cancer Institute, Kolkata; †Central India Cancer Research Institute, Nagpur; ‡Ruby Hall Clinic, Pune; §Tata Memorial Hospital, Mumbai; ∥Curie Manavata Cancer Centre, Nashik; and ¶Christian Medical College Vellore, Vellore, India; and #Advaxis, Inc, Princeton, NJ
| | - Robert Petit
- *Chittaranjan National Cancer Institute, Kolkata; †Central India Cancer Research Institute, Nagpur; ‡Ruby Hall Clinic, Pune; §Tata Memorial Hospital, Mumbai; ∥Curie Manavata Cancer Centre, Nashik; and ¶Christian Medical College Vellore, Vellore, India; and #Advaxis, Inc, Princeton, NJ
| |
Collapse
|
8
|
Alice AF, Kramer G, Bambina S, Baird JR, Bahjat KS, Gough MJ, Crittenden MR. Amplifying IFN-γ Signaling in Dendritic Cells by CD11c-Specific Loss of SOCS1 Increases Innate Immunity to Infection while Decreasing Adaptive Immunity. THE JOURNAL OF IMMUNOLOGY 2017; 200:177-185. [PMID: 29150567 DOI: 10.4049/jimmunol.1700909] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/23/2017] [Indexed: 01/23/2023]
Abstract
Although prophylactic vaccines provide protective humoral immunity against infectious agents, vaccines that elicit potent CD8 T cell responses are valuable tools to shape and drive cellular immunity against cancer and intracellular infection. In particular, IFN-γ-polarized cytotoxic CD8 T cell immunity is considered optimal for protective immunity against intracellular Ags. Suppressor of cytokine signaling (SOCS)1 is a cross-functional negative regulator of TLR and cytokine receptor signaling via degradation of the receptor-signaling complex. We hypothesized that loss of SOCS1 in dendritic cells (DCs) would improve T cell responses by accentuating IFN-γ-directed immune responses. We tested this hypothesis using a recombinant Listeria monocytogenes vaccine platform that targets CD11c+ DCs in mice in which SOCS1 is selectively deleted in all CD11c+ cells. Unexpectedly, in mice lacking SOCS1 expression in CD11c+ cells, we observed a decrease in CD8+ T cell response to the L. monocytogenes vaccine. NK cell responses were also decreased in mice lacking SOCS1 expression in CD11c+ cells but did not explain the defect in CD8+ T cell immunity. We found that DCs lacking SOCS1 expression were functional in driving Ag-specific CD8+ T cell expansion in vitro but that this process was defective following infection in vivo. Instead, monocyte-derived innate TNF-α and inducible NO synthase-producing DCs dominated the antibacterial response. Thus, loss of SOCS1 in CD11c+ cells skewed the balance of immune response to infection by increasing innate responses while decreasing Ag-specific adaptive responses to infectious Ags.
Collapse
Affiliation(s)
- Alejandro F Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Gwen Kramer
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Shelly Bambina
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Jason R Baird
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Keith S Bahjat
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and .,The Oregon Clinic, Portland, OR 97213
| |
Collapse
|
9
|
Miles B, Safran HP, Monk BJ. Therapeutic options for treatment of human papillomavirus-associated cancers - novel immunologic vaccines: ADXS11-001. GYNECOLOGIC ONCOLOGY RESEARCH AND PRACTICE 2017; 4:10. [PMID: 28725449 PMCID: PMC5512733 DOI: 10.1186/s40661-017-0047-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 07/03/2017] [Indexed: 01/22/2023]
Abstract
Survival of patients with advanced, recurrent, or metastatic human papillomavirus (HPV)-associated cancer is suboptimal despite the availability of various treatment modalities. The recently developed bacterial vector Listeria monocytogenes (Lm) activates innate and adaptive immune responses and is expected to offer immunologic advantages. Axalimogene filolisbac (AXAL or ADXS11-001) is a novel immunotherapeutic based on the live, irreversibly attenuated Lm fused to the nonhemolytic fragment of listeriolysin O (Lm-LLO) and secretes the Lm-LLO-HPV E7 fusion protein targeting HPV-positive tumors. Herein are reported the development and recent results of various clinical trials in patients with HPV-associated cervical, head and neck, and anal cancers.
Collapse
Affiliation(s)
- Brett Miles
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | - Bradley J. Monk
- Division of Gynecologic Oncology, Arizona Oncology (US Oncology Network), University of Arizona College of Medicine, Creighton University School of Medicine at St. Joseph’s Hospital, 2222 E. Highland Ave, Suite 400, Phoenix, AZ 85016 USA
| |
Collapse
|
10
|
Miles BA, Monk BJ, Safran HP. Mechanistic insights into ADXS11-001 human papillomavirus-associated cancer immunotherapy. GYNECOLOGIC ONCOLOGY RESEARCH AND PRACTICE 2017; 4:9. [PMID: 28588899 PMCID: PMC5455112 DOI: 10.1186/s40661-017-0046-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/20/2017] [Indexed: 12/31/2022]
Abstract
Immune responses to the facultative intracellular bacterium Listeria monocytogenes (Lm) are robust and well characterized. Utilized for decades as a model of host-disease immunology, Lm is well suited for use as an immunotherapeutic bacterial vector for the delivery of foreign antigen. Genetic modification of Lm has been undertaken to create an attenuated organism that is deficient in its master transcriptional regulator, protein-related factor A, and incorporates a truncated, nonhemolytic version of the listeriolysin O (LLO) molecule to ensure its adjuvant properties while also preventing escape of the live organism from the phagolysosome. Delivery of a vaccine construct (Lm-LLO-E7; axalimogene filolisbac [AXAL] or ADXS11-001) in which the modified LLO molecule is fused with the E7 oncoprotein of human papillomavirus type 16 (HPV-16) consistently stimulates strong innate and E7 antigen-specific adaptive immune responses, resulting in reduction of tumor burden in animal cancer models. In the clinical setting, AXAL has shown early promise in phase I/II trials of women with cervical cancer, and several more trials are currently underway to assess the efficacy and safety of this antitumor vaccine in patients with HPV-positive head and neck and anal cancers.
Collapse
Affiliation(s)
- Brett A Miles
- Division of Head and Neck Cancer Surgery, Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Bradley J Monk
- Division of Gynecologic Oncology, University of Arizona College of Medicine, Creighton University School of Medicine at Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, AZ USA
| | - Howard P Safran
- Brown University Oncology Research Group, Providence, RI USA
| |
Collapse
|
11
|
Pakalniškytė D, Schraml BU. Tissue-Specific Diversity and Functions of Conventional Dendritic Cells. Adv Immunol 2017; 134:89-135. [DOI: 10.1016/bs.ai.2017.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
12
|
Li Y, Wang Y, Zou L, Tang X, Yang Y, Ma L, Jia Q, Ni Q, Liu S, Tang L, Lin R, Wong E, Sun W, Wang L, Wei Q, Ran H, Zhang L, Lian H, Huang W, Wu Y, Li QJ, Wan Y. Analysis of the Rab GTPase Interactome in Dendritic Cells Reveals Anti-microbial Functions of the Rab32 Complex in Bacterial Containment. Immunity 2016; 44:422-37. [PMID: 26885862 DOI: 10.1016/j.immuni.2016.01.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/28/2015] [Accepted: 11/17/2015] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) orchestrate complex membrane trafficking through an interconnected transportation network linked together by Rab GTPases. Through a tandem affinity purification strategy and mass spectrometry, we depicted an interactomic landscape of major members of the mammalian Rab GTPase family. When complemented with imaging tools, this proteomic analysis provided a global view of intracellular membrane organization. Driven by this analysis, we investigated dynamic changes to the Rab32 subnetwork in DCs induced by L. monocytogenes infection and uncovered an essential role of this subnetwork in controlling the intracellular proliferation of L. monocytogenes. Mechanistically, Rab32 formed a persistent complex with two interacting proteins, PHB and PHB2, to encompass bacteria both during early phagosome formation and after L. monocytogenes escaped the original containment vacuole. Collectively, we have provided a functional compartmentalization overview and an organizational framework of intracellular Rab-mediated vesicle trafficking that can serve as a resource for future investigations.
Collapse
Affiliation(s)
- Yuanyuan Li
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Yu Wang
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Liyun Zou
- Department of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Xiangyu Tang
- Institute of Medical Equipment, Academy of Military Medical Sciences, Tianjin 300000 China
| | - Yi Yang
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Li Ma
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Qingzhu Jia
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Qingshan Ni
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Siqi Liu
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lizhang Tang
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Regina Lin
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Elizabeth Wong
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Wei Sun
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Quanfang Wei
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Haiying Ran
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Liqun Zhang
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Hengning Lian
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Wei Huang
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Yuzhang Wu
- Department of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Qi-Jing Li
- Chongqing Key Laboratory of Cytomics, Chongqing 400038, China; Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Ying Wan
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China; Chongqing Key Laboratory of Cytomics, Chongqing 400038, China.
| |
Collapse
|
13
|
Alexandre YO, Ghilas S, Sanchez C, Le Bon A, Crozat K, Dalod M. XCR1+ dendritic cells promote memory CD8+ T cell recall upon secondary infections with Listeria monocytogenes or certain viruses. J Exp Med 2015; 213:75-92. [PMID: 26694969 PMCID: PMC4710197 DOI: 10.1084/jem.20142350] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 11/20/2015] [Indexed: 12/30/2022] Open
Abstract
Alexandre et al. demonstrate the XCR1+ DCs are instrumental in memory CD8+ T cell responses to Listeria, VSV or vaccinia virus infection, but not CMV. Depending on the infection, robust memory CTL responses require cytokine- and chemokine-dependent cross-talk between XCR1+ DCs and NK cells or other IFN-γ–producing lymphocytes. Naive CD8+ T cell priming during tumor development or many primary infections requires cross-presentation by XCR1+ dendritic cells (DCs). Memory CD8+ T lymphocytes (mCTLs) harbor a lower activation threshold as compared with naive cells. However, whether their recall responses depend on XCR1+ DCs is unknown. By using a new mouse model allowing fluorescent tracking and conditional depletion of XCR1+ DCs, we demonstrate a differential requirement of these cells for mCTL recall during secondary infections by different pathogens. XCR1+ DCs were instrumental to promote this function upon secondary challenges with Listeria monocytogenes, vesicular stomatitis virus, or Vaccinia virus, but dispensable in the case of mouse cytomegalovirus. We deciphered how XCR1+ DCs promote mCTL recall upon secondary infections with Listeria. By visualizing for the first time the in vivo choreography of XCR1+ DCs, NK cells and mCTLs during secondary immune responses, and by neutralizing in vivo candidate molecules, we demonstrate that, very early after infection, mCTLs are activated, and attracted in a CXCR3-dependent manner, by NK cell–boosted, IL-12–, and CXCL9-producing XCR1+ DCs. Hence, depending on the infectious agent, strong recall of mCTLs during secondary challenges can require cytokine- and chemokine-dependent cross-talk with XCR1+ DCs and NK cells.
Collapse
Affiliation(s)
- Yannick O Alexandre
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| | - Sonia Ghilas
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| | - Cindy Sanchez
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| | - Agnès Le Bon
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale, U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Karine Crozat
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| | - Marc Dalod
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| |
Collapse
|
14
|
Yang X, Zhang X, Sun Y, Tu T, Fu ML, Miller M, Fu YX. A BTLA-mediated bait and switch strategy permits Listeria expansion in CD8α(+) DCs to promote long-term T cell responses. Cell Host Microbe 2015; 16:68-80. [PMID: 25011109 DOI: 10.1016/j.chom.2014.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 02/09/2014] [Accepted: 05/30/2014] [Indexed: 12/31/2022]
Abstract
Listeria monocytogenes infected CD8α(+) DCs in the spleen are essential for CD8(+) T cell generation. CD8α(+) DCs are also necessary for Listeria expansion and dissemination within the host. The mechanisms that regulate CD8α(+) DCs to allow Listeria expansion are unclear. We find that activating the B and T lymphocyte attenuator (BTLA), a coinhibitory receptor for T cells, suppresses, while blocking BTLA enhances, both the primary and memory CD8 T cell responses against Listeria. Btla(-/-) mice have lower effector and memory CD8(+) T cells while paradoxically also being more resistant to Listeria. Although bacterial entry into Btla(-/-) CD8α(+) DCs is unaffected, Listeria fails to expand within these cells. BTLA signaling limits Fas/FasL-mediated suppression of Listeria expansion within CD8α(+) DCs to more effectively alert adaptive immune cells. This study uncovers a BTLA-mediated strategy used by the host that permits Listeria proliferation to enable increasing T cell responses for long-term protection.
Collapse
Affiliation(s)
- Xuanming Yang
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Xunmin Zhang
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA; International joint Cancer Institute, The Second Military Medical University, Shanghai, 200433, China
| | - Yonglian Sun
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Tony Tu
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - May Lynne Fu
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Mendy Miller
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.
| | - Yang-Xin Fu
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
15
|
Seillet C, Belz GT, Huntington ND. Development, Homeostasis, and Heterogeneity of NK Cells and ILC1. Curr Top Microbiol Immunol 2015; 395:37-61. [PMID: 26305047 DOI: 10.1007/82_2015_474] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Natural killer (NK) cells are a population of cytotoxic innate lymphocytes that evolved prior to their adaptive counterparts and constitute one of the first lines of defense against infected or mutated cells. NK cells are rapidly activated, expressing an array of germ-line encoded receptors that allow them to scan for protein irregularities on cells and kill those deemed "altered-self." NK cells rapidly produce a broad range of cytokines and chemokines following activation by virus, bacterial, or parasitic infection and are thus key in orchestrating inflammation. NK cells have previously been viewed to represent a relatively homogeneous group of IFN-γ-producing cells that express the surface markers NK1.1 and natural killer cell p46-related protein (NKp46 or NCR1 encoded by Ncr1) and depend on the transcription factor T-bet for their development. Recently, a second subset of T-bet-dependent innate cells, the group 1 innate lymphoid cells (ILC1), has been discovered which share many attributes of conventional NK (cNK) cells. Despite the similarities between ILC1 and cNK cells , they differ in several important aspects including their localization, transcriptional regulation, and phenotype suggesting each subset has distinct origins and functions in immune responses. Previously, the ability to detect and spontaneously kill cells that exhibit "altered-self" which is central to tumor and viral immunity has been thought to be an attribute restricted solely to cNK cells. The identification of ILC1 challenges this notion and suggests that key contributions from ILC1 may have gone unrecognized. Thus, understanding the different rules that govern the behavior of ILC1 and cNK cells in immune responses may potentially open unexpected doorways to uncover novel strategies to manipulate these cells in treating disease. Here, we review recent advances in our understanding of peripheral cNK cell and ILC1 heterogeneity in terms of their development, phenotype, homeostasis, and effector functions.
Collapse
Affiliation(s)
- Cyril Seillet
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Gabrielle T Belz
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
16
|
Chandrabos C, M'Homa Soudja S, Weinrick B, Gros M, Frangaj A, Rahmoun M, Jacobs WR, Lauvau G. The p60 and NamA autolysins from Listeria monocytogenes contribute to host colonization and induction of protective memory. Cell Microbiol 2014; 17:147-63. [PMID: 25225110 PMCID: PMC4457399 DOI: 10.1111/cmi.12362] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 09/01/2014] [Accepted: 09/08/2014] [Indexed: 12/14/2022]
Abstract
Inducing long-term protective memory CD8(+) T-cells is a desirable goal for vaccines against intracellular pathogens. However, the mechanisms of differentiation of CD8(+) T-cells into long-lived memory cells capable of mediating protection of immunized hosts remain incompletely understood. We have developed an experimental system using mice immunized with wild type (WT) or mutants of the intracellular bacterium Listeria monocytogenes (Lm) that either do or do not develop protective memory CD8(+) T-cells. We previously reported that mice immunized with Lm lacking functional SecA2, an auxiliary secretion system of gram-positive bacteria, did not differentiate functional memory CD8(+) T-cells that protected against a challenge infection with WT Lm. Herein we hypothesized that the p60 and NamA autolysins of Lm, which are major substrates of the SecA2 pathway, account for this phenotype. We generated Lm genetically deficient for genes encoding for the p60 and NamA proteins, ΔiapΔmurA Lm, and further characterized this mutant. Δp60ΔNamA Lm exhibited a strong filamentous phenotype, inefficiently colonized host tissues, and grew mostly outside cells. When Δp60ΔNamA Lm was made single unit, cell invasion was restored to WT levels during vaccination, yet induced memory T-cells still did not protect immunized hosts against recall infection. Recruitment of blood phagocytes and antigen-presenting cell activation was close to that of mice immunized with ΔActA Lm, which develop protective memory. However, key inflammatory factors involved in optimal T-cell programming such as IL-12 and type I IFN (IFN-I) were lacking, suggesting that cytokine signals may largely account for the observed phenotype. Thus, altogether, these results establish that p60 and NamA secreted by Lm promote primary host cell invasion, the inflammatory response and the differentiation of functional memory CD8(+) T-cells, by preventing Lm filamentation during growth and subsequent triggering of innate sensing mechanisms.
Collapse
Affiliation(s)
- Ceena Chandrabos
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Arnold-Schrauf C, Dudek M, Dielmann A, Pace L, Swallow M, Kruse F, Kühl AA, Holzmann B, Berod L, Sparwasser T. Dendritic cells coordinate innate immunity via MyD88 signaling to control Listeria monocytogenes infection. Cell Rep 2014; 6:698-708. [PMID: 24529704 DOI: 10.1016/j.celrep.2014.01.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/19/2013] [Accepted: 01/17/2014] [Indexed: 12/15/2022] Open
Abstract
Listeria monocytogenes (LM), a facultative intracellular Gram-positive pathogen, can cause life-threatening infections in humans. In mice, the signaling cascade downstream of the myeloid differentiation factor 88 (MyD88) is essential for proper innate immune activation against LM, as MyD88-deficient mice succumb early to infection. Here, we show that MyD88 signaling in dendritic cells (DCs) is sufficient to mediate the protective innate response, including the production of proinflammatory cytokines, neutrophil infiltration, bacterial clearance, and full protection from lethal infection. We also demonstrate that MyD88 signaling by DCs controls the infection rates of CD8α(+) cDCs and thus limits the spread of LM to the T cell areas. Furthermore, in mice expressing MyD88 in DCs, inflammatory monocytes, which are required for bacterial clearance, are activated independently of intrinsic MyD88 signaling. In conclusion, CD11c(+) conventional DCs critically integrate pathogen-derived signals via MyD88 signaling during early infection with LM in vivo.
Collapse
Affiliation(s)
- Catharina Arnold-Schrauf
- Institute for Infection Immunology, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI), 30625 Hannover, Germany
| | - Markus Dudek
- Institute for Infection Immunology, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI), 30625 Hannover, Germany
| | - Anastasia Dielmann
- Institute for Infection Immunology, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI), 30625 Hannover, Germany
| | - Luigia Pace
- Institut National de la Santé et de la Recherche Médicale (INSERM) U932, Institut Curie, 75005 Paris, France
| | - Maxine Swallow
- Institute for Infection Immunology, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI), 30625 Hannover, Germany
| | - Friederike Kruse
- Institute for Infection Immunology, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI), 30625 Hannover, Germany
| | - Anja A Kühl
- Department of Medicine I for Gastroenterology, Infectious Disease and Rheumatology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Bernhard Holzmann
- Chirurgische Klinik und Poliklinik, Technische Universität München, 81675 Munich, Germany
| | - Luciana Berod
- Institute for Infection Immunology, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI), 30625 Hannover, Germany
| | - Tim Sparwasser
- Institute for Infection Immunology, TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI), 30625 Hannover, Germany.
| |
Collapse
|
18
|
Opata MM, Stephens R. Early Decision: Effector and Effector Memory T Cell Differentiation in Chronic Infection. ACTA ACUST UNITED AC 2014; 9:190-206. [PMID: 24790593 PMCID: PMC4000274 DOI: 10.2174/1573395509666131126231209] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/08/2013] [Accepted: 11/19/2013] [Indexed: 11/22/2022]
Abstract
As effector memory T cells (Tem) are the predominant population elicited by chronic parasitic infections,
increasing our knowledge of their function, survival and derivation, as phenotypically and functionally distinct from
central memory and effector T cells will be critical to vaccine development for these diseases. In some infections, memory
T cells maintain increased effector functions, however; this may require the presence of continued antigen, which can also
lead to T cell exhaustion. Alternatively, in the absence of antigen, only the increase in the number of memory cells
remains, without enhanced functionality as central memory. In order to understand the requirement for antigen and the
potential for longevity or protection, the derivation of each type of memory must be understood. A thorough review of the
data establishes the existence of both memory (Tmem) precursors and effector T cells (Teff) from the first hours of an
immune response. This suggests a new paradigm of Tmem differentiation distinct from the proposition that Tmem only
appear after the contraction of Teff. Several signals have been shown to be important in the generation of memory T cells,
such as the integrated strength of “signals 1-3” of antigen presentation (antigen receptor, co-stimulation, cytokines) as
perceived by each T cell clone. Given that these signals integrated at antigen presentation cells have been shown to
determine the outcome of Teff and Tmem phenotypes and numbers, this decision must be made at a very early stage. It
would appear that the overwhelming expansion of effector T cells and the inability to phenotypically distinguish memory
T cells at early time points has masked this important decision point. This does not rule out an effect of repeated
stimulation or chronic inflammatory milieu on populations generated in these early stages. Recent studies suggest that
Tmem are derived from early Teff, and we suggest that this includes Tem as well as Tcm. Therefore, we propose a
testable model for the pathway of differentiation from naïve to memory that suggests that Tem are not fully differentiated
effector cells, but derived from central memory T cells as originally suggested by Sallusto et al. in 1999, but much
debated since.
Collapse
Affiliation(s)
- Michael M Opata
- University of Texas Medical Branch, Department of Internal Medicine, Division of Infectious Disease, 300 University Avenue, Galveston, TX 77555-0435, USA
| | - Robin Stephens
- University of Texas Medical Branch, Department of Internal Medicine, Division of Infectious Disease, 300 University Avenue, Galveston, TX 77555-0435, USA
| |
Collapse
|
19
|
Rothman J, Paterson Y. Live-attenuatedListeria-based immunotherapy. Expert Rev Vaccines 2014; 12:493-504. [DOI: 10.1586/erv.13.34] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Li G, Smithey MJ, Rudd BD, Nikolich‐Žugich J. Age-associated alterations in CD8α+ dendritic cells impair CD8 T-cell expansion in response to an intracellular bacterium. Aging Cell 2012; 11:968-77. [PMID: 22862959 PMCID: PMC3533767 DOI: 10.1111/j.1474-9726.2012.00867.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Age-associated decline in immunity to infection has been documented across multiple pathogens, yet the relative contributions of the aged priming environment and of lymphocyte-intrinsic defects remain unclear. To address the impact of the aging environment on T-cell priming, adult naïve OT-I TCR transgenic CD8 T cells, specific for the H-2Kb-restricted immunodominant OVA257-264 epitope, were transferred into adult or old recipient mice infected with the recombinant intracellular bacterium Listeria monocytogenes carrying the chicken ovalbumin protein (Lm-OVA). We consistently found that adult OT-I CD8 expansion was reduced in aged recipient mice, and this correlated with numeric, phenotypic, and functional defects selectively affecting CD8α+ dendritic cells (DC). Following Lm-OVA infection, aged mice failed to accumulate CD8α+ DC in the spleen, and these cells expressed much lower levels of critical costimulatory molecules in the first three days following infection. Further, aged CD8α+ DC showed impaired uptake of the bacteria at very early time points following infection. Treatment of aged mice with Flt3 ligand (Flt3L) improved the number of DC present in the spleen prior to Lm-OVA infection, and improved, but did not reconstitute, OT-I expansion to Lm-OVA infection. These results suggest that age-associated changes in antigen uptake, pathogen sensing, and/or antigen presentation contribute to impaired adaptive immune responses to microbial pathogens with aging.
Collapse
Affiliation(s)
| | | | - Brian D. Rudd
- Department of Immunobiology and the Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | | |
Collapse
|
21
|
Pontiroli F, Dussurget O, Zanoni I, Urbano M, Beretta O, Granucci F, Ricciardi-Castagnoli P, Cossart P, Foti M. The timing of IFNβ production affects early innate responses to Listeria monocytogenes and determines the overall outcome of lethal infection. PLoS One 2012; 7:e43455. [PMID: 22912878 PMCID: PMC3422257 DOI: 10.1371/journal.pone.0043455] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/25/2012] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) and natural killer (NK) cells are essential components of the innate immunity and play a crucial role in the first phase of host defense against infections and tumors. Listeria monocytogenes (Lm) is an intracellular pathogen that colonizes the cytosol of eukaryotic cells. Recent findings have shown Lm specifically in splenic CD8a+ DCs shortly after intravenous infection. We examined gene expression profiles of mouse DCs exposed to Lm to elucidate the molecular mechanisms underlying DCs interaction with Lm. Using a functional genomics approach, we found that Lm infection induced a cluster of late response genes including type I IFNs and interferon responsive genes (IRGs) in DCs. Type I INFs were produced at the maximal level only at 24 h post infection indicating that the regulation of IFNs in the context of Lm infection is delayed compared to the rapid response observed with viral pathogens. We showed that during Lm infection, IFNγ production and cytotoxic activity were severely impaired in NK cells compared to E. coli infection. These defects were restored by providing an exogenous source of IFNβ during the initial phase of bacterial challenge. Moreover, when treated with IFNβ during early infection, NK cells were able to reduce bacterial titer in the spleen and significantly improve survival of infected mice. These findings show that the timing of IFNβ production is fundamental to the efficient control of the bacterium during the early innate phase of Lm infection.
Collapse
Affiliation(s)
- Francesca Pontiroli
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | - Olivier Dussurget
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Paris, France
- Inserm U604, Paris, France
- INRA USC2020, Paris, France
| | - Ivan Zanoni
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | - Matteo Urbano
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | - Ottavio Beretta
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
- Genopolis Consortium, University of Milano-Bicocca, Milan, Italy
| | - Francesca Granucci
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | | | - Pascale Cossart
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Paris, France
- Inserm U604, Paris, France
- INRA USC2020, Paris, France
| | - Maria Foti
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
- Genopolis Consortium, University of Milano-Bicocca, Milan, Italy
- * E-mail:
| |
Collapse
|
22
|
Nikolich-Žugich J, Li G, Uhrlaub JL, Renkema KR, Smithey MJ. Age-related changes in CD8 T cell homeostasis and immunity to infection. Semin Immunol 2012; 24:356-64. [PMID: 22554418 DOI: 10.1016/j.smim.2012.04.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 04/13/2012] [Indexed: 01/10/2023]
Abstract
Studies of CD8 T cell responses to vaccination or infection with various pathogens in both animal models and human subjects have revealed a markedly consistent array of age-related defects. In general, recent work shows that aged CD8 T cell responses are decreased in magnitude, and show poor differentiation into effector cells, with a reduced arsenal of effector functions. Here we review potential mechanisms underlying these defects. We specifically address phenotypic and numeric changes to the naïve CD8 T cell precursor pool, the impact of persistent viral infection(s) and inflammation, and contributions of the aging environment in which these cells are activated.
Collapse
Affiliation(s)
- Janko Nikolich-Žugich
- Department of Immunobiology and the Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ 85724, United States.
| | | | | | | | | |
Collapse
|
23
|
Lm-LLO-Based Immunotherapies and HPV-Associated Disease. JOURNAL OF ONCOLOGY 2012; 2012:542851. [PMID: 22481930 PMCID: PMC3307007 DOI: 10.1155/2012/542851] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 10/09/2011] [Indexed: 01/06/2023]
Abstract
HPV infection is a direct cause of neoplasia and malignancy. Cellular immunologic activity against cells expressing HPV E6 and E7 is sufficient to eliminate the presence of dysplastic or neoplastic tissue driven by HPV infection. Live attenuated Listeria monocytogenes- (Lm-) based immunotherapy (ADXS11-001) has been developed for the treatment of HPV-associated diseases. ADXS11-001 secretes an antigen-adjuvant fusion (Lm-LLO) protein consisting of a truncated fragment of the Lm protein listeriolysin O (LLO) fused to HPV-16 E7. In preclinical models, this construct has been found to stimulate immune responses and affect therapeutic outcome. ADXS11-001 is currently being evaluated in Phase 2 clinical trials for cervical intraepithelial neoplasia, cervical cancer, and HPV-positive head and neck cancer. The use of a live attenuated bacterium is a more complex and complete method of cancer immunotherapy, as over millennia Lm has evolved to infect humans and humans have evolved to prevent and reject this infection over millennia. This evolution has resulted in profound pathogen-associated immune mechanisms which are genetically conserved, highly efficacious, resistant to tolerance, and can be uniquely invoked using this novel platform technology.
Collapse
|
24
|
Abstract
Dendritic cells (DCs) represent a unique collection of innate immune cells present throughout the body as distinct subpopulations generally sharing the functions of pathogen recognition, cytokine production, and antigen presentation. A large body of work in recent years has examined DC functions during infection with Listeria monocytogenes (Lm), particularly in the murine model. Here, I review several aspects of DC biology in this model, with particular emphasis on the role DCs play in the establishment of a productive Lm infection and the role of DCs as cytokine producers and antigen-presenting cells in this system.
Collapse
Affiliation(s)
- Brian T Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
25
|
Edelson BT, Bradstreet TR, Hildner K, Carrero JA, Frederick KE, KC W, Belizaire R, Aoshi T, Schreiber RD, Miller MJ, Murphy TL, Unanue ER, Murphy KM. CD8α(+) dendritic cells are an obligate cellular entry point for productive infection by Listeria monocytogenes. Immunity 2011; 35:236-48. [PMID: 21867927 PMCID: PMC3172670 DOI: 10.1016/j.immuni.2011.06.012] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 05/26/2011] [Accepted: 06/15/2011] [Indexed: 11/28/2022]
Abstract
CD8α(+) dendritic cells (DCs) prime cytotoxic T lymphocytes during viral infections and produce interleukin-12 in response to pathogens. Although the loss of CD8α(+) DCs in Batf3(-/-) mice increases their susceptibility to several pathogens, we observed that Batf3(-/-) mice exhibited enhanced resistance to the intracellular bacterium Listeria monocytogenes. In wild-type mice, Listeria organisms, initially located in the splenic marginal zone, migrated to the periarteriolar lymphoid sheath (PALS) where they grew exponentially and induced widespread lymphocyte apoptosis. In Batf3(-/-) mice, however, Listeria organisms remain trapped in the marginal zone, failed to traffic into the PALS, and were rapidly cleared by phagocytes. In addition, Batf3(-/-) mice, which lacked the normal population of hepatic CD103(+) peripheral DCs, also showed protection from liver infection. These results suggest that Batf3-dependent CD8α(+) and CD103(+) DCs provide initial cellular entry points within the reticuloendothelial system by which Listeria establishes productive infection.
Collapse
Affiliation(s)
- Brian T Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|