Evaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapia

ilustraciones, diagramas

Autores:
Alfaro Marenco, María Alejandra
Tipo de recurso:
Fecha de publicación:
2024
Institución:
Universidad Nacional de Colombia
Repositorio:
Universidad Nacional de Colombia
Idioma:
OAI Identifier:
oai:repositorio.unal.edu.co:unal/86203
Acceso en línea:
https://repositorio.unal.edu.co/handle/unal/86203
https://repositorio.unal.edu.co/
Palabra clave:
610 - Medicina y salud::616 - Enfermedades
610 - Medicina y salud::612 - Fisiología humana
Inmunoterapia/métodos
Vacunas contra el Cáncer
Antígenos/uso terapéutico
Immunotherapy/methods
Cancer Vaccines
Antigens/therapeutic use
Linfocitos T CD8+
Muerte celular inmunogénica
Citometría de flujo
Células dendríticas derivadas de monocitos
Formulaciones del antígeno
Antígenos modelo
Monocyte-derived dendritic cells
Antigen formulations
Model antigens
CD8+ T cells
Tumor cell lines
immunogenic cell death
Flow cytometry
Rights
openAccess
License
Atribución-NoComercial-SinDerivadas 4.0 Internacional
id UNACIONAL2_49b2ea00ab94bf624f9bae99e5443d7a
oai_identifier_str oai:repositorio.unal.edu.co:unal/86203
network_acronym_str UNACIONAL2
network_name_str Universidad Nacional de Colombia
repository_id_str
dc.title.spa.fl_str_mv Evaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapia
dc.title.translated.eng.fl_str_mv Assessment of the immunogenicity of dendritic cells pulsed with different antigen formulations for immunotherapy purposes
title Evaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapia
spellingShingle Evaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapia
610 - Medicina y salud::616 - Enfermedades
610 - Medicina y salud::612 - Fisiología humana
Inmunoterapia/métodos
Vacunas contra el Cáncer
Antígenos/uso terapéutico
Immunotherapy/methods
Cancer Vaccines
Antigens/therapeutic use
Linfocitos T CD8+
Muerte celular inmunogénica
Citometría de flujo
Células dendríticas derivadas de monocitos
Formulaciones del antígeno
Antígenos modelo
Monocyte-derived dendritic cells
Antigen formulations
Model antigens
CD8+ T cells
Tumor cell lines
immunogenic cell death
Flow cytometry
title_short Evaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapia
title_full Evaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapia
title_fullStr Evaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapia
title_full_unstemmed Evaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapia
title_sort Evaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapia
dc.creator.fl_str_mv Alfaro Marenco, María Alejandra
dc.contributor.advisor.spa.fl_str_mv Bernal Estévez, David Andres
Parra López, Carlos Alberto
dc.contributor.author.spa.fl_str_mv Alfaro Marenco, María Alejandra
dc.subject.ddc.spa.fl_str_mv 610 - Medicina y salud::616 - Enfermedades
610 - Medicina y salud::612 - Fisiología humana
topic 610 - Medicina y salud::616 - Enfermedades
610 - Medicina y salud::612 - Fisiología humana
Inmunoterapia/métodos
Vacunas contra el Cáncer
Antígenos/uso terapéutico
Immunotherapy/methods
Cancer Vaccines
Antigens/therapeutic use
Linfocitos T CD8+
Muerte celular inmunogénica
Citometría de flujo
Células dendríticas derivadas de monocitos
Formulaciones del antígeno
Antígenos modelo
Monocyte-derived dendritic cells
Antigen formulations
Model antigens
CD8+ T cells
Tumor cell lines
immunogenic cell death
Flow cytometry
dc.subject.decs.spa.fl_str_mv Inmunoterapia/métodos
Vacunas contra el Cáncer
Antígenos/uso terapéutico
dc.subject.decs.eng.fl_str_mv Immunotherapy/methods
Cancer Vaccines
Antigens/therapeutic use
dc.subject.proposal.spa.fl_str_mv Linfocitos T CD8+
Muerte celular inmunogénica
Citometría de flujo
Células dendríticas derivadas de monocitos
Formulaciones del antígeno
Antígenos modelo
dc.subject.proposal.eng.fl_str_mv Monocyte-derived dendritic cells
Antigen formulations
Model antigens
CD8+ T cells
Tumor cell lines
immunogenic cell death
Flow cytometry
description ilustraciones, diagramas
publishDate 2024
dc.date.accessioned.none.fl_str_mv 2024-06-04T21:14:14Z
dc.date.available.none.fl_str_mv 2024-06-04T21:14:14Z
dc.date.issued.none.fl_str_mv 2024
dc.type.spa.fl_str_mv Trabajo de grado - Maestría
dc.type.driver.spa.fl_str_mv info:eu-repo/semantics/masterThesis
dc.type.version.spa.fl_str_mv info:eu-repo/semantics/acceptedVersion
dc.type.content.spa.fl_str_mv Text
dc.type.redcol.spa.fl_str_mv http://purl.org/redcol/resource_type/TM
status_str acceptedVersion
dc.identifier.uri.none.fl_str_mv https://repositorio.unal.edu.co/handle/unal/86203
dc.identifier.instname.spa.fl_str_mv Universidad Nacional de Colombia
dc.identifier.reponame.spa.fl_str_mv Repositorio Institucional Universidad Nacional de Colombia
dc.identifier.repourl.spa.fl_str_mv https://repositorio.unal.edu.co/
url https://repositorio.unal.edu.co/handle/unal/86203
https://repositorio.unal.edu.co/
identifier_str_mv Universidad Nacional de Colombia
Repositorio Institucional Universidad Nacional de Colombia
dc.relation.references.spa.fl_str_mv Gierlich P, Lex V, Technau A, Keupp A, Morper L, Glunz A, et al. Prostaglandin E(2) in a TLR3- and 7/8-agonist-based DC maturation cocktail generates mature, cytokine-producing, migratory DCs but impairs antigen cross-presentation to CD8(+) T cells. Cancer Immunol Immunother. 2020;69(6):1029-42.
Chamucero-Millares JA, Bernal-Estevez DA, Parra-Lopez CA. Usefulness of IL-21, IL-7, and IL- 15 conditioned media for expansion of antigen-specific CD8+ T cells from healthy donor-PBMCs suitable for immunotherapy. Cell Immunol. 2021;360:104257.
Patente TA, Pinho MP, Oliveira AA, Evangelista GCM, Bergami-Santos PC, Barbuto JAM. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front Immunol. 2018;9:3176.
Granot T, Senda T, Carpenter DJ, Matsuoka N, Weiner J, Gordon CL, et al. Dendritic Cells Display Subset and Tissue-Specific Maturation Dynamics over Human Life. Immunity. 2017;46(3):504-15.
Lamberti MJ, Nigro A, Mentucci FM, Rumie Vittar NB, Casolaro V, Dal Col J. Dendritic Cells and Immunogenic Cancer Cell Death: A Combination for Improving Antitumor Immunity. Pharmaceutics. 2020;12(3).
Bol KF, Schreibelt G, Gerritsen WR, de Vries IJ, Figdor CG. Dendritic Cell-Based Immunotherapy: State of the Art and Beyond. Clin Cancer Res. 2016;22(8):1897-906.
Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based immunotherapy. Cell Res. 2017;27(1):74-95.
Hu Z, Ott PA, Wu CJ. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat Rev Immunol. 2018;18(3):168-82.
Thomas R, Al-Khadairi G, Roelands J, Hendrickx W, Dermime S, Bedognetti D, et al. NY-ESO-1 Based Immunotherapy of Cancer: Current Perspectives. Front Immunol. 2018;9:947.
Mastelic-Gavillet B, Balint K, Boudousquie C, Gannon PO, Kandalaft LE. Personalized Dendritic Cell Vaccines-Recent Breakthroughs and Encouraging Clinical Results. Front Immunol. 2019;10:766.
Sarivalasis A, Boudousquie C, Balint K, Stevenson BJ, Gannon PO, Iancu EM, et al. A Phase I/II trial comparing autologous dendritic cell vaccine pulsed either with personalized peptides (PEP-DC) or with tumor lysate (OC-DC) in patients with advanced high-grade ovarian serous carcinoma. J Transl Med. 2019;17(1):391.
Prins RM, Wang X, Soto H, Young E, Lisiero DN, Fong B, et al. Comparison of gliomaassociated antigen peptide-loaded versus autologous tumor lysate-loaded dendritic cell vaccination in malignant glioma patients. J Immunother. 2013;36(2):152-7.
Castiello L, Sabatino M, Jin P, Clayberger C, Marincola FM, Krensky AM, et al. Monocytederived DC maturation strategies and related pathways: a transcriptional view. Cancer Immunol Immunother. 2011;60(4):457-66.
C DEW, M VDB, Hoefnagel M. Regulatory perspective on in vitro potency assays for human dendritic cells used in anti-tumor immunotherapy. Cytotherapy. 2018;20(11):1289-308.
Lee J, Breton G, Oliveira TY, Zhou YJ, Aljoufi A, Puhr S, et al. Restricted dendritic cell and monocyte progenitors in human cord blood and bone marrow. J Exp Med. 2015;212(3):385-99.
Gerlini G, Urso C, Mariotti G, Di Gennaro P, Palli D, Brandani P, et al. Plasmacytoid dendritic cells represent a major dendritic cell subset in sentinel lymph nodes of melanoma patients and accumulate in metastatic nodes. Clin Immunol. 2007;125(2):184-93.
Wylie B, Macri C, Mintern JD, Waithman J. Dendritic Cells and Cancer: From Biology to Therapeutic Intervention. Cancers (Basel). 2019;11(4).
Huysamen C, Willment JA, Dennehy KM, Brown GD. CLEC9A is a novel activation C-type lectin-like receptor expressed on BDCA3+ dendritic cells and a subset of monocytes. J Biol Chem. 2008;283(24):16693-701.
Sancho D, Joffre OP, Keller AM, Rogers NC, Martinez D, Hernanz-Falcon P, et al. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature. 2009;458(7240):899-903.
Constantino J, Gomes C, Falcao A, Neves BM, Cruz MT. Dendritic cell-based immunotherapy: a basic review and recent advances. Immunol Res. 2017;65(4):798-810.
Calmeiro J, Mendes L, Duarte IF, Leitao C, Tavares AR, Ferreira DA, et al. In-Depth Analysis of the Impact of Different Serum-Free Media on the Production of Clinical Grade Dendritic Cells for Cancer Immunotherapy. Front Immunol. 2020;11:593363.
Munz C, Dao T, Ferlazzo G, de Cos MA, Goodman K, Young JW. Mature myeloid dendritic cell subsets have distinct roles for activation and viability of circulating human natural killer cells. Blood. 2005;105(1):266-73.
Luo XL, Dalod M. The quest for faithful in vitro models of human dendritic cells types. Mol Immunol. 2020;123:40-59.
Chiang CL, Kandalaft LE, Tanyi J, Hagemann AR, Motz GT, Svoronos N, et al. A dendritic cell vaccine pulsed with autologous hypochlorous acid-oxidized ovarian cancer lysate primes effective broad antitumor immunity: from bench to bedside. Clin Cancer Res. 2013;19(17):4801-15.
Mailliard RB, Wankowicz-Kalinska A, Cai Q, Wesa A, Hilkens CM, Kapsenberg ML, et al. alphatype-1 polarized dendritic cells: a novel immunization tool with optimized CTL-inducing activity. Cancer Res. 2004;64(17):5934-7.
Lee JJ, Foon KA, Mailliard RB, Muthuswamy R, Kalinski P. Type 1-polarized dendritic cells loaded with autologous tumor are a potent immunogen against chronic lymphocytic leukemia. J Leukoc Biol. 2008;84(1):319-25.
Garris CS, Arlauckas SP, Kohler RH, Trefny MP, Garren S, Piot C, et al. Successful Anti-PD-1 Cancer Immunotherapy Requires T Cell-Dendritic Cell Crosstalk Involving the Cytokines IFN-gamma and IL-12. Immunity. 2018;49(6):1148-61 e7.
Del Prete A, Sozio F, Barbazza I, Salvi V, Tiberio L, Laffranchi M, et al. Functional Role of Dendritic Cell Subsets in Cancer Progression and Clinical Implications. Int J Mol Sci. 2020;21(11).
Cannon MJ, Schmid DS, Hyde TB. Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection. Rev Med Virol. 2010;20(4):202-13.
Terrazzini N, Kern F. Cell-mediated immunity to human CMV infection: a brief overview. F1000Prime Rep. 2014;6:28.
Schober K, Buchholz VR, Busch DH. TCR repertoire evolution during maintenance of CMVspecific T-cell populations. Immunol Rev. 2018;283(1):113-28.
O'Hara GA, Welten SP, Klenerman P, Arens R. Memory T cell inflation: understanding cause and effect. Trends Immunol. 2012;33(2):84-90.
Khan N, Best D, Bruton R, Nayak L, Rickinson AB, Moss PA. T cell recognition patterns of immunodominant cytomegalovirus antigens in primary and persistent infection. J Immunol. 2007;178(7):4455-65.
Wills MR, Carmichael AJ, Mynard K, Jin X, Weekes MP, Plachter B, et al. The human cytotoxic T-lymphocyte (CTL) response to cytomegalovirus is dominated by structural protein pp65: frequency, specificity, and T-cell receptor usage of pp65-specific CTL. J Virol. 1996;70(11):7569-79.
Pittet MJ, Zippelius A, Valmori D, Speiser DE, Cerottini JC, Romero P. Melan-A/MART-1-specific CD8 T cells: from thymus to tumor. Trends Immunol. 2002;23(7):325-8.
Coulie PG, Brichard V, Van Pel A, Wolfel T, Schneider J, Traversari C, et al. A new gene coding for a differentiation antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J Exp Med. 1994;180(1):35-42.
Pittet MJ, Valmori D, Dunbar PR, Speiser DE, Lienard D, Lejeune F, et al. High frequencies of naive Melan-A/MART-1-specific CD8(+) T cells in a large proportion of human histocompatibility leukocyte antigen (HLA)-A2 individuals. J Exp Med. 1999;190(5):705-15.
Alanio C, Lemaitre F, Law HK, Hasan M, Albert ML. Enumeration of human antigen-specific naive CD8+ T cells reveals conserved precursor frequencies. Blood. 2010;115(18):3718-25.
Valmori D, Fonteneau JF, Lizana CM, Gervois N, Lienard D, Rimoldi D, et al. Enhanced generation of specific tumor-reactive CTL in vitro by selected Melan-A/MART-1 immunodominant peptide analogues. J Immunol. 1998;160(4):1750-8.
van Elsas A, van der Burg SH, van der Minne CE, Borghi M, Mourer JS, Melief CJ, et al. Peptide-pulsed dendritic cells induce tumoricidal cytotoxic T lymphocytes from healthy donors against stably HLA-A*0201-binding peptides from the Melan-A/MART-1 self antigen. Eur J Immunol. 1996;26(8):1683-9.
Bakker AB, Marland G, de Boer AJ, Huijbens RJ, Danen EH, Adema GJ, et al. Generation of antimelanoma cytotoxic T lymphocytes from healthy donors after presentation of melanomaassociated antigen-derived epitopes by dendritic cells in vitro. Cancer Res. 1995;55(22):5330-4.
Cebon J, Knights A, Ebert L, Jackson H, Chen W. Evaluation of cellular immune responses in cancer vaccine recipients: lessons from NY-ESO-1. Expert Rev Vaccines. 2010;9(6):617-29.
Chen YT, Scanlan MJ, Sahin U, Tureci O, Gure AO, Tsang S, et al. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci U S A. 1997;94(5):1914-8.
Sabbatini P, Tsuji T, Ferran L, Ritter E, Sedrak C, Tuballes K, et al. Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients. Clin Cancer Res. 2012;18(23):6497-508.
Raza A, Merhi M, Inchakalody VP, Krishnankutty R, Relecom A, Uddin S, et al. Unleashing the immune response to NY-ESO-1 cancer testis antigen as a potential target for cancer immunotherapy. J Transl Med. 2020;18(1):140.
Slingluff CL, Jr. The present and future of peptide vaccines for cancer: single or multiple, long or short, alone or in combination? Cancer J. 2011;17(5):343-50.
Yamshchikov GV, Barnd DL, Eastham S, Galavotti H, Patterson JW, Deacon DH, et al. Evaluation of peptide vaccine immunogenicity in draining lymph nodes and peripheral blood of melanoma patients. Int J Cancer. 2001;92(5):703-11.
Rosenberg SA, Sherry RM, Morton KE, Scharfman WJ, Yang JC, Topalian SL, et al. Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma. J Immunol. 2005;175(9):6169-76.
Toes RE, Blom RJ, Offringa R, Kast WM, Melief CJ. Enhanced tumor outgrowth after peptide vaccination. Functional deletion of tumor-specific CTL induced by peptide vaccination can lead to the inability to reject tumors. J Immunol. 1996;156(10):3911-8.
Toes RE, Offringa R, Blom RJ, Melief CJ, Kast WM. Peptide vaccination can lead to enhanced tumor growth through specific T-cell tolerance induction. Proc Natl Acad Sci U S A. 1996;93(15):7855-60.
Janssen EM, Droin NM, Lemmens EE, Pinkoski MJ, Bensinger SJ, Ehst BD, et al. CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-mediated activation-induced cell death. Nature. 2005;434(7029):88-93.
Srinivasan M, Domanico SZ, Kaumaya PT, Pierce SK. Peptides of 23 residues or greater are required to stimulate a high affinity class II-restricted T cell response. Eur J Immunol. 1993;23(5):1011-6.
Zwaveling S, Ferreira Mota SC, Nouta J, Johnson M, Lipford GB, Offringa R, et al. Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J Immunol. 2002;169(1):350-8.
Ma Y, Adjemian S, Mattarollo SR, Yamazaki T, Aymeric L, Yang H, et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity. 2013;38(4):729-41.
Pathak SK, Skold AE, Mohanram V, Persson C, Johansson U, Spetz AL. Activated apoptotic cells induce dendritic cell maturation via engagement of Toll-like receptor 4 (TLR4), dendritic cellspecific intercellular adhesion molecule 3 (ICAM-3)-grabbing nonintegrin (DC-SIGN), and beta2 integrins. J Biol Chem. 2012;287(17):13731-42.
Han Q, Bagheri N, Bradshaw EM, Hafler DA, Lauffenburger DA, Love JC. Polyfunctional responses by human T cells result from sequential release of cytokines. Proc Natl Acad Sci U S A. 2012;109(5):1607-12.
de Araujo-Souza PS, Hanschke SCH, Nardy A, Secca C, Oliveira-Vieira B, Silva KL, et al. Differential interferon-gamma production by naive and memory-like CD8 T cells. J Leukoc Biol. 2020;108(4):1329-37.
Bhat P, Leggatt G, Waterhouse N, Frazer IH. Interferon-gamma derived from cytotoxic lymphocytes directly enhances their motility and cytotoxicity. Cell Death Dis. 2017;8(6):e2836.
Lachmann R, Bajwa M, Vita S, Smith H, Cheek E, Akbar A, et al. Polyfunctional T cells accumulate in large human cytomegalovirus-specific T cell responses. J Virol. 2012;86(2):1001-9.
Gubser PM, Bantug GR, Razik L, Fischer M, Dimeloe S, Hoenger G, et al. Rapid effector function of memory CD8+ T cells requires an immediate-early glycolytic switch. Nat Immunol. 2013;14(10):1064-72.
Sathaliyawala T, Kubota M, Yudanin N, Turner D, Camp P, Thome JJ, et al. Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity. 2013;38(1):187-97.
Mehta AK, Gracias DT, Croft M. TNF activity and T cells. Cytokine. 2018;101:14-8.
Brehm MA, Daniels KA, Welsh RM. Rapid production of TNF-alpha following TCR engagement of naive CD8 T cells. J Immunol. 2005;175(8):5043-9.
Harari A, Dutoit V, Cellerai C, Bart PA, Du Pasquier RA, Pantaleo G. Functional signatures of protective antiviral T-cell immunity in human virus infections. Immunol Rev. 2006;211:236-54.
Healy ZR, Weinhold KJ, Murdoch DM. Transcriptional Profiling of CD8+ CMV-Specific T Cell Functional Subsets Obtained Using a Modified Method for Isolating High-Quality RNA From Fixed and Permeabilized Cells. Front Immunol. 2020;11:1859.
Imai N, Tawara I, Yamane M, Muraoka D, Shiku H, Ikeda H. CD4(+) T cells support polyfunctionality of cytotoxic CD8(+) T cells with memory potential in immunological control of tumor. Cancer Sci. 2020;111(6):1958-68.
Reddy M, Eirikis E, Davis C, Davis HM, Prabhakar U. Comparative analysis of lymphocyte activation marker expression and cytokine secretion profile in stimulated human peripheral blood mononuclear cell cultures: an in vitro model to monitor cellular immune function. J Immunol Methods. 2004;293(1-2):127-42.
Adamczyk M, Bartosinska J, Raczkiewicz D, Kowal M, Surdacka A, Krasowska D, et al. The Expression of Activation Markers CD25 and CD69 Increases during Biologic Treatment of Psoriasis. J Clin Med. 2023;12(20).
Cebrian M, Yague E, Rincon M, Lopez-Botet M, de Landazuri MO, Sanchez-Madrid F. Triggering of T cell proliferation through AIM, an activation inducer molecule expressed on activated human lymphocytes. J Exp Med. 1988;168(5):1621-37.
Risso A, Smilovich D, Capra MC, Baldissarro I, Yan G, Bargellesi A, et al. CD69 in resting and activated T lymphocytes. Its association with a GTP binding protein and biochemical requirements for its expression. J Immunol. 1991;146(12):4105-14.
Cibrian D, Sanchez-Madrid F. CD69: from activation marker to metabolic gatekeeper. Eur J Immunol. 2017;47(6):946-53.
Ziegler SF, Ramsdell F, Alderson MR. The activation antigen CD69. Stem Cells. 1994;12(5):456-65.
Gonzalez-Amaro R, Cortes JR, Sanchez-Madrid F, Martin P. Is CD69 an effective brake to control inflammatory diseases? Trends Mol Med. 2013;19(10):625-32.
de la Fuente H, Cruz-Adalia A, Martinez Del Hoyo G, Cibrian-Vera D, Bonay P, Perez-Hernandez D, et al. The leukocyte activation receptor CD69 controls T cell differentiation through its interaction with galectin-1. Mol Cell Biol. 2014;34(13):2479-87.
Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155(3):1151-64.
Brusko TM, Wasserfall CH, Hulme MA, Cabrera R, Schatz D, Atkinson MA. Influence of membrane CD25 stability on T lymphocyte activity: implications for immunoregulation. PLoS One. 2009;4(11):e7980.
Green DR, Droin N, Pinkoski M. Activation-induced cell death in T cells. Immunol Rev. 2003;193:70-81.
Poulton TA, Gallagher A, Potts RC, Beck JS. Changes in activation markers and cell membrane receptors on human peripheral blood T lymphocytes during cell cycle progression after PHA stimulation. Immunology. 1988;64(3):419-25.
Watts TH. TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol. 2005;23:23-68.
Wolfl M, Kuball J, Eyrich M, Schlegel PG, Greenberg PD. Use of CD137 to study the full repertoire of CD8+ T cells without the need to know epitope specificities. Cytometry A. 2008;73(11):1043-9.
Wen T, Bukczynski J, Watts TH. 4-1BB ligand-mediated costimulation of human T cells induces CD4 and CD8 T cell expansion, cytokine production, and the development of cytolytic effector function. J Immunol. 2002;168(10):4897-906.
Halstead ES, Mueller YM, Altman JD, Katsikis PD. In vivo stimulation of CD137 broadens primary antiviral CD8+ T cell responses. Nat Immunol. 2002;3(6):536-41.
Vinay DS, Kwon BS. Role of 4-1BB in immune responses. Semin Immunol. 1998;10(6):481-9.
Etxeberria I, Glez-Vaz J, Teijeira A, Melero I. New emerging targets in cancer immunotherapy: CD137/4-1BB costimulatory axis. ESMO Open. 2020;4(Suppl 3):e000733.
Alosaimi MF, Hoenig M, Jaber F, Platt CD, Jones J, Wallace J, et al. Immunodeficiency and EBV-induced lymphoproliferation caused by 4-1BB deficiency. J Allergy Clin Immunol. 2019;144(2):574-83 e5.
Zhu Y, Zhu G, Luo L, Flies AS, Chen L. CD137 stimulation delivers an antigen-independent growth signal for T lymphocytes with memory phenotype. Blood. 2007;109(11):4882-9.
Melero I, Shuford WW, Newby SA, Aruffo A, Ledbetter JA, Hellstrom KE, et al. Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med. 1997;3(6):682-5.
Grewal IS, Flavell RA. CD40 and CD154 in cell-mediated immunity. Annu Rev Immunol. 1998;16:111-35.
Caux C, Massacrier C, Vanbervliet B, Dubois B, Van Kooten C, Durand I, et al. Activation of human dendritic cells through CD40 cross-linking. J Exp Med. 1994;180(4):1263-72.
Bennett SR, Carbone FR, Karamalis F, Flavell RA, Miller JF, Heath WR. Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature. 1998;393(6684):478-80.
Schoenberger SP, Toes RE, van der Voort EI, Offringa R, Melief CJ. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature. 1998;393(6684):480-3.
Tay NQ, Lee DCP, Chua YL, Prabhu N, Gascoigne NRJ, Kemeny DM. CD40L Expression Allows CD8(+) T Cells to Promote Their Own Expansion and Differentiation through Dendritic Cells. Front Immunol. 2017;8:1484.
Wong KL, Lew FC, MacAry PA, Kemeny DM. CD40L-expressing CD8 T cells prime CD8alpha(+) DC for IL-12p70 production. Eur J Immunol. 2008;38(8):2251-62.
Wong KL, Tang LF, Lew FC, Wong HS, Chua YL, MacAry PA, et al. CD44high memory CD8 T cells synergize with CpG DNA to activate dendritic cell IL-12p70 production. J Immunol. 2009;183(1):41-50.
Bourgeois C, Rocha B, Tanchot C. A role for CD40 expression on CD8+ T cells in the generation of CD8+ T cell memory. Science. 2002;297(5589):2060-3.
Hernandez MG, Shen L, Rock KL. CD40-CD40 ligand interaction between dendritic cells and CD8+ T cells is needed to stimulate maximal T cell responses in the absence of CD4+ T cell help. J Immunol. 2007;178(5):2844-52.
Shugart JA, Bambina S, Alice AF, Montler R, Bahjat KS. A self-help program for memory CD8+ T cells: positive feedback via CD40-CD40L signaling as a critical determinant of secondary expansion. PLoS One. 2013;8(5):e64878.
Olson MR, Seah SG, Edenborough K, Doherty PC, Lew AM, Turner SJ. CD154+ CD4+ T-cell dependence for effective memory influenza virus-specific CD8+ T-cell responses. Immunol Cell Biol. 2014;92(7):605-11.
Stinchcombe JC, Majorovits E, Bossi G, Fuller S, Griffiths GM. Centrosome polarization delivers secretory granules to the immunological synapse. Nature. 2006;443(7110):462-5.
Ballesteros-Tato A, Leon B, Lee BO, Lund FE, Randall TD. Epitope-specific regulation of memory programming by differential duration of antigen presentation to influenza-specific CD8(+) T cells. Immunity. 2014;41(1):127-40.
Blank CU, Haining WN, Held W, Hogan PG, Kallies A, Lugli E, et al. Defining 'T cell exhaustion'. Nat Rev Immunol. 2019;19(11):665-74.
Ahn E, Araki K, Hashimoto M, Li W, Riley JL, Cheung J, et al. Role of PD-1 during effector CD8 T cell differentiation. Proc Natl Acad Sci U S A. 2018;115(18):4749-54.
Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492-9.
Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8(3):239-45.
Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017;355(6332):1428-33.
Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol. 2005;25(21):9543- 53.
Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates Tcell proliferation and interleukin-10 secretion. Nat Med. 1999;5(12):1365-9.
Tseng SY, Otsuji M, Gorski K, Huang X, Slansky JE, Pai SI, et al. B7-DC, a new dendritic cell molecule with potent costimulatory properties for T cells. J Exp Med. 2001;193(7):839-46.
Keir ME, Freeman GJ, Sharpe AH. PD-1 regulates self-reactive CD8+ T cell responses to antigen in lymph nodes and tissues. J Immunol. 2007;179(8):5064-70.
Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455-65.
Meng X, Liu X, Guo X, Jiang S, Chen T, Hu Z, et al. FBXO38 mediates PD-1 ubiquitination and regulates anti-tumour immunity of T cells. Nature. 2018;564(7734):130-5.
Chan DV, Gibson HM, Aufiero BM, Wilson AJ, Hafner MS, Mi QS, et al. Differential CTLA-4 expression in human CD4+ versus CD8+ T cells is associated with increased NFAT1 and inhibition of CD4+ proliferation. Genes Immun. 2014;15(1):25-32.
Dariavach P, Mattei MG, Golstein P, Lefranc MP. Human Ig superfamily CTLA-4 gene: chromosomal localization and identity of protein sequence between murine and human CTLA-4 cytoplasmic domains. Eur J Immunol. 1988;18(12):1901-5.
Harper K, Balzano C, Rouvier E, Mattei MG, Luciani MF, Golstein P. CTLA-4 and CD28 activated lymphocyte molecules are closely related in both mouse and human as to sequence, message expression, gene structure, and chromosomal location. J Immunol. 1991;147(3):1037-44.
Auchincloss H, Turka LA. CTLA-4: not all costimulation is stimulatory. J Immunol. 2011;187(7):3457-8.
Lindsten T, Lee KP, Harris ES, Petryniak B, Craighead N, Reynolds PJ, et al. Characterization of CTLA-4 structure and expression on human T cells. J Immunol. 1993;151(7):3489-99.
Gajewski TF, Fallarino F, Fields PE, Rivas F, Alegre ML. Absence of CTLA-4 lowers the activation threshold of primed CD8+ TCR-transgenic T cells: lack of correlation with Src homology domain 2-containing protein tyrosine phosphatase. J Immunol. 2001;166(6):3900-7.
Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994;1(5):405-13.
Triebel F, Jitsukawa S, Baixeras E, Roman-Roman S, Genevee C, Viegas-Pequignot E, et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med. 1990;171(5):1393-405.
Workman CJ, Cauley LS, Kim IJ, Blackman MA, Woodland DL, Vignali DA. Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell population following antigen activation in vivo. J Immunol. 2004;172(9):5450-5.
Hannier S, Tournier M, Bismuth G, Triebel F. CD3/TCR complex-associated lymphocyte activation gene-3 molecules inhibit CD3/TCR signaling. J Immunol. 1998;161(8):4058-65.
Workman CJ, Dugger KJ, Vignali DA. Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J Immunol. 2002;169(10):5392-5.
Huard B, Tournier M, Hercend T, Triebel F, Faure F. Lymphocyte-activation gene 3/major histocompatibility complex class II interaction modulates the antigenic response of CD4+ T lymphocytes. Eur J Immunol. 1994;24(12):3216-21.
Goldberg MV, Drake CG. LAG-3 in Cancer Immunotherapy. Curr Top Microbiol Immunol. 2011;344:269-78.
Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10(1):29-37.
Bijker MS, van den Eeden SJ, Franken KL, Melief CJ, van der Burg SH, Offringa R. Superior induction of anti-tumor CTL immunity by extended peptide vaccines involves prolonged, DC-focused antigen presentation. Eur J Immunol. 2008;38(4):1033-42.
Gutierrez-Martinez E, Planes R, Anselmi G, Reynolds M, Menezes S, Adiko AC, et al. Cross-Presentation of Cell-Associated Antigens by MHC Class I in Dendritic Cell Subsets. Front Immunol. 2015;6:363.
Alloatti A, Kotsias F, Magalhaes JG, Amigorena S. Dendritic cell maturation and crosspresentation: timing matters! Immunol Rev. 2016;272(1):97-108.
Menager J, Ebstein F, Oger R, Hulin P, Nedellec S, Duverger E, et al. 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(2):e89897.
Tang-Huau TL, Gueguen P, Goudot C, Durand M, Bohec M, Baulande S, et al. Human in vivogenerated monocyte-derived dendritic cells and macrophages cross-present antigens through a vacuolar pathway. Nat Commun. 2018;9(1):2570.
Fonteneau JF, Kavanagh DG, Lirvall M, Sanders C, Cover TL, Bhardwaj N, et al. Characterization of the MHC class I cross-presentation pathway for cell-associated antigens by human dendritic cells. Blood. 2003;102(13):4448-55.
Du J MA, Widlund HR, Horstmann MA, Ramaswamy S, Fisher DE. MLANA/MART1 and SILV/PMEL17/GP100 are transcriptionally regulated by MITF in melanocytes and melanoma. Am J Pathol. 2003(Jul;163(1):333-43.).
Li J, Song JS, Bell RJ, Tran TN, Haq R, Liu H, et al. YY1 regulates melanocyte development and function by cooperating with MITF. PLoS Genet. 2012;8(5):e1002688.
Vidard L. 4-1BB and cytokines trigger human NK, gammadelta T, and CD8(+) T cell proliferation and activation, but are not required for their effector functions. Immun Inflamm Dis. 2023;11(1):e749.
Almunia C, Bretaudeau M, Held G, Babon A, Marchetti C, Castelli FA, et al. Bee Venom Phospholipase A2, a Good "Chauffeur" for Delivering Tumor Antigen to the MHC I and MHC II Peptide-Loading Compartments of the Dendritic Cells: The Case of NY-ESO-1. PLoS One. 2013;8(6):e67645.
Levy F, Muehlethaler K, Salvi S, Peitrequin AL, Lindholm CK, Cerottini JC, et al. Ubiquitylation of a melanosomal protein by HECT-E3 ligases serves as sorting signal for lysosomal degradation. Mol Biol Cell. 2005;16(4):1777-87.
Sabbatino F, Wang Y, Scognamiglio G, Favoino E, Feldman SA, Villani V, et al. Antitumor Activity of BRAF Inhibitor and IFNalpha Combination in BRAF-Mutant Melanoma. J Natl Cancer Inst. 2016;108(7).
Suriano R, Rajoria S, A LG, Geliebter J, Wallack M, Tiwari RK. Ex vivo derived primary melanoma cells: implications for immunotherapeutic vaccines. J Cancer. 2013;4(5):371-82.
Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 1994;179(4):1109-18.
Jonuleit H, Kuhn U, Muller G, Steinbrink K, Paragnik L, Schmitt E, et al. Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. Eur J Immunol. 1997;27(12):3135-42.
Martinuzzi E, Afonso G, Gagnerault MC, Naselli G, Mittag D, Combadiere B, et al. acDCs enhance human antigen-specific T-cell responses. Blood. 2011;118(8):2128-37.
Aspord C, Leloup C, Reche S, Plumas J. pDCs efficiently process synthetic long peptides to induce functional virus- and tumour-specific T-cell responses. Eur J Immunol. 2014;44(10):2880-92.
Wada H, Isobe M, Kakimi K, Mizote Y, Eikawa S, Sato E, et al. Vaccination with NY-ESO-1 overlapping peptides mixed with Picibanil OK-432 and montanide ISA-51 in patients with cancers expressing the NY-ESO-1 antigen. J Immunother. 2014;37(2):84-92.
Martínez-Enríquez L. Identificación y caracterización de linfocitos T neoantígeno específicos de donantes sanos con fines de inmunoterapia en cáncer: Universidad Nacional de Colombia; 2022.
Weekes MP, Wills MR, Mynard K, Carmichael AJ, Sissons JG. The memory cytotoxic Tlymphocyte (CTL) response to human cytomegalovirus infection contains individual peptide-specific CTL clones that have undergone extensive expansion in vivo. J Virol. 1999;73(3):2099-108.
Solache A, Morgan CL, Dodi AI, Morte C, Scott I, Baboonian C, et al. Identification of three HLA-A*0201-restricted cytotoxic T cell epitopes in the cytomegalovirus protein pp65 that are conserved between eight strains of the virus. J Immunol. 1999;163(10):5512-8.
Bruggner RV, Bodenmiller B, Dill DL, Tibshirani RJ, Nolan GP. Automated identification of stratifying signatures in cellular subpopulations. Proc Natl Acad Sci U S A. 2014;111(26):E2770-7.
Carmichael J, DeGraff WG, Gazdar AF, Minna JD, Mitchell JB. Evaluation of a tetrazoliumbased semiautomated colorimetric assay: assessment of chemosensitivity testing. Cancer Res. 1987;47(4):936-42.
Infante-Crúz A. Activación de respuesta inmune innata a partir de la inducción de muerte tumoral inmunogénica.: Universidad Nacional de Colombia; 2016.
Yadav B, Wennerberg K, Aittokallio T, Tang J. Searching for Drug Synergy in Complex Dose-Response Landscapes Using an Interaction Potency Model. Comput Struct Biotechnol J. 2015;13:504-13.
Altomonte M, Gloghini A, Bertola G, Gasparollo A, Carbone A, Ferrone S, et al. Differential expression of cell adhesion molecules CD54/CD11a and CD58/CD2 by human melanoma cells and functional role in their interaction with cytotoxic cells. Cancer Res. 1993;53(14):3343-8.
Bernal Estévez D. Evaluación de la capacidad inmuno-estimulante de la terapia neoadyuvante con Doxorrubicina y Ciclofosfamida en pacientes con cáncer de mama: Universidad Nacional de Colombia Sede Bogotá 2017.
Mariotti S, Nisini R. Generation of human T cell clones. Methods Mol Biol. 2009;514:65-93.
Klapper JA, Thomasian AA, Smith DM, Gorgas GC, Wunderlich JR, Smith FO, et al. Single-pass, closed-system rapid expansion of lymphocyte cultures for adoptive cell therapy. J Immunol Methods. 2009;345(1-2):90-9.
Rimoldi D, Muehlethaler K, Salvi S, Valmori D, Romero P, Cerottini JC, et al. Subcellular localization of the melanoma-associated protein Melan-AMART-1 influences the processing of its HLA-A2-restricted epitope. J Biol Chem. 2001;276(46):43189-96.
Busam KJ JA. Melan-A, a new melanocytic differentiation marker. Adv Anat Pathol. 1999(Jan;6(1):12-8.).
Blass E, Ott PA. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat Rev Clin Oncol. 2021;18(4):215-29.
Wahida A, Buschhorn L, Frohling S, Jost PJ, Schneeweiss A, Lichter P, et al. The coming decade in precision oncology: six riddles. Nat Rev Cancer. 2023;23(1):43-54.
Olivier T, Haslam A, Tuia J, Prasad V. Eligibility for Human Leukocyte Antigen-Based Therapeutics by Race and Ethnicity. JAMA Netw Open. 2023;6(10):e2338612.
Welten SPM, Baumann NS, Oxenius A. Fuel and brake of memory T cell inflation. Med Microbiol Immunol. 2019;208(3-4):329-38.
van der Leun AM, Thommen DS, Schumacher TN. CD8(+) T cell states in human cancer: insights from single-cell analysis. Nat Rev Cancer. 2020;20(4):218-32.
Rodriguez IJ, Bernal-Estevez DA, Llano-Leon M, Bonilla CE, Parra-Lopez CA. Neoadjuvant chemotherapy modulates exhaustion of T cells in breast cancer patients. PLoS One. 2023;18(2):e0280851.
Zhou LJ, Schwarting R, Smith HM, Tedder TF. A novel cell-surface molecule expressed by human interdigitating reticulum cells, Langerhans cells, and activated lymphocytes is a new member of the Ig superfamily. J Immunol. 1992;149(2):735-42.
Zhou LJ, Tedder TF. Human blood dendritic cells selectively express CD83, a member of the immunoglobulin superfamily. J Immunol. 1995;154(8):3821-35.
Lechmann M, Berchtold S, Hauber J, Steinkasserer A. CD83 on dendritic cells: more than just a marker for maturation. Trends Immunol. 2002;23(6):273-5.
Li Z, Ju X, Silveira PA, Abadir E, Hsu WH, Hart DNJ, et al. CD83: Activation Marker for Antigen Presenting Cells and Its Therapeutic Potential. Front Immunol. 2019;10:1312.
Trepiakas R, Pedersen AE, Met O, Hansen MH, Berntsen A, Svane IM. Comparison of alpha-Type-1 polarizing and standard dendritic cell cytokine cocktail for maturation of therapeutic monocyte-derived dendritic cell preparations from cancer patients. Vaccine. 2008;26(23):2824-32.
Li JG, Du YM, Yan ZD, Yan J, Zhuansun YX, Chen R, et al. CD80 and CD86 knockdown in dendritic cells regulates Th1/Th2 cytokine production in asthmatic mice. Exp Ther Med. 2016;11(3):878-84.
Dustin ML, Rothlein R, Bhan AK, Dinarello CA, Springer TA. Induction by IL 1 and interferongamma: tissue distribution, biochemistry, and function of a natural adherence molecule (ICAM-1). J Immunol. 1986;137(1):245-54.
Carrasco YR, Fleire SJ, Cameron T, Dustin ML, Batista FD. LFA-1/ICAM-1 interaction lowers the threshold of B cell activation by facilitating B cell adhesion and synapse formation. Immunity. 2004;20(5):589-99.
Sheikh NA, Jones LA. CD54 is a surrogate marker of antigen presenting cell activation. Cancer Immunol Immunother. 2008;57(9):1381-90.
Josephs TM, Grant EJ, Gras S. Molecular challenges imposed by MHC-I restricted long epitopes on T cell immunity. Biol Chem. 2017;398(9):1027-36.
Jimenez-Fernandez M, de la Fuente H, Martin P, Cibrian D, Sanchez-Madrid F. Unraveling CD69 signaling pathways, ligands and laterally associated molecules. EXCLI J. 2023;22:334-51.
Hosono M, de Boer OJ, van der Wal AC, van der Loos CM, Teeling P, Piek JJ, et al. Increased expression of T cell activation markers (CD25, CD26, CD40L and CD69) in atherectomy specimens of patients with unstable angina and acute myocardial infarction. Atherosclerosis. 2003;168(1):73-80.
Engelhard VH. Structure of peptides associated with class I and class II MHC molecules. Annu Rev Immunol. 1994;12:181-207.
Wang M, Larsen MV, Nielsen M, Harndahl M, Justesen S, Dziegiel MH, et al. HLA class I binding 9mer peptides from influenza A virus induce CD4 T cell responses. PLoS One. 2010;5(5):e10533.
Wang M, Tang ST, Stryhn A, Justesen S, Larsen MV, Dziegiel MH, et al. Identification of MHC class II restricted T-cell-mediated reactivity against MHC class I binding Mycobacterium tuberculosis peptides. Immunology. 2011;132(4):482-91.
Bioley G, Jandus C, Tuyaerts S, Rimoldi D, Kwok WW, Speiser DE, et al. Melan-A/MART-1-specific CD4 T cells in melanoma patients: identification of new epitopes and ex vivo visualization of specific T cells by MHC class II tetramers. J Immunol. 2006;177(10):6769-79.
Jandus C, Bioley G, Dojcinovic D, Derre L, Baitsch L, Wieckowski S, et al. Tumor antigenspecific FOXP3+ CD4 T cells identified in human metastatic melanoma: peptide vaccination results in selective expansion of Th1-like counterparts. Cancer Res. 2009;69(20):8085-93.
Gross S, Lennerz V, Gallerani E, Mach N, Bohm S, Hess D, et al. Short Peptide Vaccine Induces CD4+ T Helper Cells in Patients with Different Solid Cancers. Cancer Immunol Res. 2016;4(1):18-25.
Hemmer B, Kondo T, Gran B, Pinilla C, Cortese I, Pascal J, et al. Minimal peptide length requirements for CD4(+) T cell clones--implications for molecular mimicry and T cell survival. Int Immunol. 2000;12(3):375-83.
Meeuwsen MH, Wouters AK, Hagedoorn RS, Kester MGD, Remst DFG, van der Steen DM, et al. Cutting Edge: Unconventional CD8(+) T Cell Recognition of a Naturally Occurring HLA-A*02:01- Restricted 20mer Epitope. J Immunol. 2022;208(8):1851-6.
Jimenez-Fernandez M, Rodriguez-Sinovas C, Canes L, Ballester-Servera C, Vara A, Requena S, et al. CD69-oxLDL ligand engagement induces Programmed Cell Death 1 (PD-1) expression in human CD4 + T lymphocytes. Cell Mol Life Sci. 2022;79(8):468.
Hu ZW, Sun W, Wen YH, Ma RQ, Chen L, Chen WQ, et al. CD69 and SBK1 as potential predictors of responses to PD-1/PD-L1 blockade cancer immunotherapy in lung cancer and melanoma. Front Immunol. 2022;13:952059.
Maruhashi T, Okazaki IM, Sugiura D, Takahashi S, Maeda TK, Shimizu K, et al. LAG-3 inhibits the activation of CD4(+) T cells that recognize stable pMHCII through its conformation-dependent recognition of pMHCII. Nat Immunol. 2018;19(12):1415-26.
MacLachlan BJ, Mason GH, Greenshields-Watson A, Triebel F, Gallimore A, Cole DK, et al. Molecular characterization of HLA class II binding to the LAG-3 T cell co-inhibitory receptor. Eur J Immunol. 2021;51(2):331-41.
Liu W, Tang L, Zhang G, Wei H, Cui Y, Guo L, et al. Characterization of a novel C-type lectin-like gene, LSECtin: demonstration of carbohydrate binding and expression in sinusoidal endothelial cells of liver and lymph node. J Biol Chem. 2004;279(18):18748-58.
Kouo T, Huang L, Pucsek AB, Cao M, Solt S, Armstrong T, et al. Galectin-3 Shapes Antitumor Immune Responses by Suppressing CD8+ T Cells via LAG-3 and Inhibiting Expansion of Plasmacytoid Dendritic Cells. Cancer Immunol Res. 2015;3(4):412-23.
Lichtenegger FS, Rothe M, Schnorfeil FM, Deiser K, Krupka C, Augsberger C, et al. Targeting LAG-3 and PD-1 to Enhance T Cell Activation by Antigen-Presenting Cells. Front Immunol. 2018;9:385.
Andreae S, Piras F, Burdin N, Triebel F. Maturation and activation of dendritic cells induced by lymphocyte activation gene-3 (CD223). J Immunol. 2002;168(8):3874-80.
Tiago M, de Oliveira EM, Brohem CA, Pennacchi PC, Paes RD, Haga RB, et al. Fibroblasts protect melanoma cells from the cytotoxic effects of doxorubicin. Tissue Eng Part A. 2014;20(17-18):2412-21.
Mehraj U, Mir IA, Hussain MU, Alkhanani M, Wani NA, Mir MA. Adapalene and Doxorubicin Synergistically Promote Apoptosis of TNBC Cells by Hyperactivation of the ERK1/2 Pathway Through ROS Induction. Front Oncol. 2022;12:938052.
Licarete E, Rauca VF, Luput L, Drotar D, Stejerean I, Patras L, et al. Overcoming Intrinsic Doxorubicin Resistance in Melanoma by Anti-Angiogenic and Anti-Metastatic Effects of Liposomal Prednisolone Phosphate on Tumor Microenvironment. Int J Mol Sci. 2020;21(8).
Bernard S, Poon AC, Tam PM, Mutsaers AJ. Investigation of the effects of mTOR inhibitors rapamycin and everolimus in combination with carboplatin on canine malignant melanoma cells. BMC Vet Res. 2021;17(1):382.
Liebmann JE, Cook JA, Lipschultz C, Teague D, Fisher J, Mitchell JB. Cytotoxic studies of paclitaxel (Taxol) in human tumour cell lines. Br J Cancer. 1993;68(6):1104-9.
Fucikova J, Kepp O, Kasikova L, Petroni G, Yamazaki T, Liu P, et al. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020;11(11):1013.
Kim DY, Pyo A, Yun M, Thangam R, You SH, Zhang Y, et al. Imaging Calreticulin for Early Detection of Immunogenic Cell Death During Anticancer Treatment. J Nucl Med. 2021;62(7):956-60.
Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13(1):54-61.
Liu P, Zhao L, Kepp O, Kroemer G. Quantitation of calreticulin exposure associated with immunogenic cell death. Methods Enzymol. 2020;632:1-13.
Mistarz A, Graczyk M, Winkler M, Singh PK, Cortes E, Miliotto A, et al. Induction of cell death in ovarian cancer cells by doxorubicin and oncolytic vaccinia virus is associated with CREB3L1 activation. Mol Ther Oncolytics. 2021;23:38-50.
Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology. 2014;3:e28518.
Schaer DA, Geeganage S, Amaladas N, Lu ZH, Rasmussen ER, Sonyi A, et al. The Folate Pathway Inhibitor Pemetrexed Pleiotropically Enhances Effects of Cancer Immunotherapy. Clin Cancer Res. 2019;25(23):7175-88.
Lau TS, Chan LKY, Man GCW, Wong CH, Lee JHS, Yim SF, et al. Paclitaxel Induces Immunogenic Cell Death in Ovarian Cancer via TLR4/IKK2/SNARE-Dependent Exocytosis. Cancer Immunol Res. 2020;8(8):1099-111.
Arandjelovic S, Ravichandran KS. Phagocytosis of apoptotic cells in homeostasis. Nat Immunol. 2015;16(9):907-17.
Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-Ullrich JE, et al. Cellsurface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell. 2005;123(2):321-34.
Liu X, Li J, Liu Y, Ding J, Tong Z, Liu Y, et al. Calreticulin acts as an adjuvant to promote dendritic cell maturation and enhances antigen-specific cytotoxic T lymphocyte responses against non-small cell lung cancer cells. Cell Immunol. 2016;300:46-53.
Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology. 2014;3(9):e955691.
dc.rights.coar.fl_str_mv http://purl.org/coar/access_right/c_abf2
dc.rights.license.spa.fl_str_mv Atribución-NoComercial-SinDerivadas 4.0 Internacional
dc.rights.uri.spa.fl_str_mv http://creativecommons.org/licenses/by-nc-nd/4.0/
dc.rights.accessrights.spa.fl_str_mv info:eu-repo/semantics/openAccess
rights_invalid_str_mv Atribución-NoComercial-SinDerivadas 4.0 Internacional
http://creativecommons.org/licenses/by-nc-nd/4.0/
http://purl.org/coar/access_right/c_abf2
eu_rights_str_mv openAccess
dc.format.extent.spa.fl_str_mv 156 páginas
dc.format.mimetype.spa.fl_str_mv application/pdf
dc.publisher.spa.fl_str_mv Universidad Nacional de Colombia
dc.publisher.program.spa.fl_str_mv Bogotá - Medicina - Maestría en Inmunología
dc.publisher.faculty.spa.fl_str_mv Facultad de Medicina
dc.publisher.place.spa.fl_str_mv Bogotá, Colombia
dc.publisher.branch.spa.fl_str_mv Universidad Nacional de Colombia - Sede Bogotá
institution Universidad Nacional de Colombia
bitstream.url.fl_str_mv https://repositorio.unal.edu.co/bitstream/unal/86203/1/license.txt
https://repositorio.unal.edu.co/bitstream/unal/86203/2/1026272802.2024.pdf
https://repositorio.unal.edu.co/bitstream/unal/86203/3/1026272802.2024.pdf.jpg
bitstream.checksum.fl_str_mv eb34b1cf90b7e1103fc9dfd26be24b4a
6f54f9ece62a46be6843d08a1742e2a2
8aad89a8c243229ee65c6be73b9238c9
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
MD5
repository.name.fl_str_mv Repositorio Institucional Universidad Nacional de Colombia
repository.mail.fl_str_mv repositorio_nal@unal.edu.co
_version_ 1806886417507287040
spelling Atribución-NoComercial-SinDerivadas 4.0 Internacionalhttp://creativecommons.org/licenses/by-nc-nd/4.0/info:eu-repo/semantics/openAccesshttp://purl.org/coar/access_right/c_abf2Bernal Estévez, David Andreseebb2b477da688433ca45b907a57e850600Parra López, Carlos Alberto29924f3507fcedbca9501300464d5b61600Alfaro Marenco, María Alejandraf5d9273f2e9e206b32f62581a8fc68762024-06-04T21:14:14Z2024-06-04T21:14:14Z2024https://repositorio.unal.edu.co/handle/unal/86203Universidad Nacional de ColombiaRepositorio Institucional Universidad Nacional de Colombiahttps://repositorio.unal.edu.co/ilustraciones, diagramasLas vacunas basadas en células dendríticas (DCs) se han proyectado como una importante alternativa para la inmunoterapia del cáncer. Su diseño típicamente se fundamenta en la estimulación ex vivo de DCs derivadas de monocitos (MoDCs) utilizando diferentes formulaciones de antígenos y condiciones de maduración. Sin embargo, no hay consenso frente a la formulación óptima del antígeno a la hora de diseñar vacunas basadas en DCs. Por otro lado, se ha descrito que el estímulo de maduración en las MoDCs podría influir en su capacidad para presentar determinados formatos del antígeno. Por lo anterior, el presente trabajo se propuso explorar las implicaciones de diversas formulaciones del antígeno en el desarrollo de respuestas inmunes antitumorales por LT CD8+ cultivados con MoDCs tratadas bajo distintas condiciones. Para ello, se identificaron donantes sanos con LT CD8+ específicos contra epítopes modelo, utilizando el método de citometría basado en tetrámero. De forma paralela, se derivaron MoDCs que fueron estimuladas con dichos antígenos, bajo las formulaciones de péptido corto, péptido largo, proteína recombinante y célula tumoral, todas portadoras de la epítope inmunogénica de cada antígeno. Las MoDCs fueron maduradas empleando diferentes cocteles de citoquinas y se analizó la expresión diferencial de marcadores de maduración. Las líneas tumorales fueron tratadas con agentes quimioterapéuticos, a fin de determinar la expresión de calreticulina (CLR) como indicador de muerte celular inmunogénica (ICD) y su capacidad de favorecer su fagocitosis por MoDCs. Finalmente, se estableció un cocultivo de MoDCs y LT CD8+ autólogos antígeno-específicos o clones de LT CD8+ alogénicos específicos para las epítopes de interés; cuya respuesta efectora fue evaluada por medio de la producción de citoquinas y la expresión de marcadores de activación, agotamiento y memoria, utilizando citometría de flujo multiparamétrica. Se logró la expansión de LT CD8+ HLA-A*02:01 tetrámero-positivos para una epítope viral y una tumoral, con los cuales se realizaron los ensayos de comparación entre formatos del antígeno y cocteles de maduración. Como se ha reportado clásicamente en la literatura, las formulaciones sencillas y directas basadas en péptidos promovieron respuestas robustas de activación y producción de citoquinas. Por el contrario, las formulaciones complejas como la proteína recombinante y las células tumorales tratadas mostraron respuestas efectoras menos evidentes. Los fenotipos de activación y agotamiento exhibieron una expresión dependiente principalmente del formato del antígeno utilizado, aunque se observó heterogeneidad entre los individuos. Adicionalmente, el marcador LAG-3 se vio influenciado por el coctel de maduración utilizado en las MoDCs. Por último, se logró inducir la expresión de CRL en las líneas tumorales tratadas y su fagocitosis por iDCs, aunque no se evidenció presentación cruzada del antígeno. Los resultados de este proyecto proporcionan información valiosa para el diseño de vacunas basadas en DCs y su potencial en inmunoterapia del cáncer. (Texto tomado de la fuente).Dendritic cell (DC)-based vaccines have been projected as a significant alternative for cancer immunotherapy. Typically, these vaccines are designed by stimulating monocyte-derived dendritic cells (MoDCs) ex vivo, employing diverse antigen formulations and maturation conditions. However, there is no consensus regarding the optimal antigen formulation when designing DC-based vaccines. On the other hand, it has been noted that the maturation stimulus in MoDCs could influence their ability to present specific antigen formats. This project aimed to investigate the effects of various antigen formulations on the development of anti-tumor immune responses by CD8+ T cells cultured with monocyte-derived dendritic cells (MoDCs) treated under different conditions. To achieve this, healthy donors with antigen-specific CD8+ T cell precursors for model epitopes were identified using tetramer-based flow cytometry. Simultaneously, MoDCs were derived and stimulated employing short peptide, long peptide, recombinant protein, or tumor cell formulations, all carrying the immunogenic epitope of each antigen. The MoDCs were matured using different cytokine cocktails, and the differential expression of maturation markers was analyzed. Tumor cell lines were treated with chemotherapeutic agents to determine calreticulin (CLR) expression as an indicator of immunogenic cell death (ICD). The ability of treated tumor cells to enhance phagocytosis by MoDCs was also assessed. Finally, a co-culture of autologous MoDCs and antigen-specific CD8+ T cells or allogeneic CD8+ T cell clones specific for the target epitopes was established. Their effector response was evaluated through cytokine production and the expression of activation, exhaustion, and memory markers, utilizing multiparametric flow cytometry. The expansion of HLA-A*02:01 tetramer-positive CD8+ T cells was achieved for a viral and a tumor epitope. Using these, comparative assays were carried out between antigen formats and maturation cocktails. As classically reported in the literature, peptide-based formulations promoted robust activation and cytokine production responses. In contrast, more complex formulations such as recombinant protein and treated tumor cells exhibited less evident effector responses. The activation and exhaustion phenotypes showed expression primarily dependent on the antigen format used, although heterogeneity was observed among individuals. Additionally, LAG-3 was 13 influenced by the maturation cocktail used in MoDCs. Finally, the induction of calreticulin (CRL) expression in treated tumor cell lines and their phagocytosis by iDCs was achieved, although no evidence of antigen cross-presentation was observed. The findings of this project provide valuable insights for the design of DC-based vaccines and their potential in cancer immunotherapy.Fundación Salud de los AndesMaestríaMagíster en InmunologíaEstudio experimental in vitro156 páginasapplication/pdfUniversidad Nacional de ColombiaBogotá - Medicina - Maestría en InmunologíaFacultad de MedicinaBogotá, ColombiaUniversidad Nacional de Colombia - Sede Bogotá610 - Medicina y salud::616 - Enfermedades610 - Medicina y salud::612 - Fisiología humanaInmunoterapia/métodosVacunas contra el CáncerAntígenos/uso terapéuticoImmunotherapy/methodsCancer VaccinesAntigens/therapeutic useLinfocitos T CD8+Muerte celular inmunogénicaCitometría de flujoCélulas dendríticas derivadas de monocitosFormulaciones del antígenoAntígenos modeloMonocyte-derived dendritic cellsAntigen formulationsModel antigensCD8+ T cellsTumor cell linesimmunogenic cell deathFlow cytometryEvaluación de la inmunogenicidad de células dendríticas pulsadas con diferentes formulaciones del antígeno con fines de inmunoterapiaAssessment of the immunogenicity of dendritic cells pulsed with different antigen formulations for immunotherapy purposesTrabajo de grado - Maestríainfo:eu-repo/semantics/masterThesisinfo:eu-repo/semantics/acceptedVersionTexthttp://purl.org/redcol/resource_type/TMGierlich P, Lex V, Technau A, Keupp A, Morper L, Glunz A, et al. Prostaglandin E(2) in a TLR3- and 7/8-agonist-based DC maturation cocktail generates mature, cytokine-producing, migratory DCs but impairs antigen cross-presentation to CD8(+) T cells. Cancer Immunol Immunother. 2020;69(6):1029-42.Chamucero-Millares JA, Bernal-Estevez DA, Parra-Lopez CA. Usefulness of IL-21, IL-7, and IL- 15 conditioned media for expansion of antigen-specific CD8+ T cells from healthy donor-PBMCs suitable for immunotherapy. Cell Immunol. 2021;360:104257.Patente TA, Pinho MP, Oliveira AA, Evangelista GCM, Bergami-Santos PC, Barbuto JAM. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front Immunol. 2018;9:3176.Granot T, Senda T, Carpenter DJ, Matsuoka N, Weiner J, Gordon CL, et al. Dendritic Cells Display Subset and Tissue-Specific Maturation Dynamics over Human Life. Immunity. 2017;46(3):504-15.Lamberti MJ, Nigro A, Mentucci FM, Rumie Vittar NB, Casolaro V, Dal Col J. Dendritic Cells and Immunogenic Cancer Cell Death: A Combination for Improving Antitumor Immunity. Pharmaceutics. 2020;12(3).Bol KF, Schreibelt G, Gerritsen WR, de Vries IJ, Figdor CG. Dendritic Cell-Based Immunotherapy: State of the Art and Beyond. Clin Cancer Res. 2016;22(8):1897-906.Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based immunotherapy. Cell Res. 2017;27(1):74-95.Hu Z, Ott PA, Wu CJ. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat Rev Immunol. 2018;18(3):168-82.Thomas R, Al-Khadairi G, Roelands J, Hendrickx W, Dermime S, Bedognetti D, et al. NY-ESO-1 Based Immunotherapy of Cancer: Current Perspectives. Front Immunol. 2018;9:947.Mastelic-Gavillet B, Balint K, Boudousquie C, Gannon PO, Kandalaft LE. Personalized Dendritic Cell Vaccines-Recent Breakthroughs and Encouraging Clinical Results. Front Immunol. 2019;10:766.Sarivalasis A, Boudousquie C, Balint K, Stevenson BJ, Gannon PO, Iancu EM, et al. A Phase I/II trial comparing autologous dendritic cell vaccine pulsed either with personalized peptides (PEP-DC) or with tumor lysate (OC-DC) in patients with advanced high-grade ovarian serous carcinoma. J Transl Med. 2019;17(1):391.Prins RM, Wang X, Soto H, Young E, Lisiero DN, Fong B, et al. Comparison of gliomaassociated antigen peptide-loaded versus autologous tumor lysate-loaded dendritic cell vaccination in malignant glioma patients. J Immunother. 2013;36(2):152-7.Castiello L, Sabatino M, Jin P, Clayberger C, Marincola FM, Krensky AM, et al. Monocytederived DC maturation strategies and related pathways: a transcriptional view. Cancer Immunol Immunother. 2011;60(4):457-66.C DEW, M VDB, Hoefnagel M. Regulatory perspective on in vitro potency assays for human dendritic cells used in anti-tumor immunotherapy. Cytotherapy. 2018;20(11):1289-308.Lee J, Breton G, Oliveira TY, Zhou YJ, Aljoufi A, Puhr S, et al. Restricted dendritic cell and monocyte progenitors in human cord blood and bone marrow. J Exp Med. 2015;212(3):385-99.Gerlini G, Urso C, Mariotti G, Di Gennaro P, Palli D, Brandani P, et al. Plasmacytoid dendritic cells represent a major dendritic cell subset in sentinel lymph nodes of melanoma patients and accumulate in metastatic nodes. Clin Immunol. 2007;125(2):184-93.Wylie B, Macri C, Mintern JD, Waithman J. Dendritic Cells and Cancer: From Biology to Therapeutic Intervention. Cancers (Basel). 2019;11(4).Huysamen C, Willment JA, Dennehy KM, Brown GD. CLEC9A is a novel activation C-type lectin-like receptor expressed on BDCA3+ dendritic cells and a subset of monocytes. J Biol Chem. 2008;283(24):16693-701.Sancho D, Joffre OP, Keller AM, Rogers NC, Martinez D, Hernanz-Falcon P, et al. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature. 2009;458(7240):899-903.Constantino J, Gomes C, Falcao A, Neves BM, Cruz MT. Dendritic cell-based immunotherapy: a basic review and recent advances. Immunol Res. 2017;65(4):798-810.Calmeiro J, Mendes L, Duarte IF, Leitao C, Tavares AR, Ferreira DA, et al. In-Depth Analysis of the Impact of Different Serum-Free Media on the Production of Clinical Grade Dendritic Cells for Cancer Immunotherapy. Front Immunol. 2020;11:593363.Munz C, Dao T, Ferlazzo G, de Cos MA, Goodman K, Young JW. Mature myeloid dendritic cell subsets have distinct roles for activation and viability of circulating human natural killer cells. Blood. 2005;105(1):266-73.Luo XL, Dalod M. The quest for faithful in vitro models of human dendritic cells types. Mol Immunol. 2020;123:40-59.Chiang CL, Kandalaft LE, Tanyi J, Hagemann AR, Motz GT, Svoronos N, et al. A dendritic cell vaccine pulsed with autologous hypochlorous acid-oxidized ovarian cancer lysate primes effective broad antitumor immunity: from bench to bedside. Clin Cancer Res. 2013;19(17):4801-15.Mailliard RB, Wankowicz-Kalinska A, Cai Q, Wesa A, Hilkens CM, Kapsenberg ML, et al. alphatype-1 polarized dendritic cells: a novel immunization tool with optimized CTL-inducing activity. Cancer Res. 2004;64(17):5934-7.Lee JJ, Foon KA, Mailliard RB, Muthuswamy R, Kalinski P. Type 1-polarized dendritic cells loaded with autologous tumor are a potent immunogen against chronic lymphocytic leukemia. J Leukoc Biol. 2008;84(1):319-25.Garris CS, Arlauckas SP, Kohler RH, Trefny MP, Garren S, Piot C, et al. Successful Anti-PD-1 Cancer Immunotherapy Requires T Cell-Dendritic Cell Crosstalk Involving the Cytokines IFN-gamma and IL-12. Immunity. 2018;49(6):1148-61 e7.Del Prete A, Sozio F, Barbazza I, Salvi V, Tiberio L, Laffranchi M, et al. Functional Role of Dendritic Cell Subsets in Cancer Progression and Clinical Implications. Int J Mol Sci. 2020;21(11).Cannon MJ, Schmid DS, Hyde TB. Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection. Rev Med Virol. 2010;20(4):202-13.Terrazzini N, Kern F. Cell-mediated immunity to human CMV infection: a brief overview. F1000Prime Rep. 2014;6:28.Schober K, Buchholz VR, Busch DH. TCR repertoire evolution during maintenance of CMVspecific T-cell populations. Immunol Rev. 2018;283(1):113-28.O'Hara GA, Welten SP, Klenerman P, Arens R. Memory T cell inflation: understanding cause and effect. Trends Immunol. 2012;33(2):84-90.Khan N, Best D, Bruton R, Nayak L, Rickinson AB, Moss PA. T cell recognition patterns of immunodominant cytomegalovirus antigens in primary and persistent infection. J Immunol. 2007;178(7):4455-65.Wills MR, Carmichael AJ, Mynard K, Jin X, Weekes MP, Plachter B, et al. The human cytotoxic T-lymphocyte (CTL) response to cytomegalovirus is dominated by structural protein pp65: frequency, specificity, and T-cell receptor usage of pp65-specific CTL. J Virol. 1996;70(11):7569-79.Pittet MJ, Zippelius A, Valmori D, Speiser DE, Cerottini JC, Romero P. Melan-A/MART-1-specific CD8 T cells: from thymus to tumor. Trends Immunol. 2002;23(7):325-8.Coulie PG, Brichard V, Van Pel A, Wolfel T, Schneider J, Traversari C, et al. A new gene coding for a differentiation antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J Exp Med. 1994;180(1):35-42.Pittet MJ, Valmori D, Dunbar PR, Speiser DE, Lienard D, Lejeune F, et al. High frequencies of naive Melan-A/MART-1-specific CD8(+) T cells in a large proportion of human histocompatibility leukocyte antigen (HLA)-A2 individuals. J Exp Med. 1999;190(5):705-15.Alanio C, Lemaitre F, Law HK, Hasan M, Albert ML. Enumeration of human antigen-specific naive CD8+ T cells reveals conserved precursor frequencies. Blood. 2010;115(18):3718-25.Valmori D, Fonteneau JF, Lizana CM, Gervois N, Lienard D, Rimoldi D, et al. Enhanced generation of specific tumor-reactive CTL in vitro by selected Melan-A/MART-1 immunodominant peptide analogues. J Immunol. 1998;160(4):1750-8.van Elsas A, van der Burg SH, van der Minne CE, Borghi M, Mourer JS, Melief CJ, et al. Peptide-pulsed dendritic cells induce tumoricidal cytotoxic T lymphocytes from healthy donors against stably HLA-A*0201-binding peptides from the Melan-A/MART-1 self antigen. Eur J Immunol. 1996;26(8):1683-9.Bakker AB, Marland G, de Boer AJ, Huijbens RJ, Danen EH, Adema GJ, et al. Generation of antimelanoma cytotoxic T lymphocytes from healthy donors after presentation of melanomaassociated antigen-derived epitopes by dendritic cells in vitro. Cancer Res. 1995;55(22):5330-4.Cebon J, Knights A, Ebert L, Jackson H, Chen W. Evaluation of cellular immune responses in cancer vaccine recipients: lessons from NY-ESO-1. Expert Rev Vaccines. 2010;9(6):617-29.Chen YT, Scanlan MJ, Sahin U, Tureci O, Gure AO, Tsang S, et al. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci U S A. 1997;94(5):1914-8.Sabbatini P, Tsuji T, Ferran L, Ritter E, Sedrak C, Tuballes K, et al. Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients. Clin Cancer Res. 2012;18(23):6497-508.Raza A, Merhi M, Inchakalody VP, Krishnankutty R, Relecom A, Uddin S, et al. Unleashing the immune response to NY-ESO-1 cancer testis antigen as a potential target for cancer immunotherapy. J Transl Med. 2020;18(1):140.Slingluff CL, Jr. The present and future of peptide vaccines for cancer: single or multiple, long or short, alone or in combination? Cancer J. 2011;17(5):343-50.Yamshchikov GV, Barnd DL, Eastham S, Galavotti H, Patterson JW, Deacon DH, et al. Evaluation of peptide vaccine immunogenicity in draining lymph nodes and peripheral blood of melanoma patients. Int J Cancer. 2001;92(5):703-11.Rosenberg SA, Sherry RM, Morton KE, Scharfman WJ, Yang JC, Topalian SL, et al. Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma. J Immunol. 2005;175(9):6169-76.Toes RE, Blom RJ, Offringa R, Kast WM, Melief CJ. Enhanced tumor outgrowth after peptide vaccination. Functional deletion of tumor-specific CTL induced by peptide vaccination can lead to the inability to reject tumors. J Immunol. 1996;156(10):3911-8.Toes RE, Offringa R, Blom RJ, Melief CJ, Kast WM. Peptide vaccination can lead to enhanced tumor growth through specific T-cell tolerance induction. Proc Natl Acad Sci U S A. 1996;93(15):7855-60.Janssen EM, Droin NM, Lemmens EE, Pinkoski MJ, Bensinger SJ, Ehst BD, et al. CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-mediated activation-induced cell death. Nature. 2005;434(7029):88-93.Srinivasan M, Domanico SZ, Kaumaya PT, Pierce SK. Peptides of 23 residues or greater are required to stimulate a high affinity class II-restricted T cell response. Eur J Immunol. 1993;23(5):1011-6.Zwaveling S, Ferreira Mota SC, Nouta J, Johnson M, Lipford GB, Offringa R, et al. Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J Immunol. 2002;169(1):350-8.Ma Y, Adjemian S, Mattarollo SR, Yamazaki T, Aymeric L, Yang H, et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity. 2013;38(4):729-41.Pathak SK, Skold AE, Mohanram V, Persson C, Johansson U, Spetz AL. Activated apoptotic cells induce dendritic cell maturation via engagement of Toll-like receptor 4 (TLR4), dendritic cellspecific intercellular adhesion molecule 3 (ICAM-3)-grabbing nonintegrin (DC-SIGN), and beta2 integrins. J Biol Chem. 2012;287(17):13731-42.Han Q, Bagheri N, Bradshaw EM, Hafler DA, Lauffenburger DA, Love JC. Polyfunctional responses by human T cells result from sequential release of cytokines. Proc Natl Acad Sci U S A. 2012;109(5):1607-12.de Araujo-Souza PS, Hanschke SCH, Nardy A, Secca C, Oliveira-Vieira B, Silva KL, et al. Differential interferon-gamma production by naive and memory-like CD8 T cells. J Leukoc Biol. 2020;108(4):1329-37.Bhat P, Leggatt G, Waterhouse N, Frazer IH. Interferon-gamma derived from cytotoxic lymphocytes directly enhances their motility and cytotoxicity. Cell Death Dis. 2017;8(6):e2836.Lachmann R, Bajwa M, Vita S, Smith H, Cheek E, Akbar A, et al. Polyfunctional T cells accumulate in large human cytomegalovirus-specific T cell responses. J Virol. 2012;86(2):1001-9.Gubser PM, Bantug GR, Razik L, Fischer M, Dimeloe S, Hoenger G, et al. Rapid effector function of memory CD8+ T cells requires an immediate-early glycolytic switch. Nat Immunol. 2013;14(10):1064-72.Sathaliyawala T, Kubota M, Yudanin N, Turner D, Camp P, Thome JJ, et al. Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity. 2013;38(1):187-97.Mehta AK, Gracias DT, Croft M. TNF activity and T cells. Cytokine. 2018;101:14-8.Brehm MA, Daniels KA, Welsh RM. Rapid production of TNF-alpha following TCR engagement of naive CD8 T cells. J Immunol. 2005;175(8):5043-9.Harari A, Dutoit V, Cellerai C, Bart PA, Du Pasquier RA, Pantaleo G. Functional signatures of protective antiviral T-cell immunity in human virus infections. Immunol Rev. 2006;211:236-54.Healy ZR, Weinhold KJ, Murdoch DM. Transcriptional Profiling of CD8+ CMV-Specific T Cell Functional Subsets Obtained Using a Modified Method for Isolating High-Quality RNA From Fixed and Permeabilized Cells. Front Immunol. 2020;11:1859.Imai N, Tawara I, Yamane M, Muraoka D, Shiku H, Ikeda H. CD4(+) T cells support polyfunctionality of cytotoxic CD8(+) T cells with memory potential in immunological control of tumor. Cancer Sci. 2020;111(6):1958-68.Reddy M, Eirikis E, Davis C, Davis HM, Prabhakar U. Comparative analysis of lymphocyte activation marker expression and cytokine secretion profile in stimulated human peripheral blood mononuclear cell cultures: an in vitro model to monitor cellular immune function. J Immunol Methods. 2004;293(1-2):127-42.Adamczyk M, Bartosinska J, Raczkiewicz D, Kowal M, Surdacka A, Krasowska D, et al. The Expression of Activation Markers CD25 and CD69 Increases during Biologic Treatment of Psoriasis. J Clin Med. 2023;12(20).Cebrian M, Yague E, Rincon M, Lopez-Botet M, de Landazuri MO, Sanchez-Madrid F. Triggering of T cell proliferation through AIM, an activation inducer molecule expressed on activated human lymphocytes. J Exp Med. 1988;168(5):1621-37.Risso A, Smilovich D, Capra MC, Baldissarro I, Yan G, Bargellesi A, et al. CD69 in resting and activated T lymphocytes. Its association with a GTP binding protein and biochemical requirements for its expression. J Immunol. 1991;146(12):4105-14.Cibrian D, Sanchez-Madrid F. CD69: from activation marker to metabolic gatekeeper. Eur J Immunol. 2017;47(6):946-53.Ziegler SF, Ramsdell F, Alderson MR. The activation antigen CD69. Stem Cells. 1994;12(5):456-65.Gonzalez-Amaro R, Cortes JR, Sanchez-Madrid F, Martin P. Is CD69 an effective brake to control inflammatory diseases? Trends Mol Med. 2013;19(10):625-32.de la Fuente H, Cruz-Adalia A, Martinez Del Hoyo G, Cibrian-Vera D, Bonay P, Perez-Hernandez D, et al. The leukocyte activation receptor CD69 controls T cell differentiation through its interaction with galectin-1. Mol Cell Biol. 2014;34(13):2479-87.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155(3):1151-64.Brusko TM, Wasserfall CH, Hulme MA, Cabrera R, Schatz D, Atkinson MA. Influence of membrane CD25 stability on T lymphocyte activity: implications for immunoregulation. PLoS One. 2009;4(11):e7980.Green DR, Droin N, Pinkoski M. Activation-induced cell death in T cells. Immunol Rev. 2003;193:70-81.Poulton TA, Gallagher A, Potts RC, Beck JS. Changes in activation markers and cell membrane receptors on human peripheral blood T lymphocytes during cell cycle progression after PHA stimulation. Immunology. 1988;64(3):419-25.Watts TH. TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol. 2005;23:23-68.Wolfl M, Kuball J, Eyrich M, Schlegel PG, Greenberg PD. Use of CD137 to study the full repertoire of CD8+ T cells without the need to know epitope specificities. Cytometry A. 2008;73(11):1043-9.Wen T, Bukczynski J, Watts TH. 4-1BB ligand-mediated costimulation of human T cells induces CD4 and CD8 T cell expansion, cytokine production, and the development of cytolytic effector function. J Immunol. 2002;168(10):4897-906.Halstead ES, Mueller YM, Altman JD, Katsikis PD. In vivo stimulation of CD137 broadens primary antiviral CD8+ T cell responses. Nat Immunol. 2002;3(6):536-41.Vinay DS, Kwon BS. Role of 4-1BB in immune responses. Semin Immunol. 1998;10(6):481-9.Etxeberria I, Glez-Vaz J, Teijeira A, Melero I. New emerging targets in cancer immunotherapy: CD137/4-1BB costimulatory axis. ESMO Open. 2020;4(Suppl 3):e000733.Alosaimi MF, Hoenig M, Jaber F, Platt CD, Jones J, Wallace J, et al. Immunodeficiency and EBV-induced lymphoproliferation caused by 4-1BB deficiency. J Allergy Clin Immunol. 2019;144(2):574-83 e5.Zhu Y, Zhu G, Luo L, Flies AS, Chen L. CD137 stimulation delivers an antigen-independent growth signal for T lymphocytes with memory phenotype. Blood. 2007;109(11):4882-9.Melero I, Shuford WW, Newby SA, Aruffo A, Ledbetter JA, Hellstrom KE, et al. Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med. 1997;3(6):682-5.Grewal IS, Flavell RA. CD40 and CD154 in cell-mediated immunity. Annu Rev Immunol. 1998;16:111-35.Caux C, Massacrier C, Vanbervliet B, Dubois B, Van Kooten C, Durand I, et al. Activation of human dendritic cells through CD40 cross-linking. J Exp Med. 1994;180(4):1263-72.Bennett SR, Carbone FR, Karamalis F, Flavell RA, Miller JF, Heath WR. Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature. 1998;393(6684):478-80.Schoenberger SP, Toes RE, van der Voort EI, Offringa R, Melief CJ. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature. 1998;393(6684):480-3.Tay NQ, Lee DCP, Chua YL, Prabhu N, Gascoigne NRJ, Kemeny DM. CD40L Expression Allows CD8(+) T Cells to Promote Their Own Expansion and Differentiation through Dendritic Cells. Front Immunol. 2017;8:1484.Wong KL, Lew FC, MacAry PA, Kemeny DM. CD40L-expressing CD8 T cells prime CD8alpha(+) DC for IL-12p70 production. Eur J Immunol. 2008;38(8):2251-62.Wong KL, Tang LF, Lew FC, Wong HS, Chua YL, MacAry PA, et al. CD44high memory CD8 T cells synergize with CpG DNA to activate dendritic cell IL-12p70 production. J Immunol. 2009;183(1):41-50.Bourgeois C, Rocha B, Tanchot C. A role for CD40 expression on CD8+ T cells in the generation of CD8+ T cell memory. Science. 2002;297(5589):2060-3.Hernandez MG, Shen L, Rock KL. CD40-CD40 ligand interaction between dendritic cells and CD8+ T cells is needed to stimulate maximal T cell responses in the absence of CD4+ T cell help. J Immunol. 2007;178(5):2844-52.Shugart JA, Bambina S, Alice AF, Montler R, Bahjat KS. A self-help program for memory CD8+ T cells: positive feedback via CD40-CD40L signaling as a critical determinant of secondary expansion. PLoS One. 2013;8(5):e64878.Olson MR, Seah SG, Edenborough K, Doherty PC, Lew AM, Turner SJ. CD154+ CD4+ T-cell dependence for effective memory influenza virus-specific CD8+ T-cell responses. Immunol Cell Biol. 2014;92(7):605-11.Stinchcombe JC, Majorovits E, Bossi G, Fuller S, Griffiths GM. Centrosome polarization delivers secretory granules to the immunological synapse. Nature. 2006;443(7110):462-5.Ballesteros-Tato A, Leon B, Lee BO, Lund FE, Randall TD. Epitope-specific regulation of memory programming by differential duration of antigen presentation to influenza-specific CD8(+) T cells. Immunity. 2014;41(1):127-40.Blank CU, Haining WN, Held W, Hogan PG, Kallies A, Lugli E, et al. Defining 'T cell exhaustion'. Nat Rev Immunol. 2019;19(11):665-74.Ahn E, Araki K, Hashimoto M, Li W, Riley JL, Cheung J, et al. Role of PD-1 during effector CD8 T cell differentiation. Proc Natl Acad Sci U S A. 2018;115(18):4749-54.Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492-9.Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8(3):239-45.Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017;355(6332):1428-33.Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol. 2005;25(21):9543- 53.Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates Tcell proliferation and interleukin-10 secretion. Nat Med. 1999;5(12):1365-9.Tseng SY, Otsuji M, Gorski K, Huang X, Slansky JE, Pai SI, et al. B7-DC, a new dendritic cell molecule with potent costimulatory properties for T cells. J Exp Med. 2001;193(7):839-46.Keir ME, Freeman GJ, Sharpe AH. PD-1 regulates self-reactive CD8+ T cell responses to antigen in lymph nodes and tissues. J Immunol. 2007;179(8):5064-70.Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455-65.Meng X, Liu X, Guo X, Jiang S, Chen T, Hu Z, et al. FBXO38 mediates PD-1 ubiquitination and regulates anti-tumour immunity of T cells. Nature. 2018;564(7734):130-5.Chan DV, Gibson HM, Aufiero BM, Wilson AJ, Hafner MS, Mi QS, et al. Differential CTLA-4 expression in human CD4+ versus CD8+ T cells is associated with increased NFAT1 and inhibition of CD4+ proliferation. Genes Immun. 2014;15(1):25-32.Dariavach P, Mattei MG, Golstein P, Lefranc MP. Human Ig superfamily CTLA-4 gene: chromosomal localization and identity of protein sequence between murine and human CTLA-4 cytoplasmic domains. Eur J Immunol. 1988;18(12):1901-5.Harper K, Balzano C, Rouvier E, Mattei MG, Luciani MF, Golstein P. CTLA-4 and CD28 activated lymphocyte molecules are closely related in both mouse and human as to sequence, message expression, gene structure, and chromosomal location. J Immunol. 1991;147(3):1037-44.Auchincloss H, Turka LA. CTLA-4: not all costimulation is stimulatory. J Immunol. 2011;187(7):3457-8.Lindsten T, Lee KP, Harris ES, Petryniak B, Craighead N, Reynolds PJ, et al. Characterization of CTLA-4 structure and expression on human T cells. J Immunol. 1993;151(7):3489-99.Gajewski TF, Fallarino F, Fields PE, Rivas F, Alegre ML. Absence of CTLA-4 lowers the activation threshold of primed CD8+ TCR-transgenic T cells: lack of correlation with Src homology domain 2-containing protein tyrosine phosphatase. J Immunol. 2001;166(6):3900-7.Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994;1(5):405-13.Triebel F, Jitsukawa S, Baixeras E, Roman-Roman S, Genevee C, Viegas-Pequignot E, et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med. 1990;171(5):1393-405.Workman CJ, Cauley LS, Kim IJ, Blackman MA, Woodland DL, Vignali DA. Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell population following antigen activation in vivo. J Immunol. 2004;172(9):5450-5.Hannier S, Tournier M, Bismuth G, Triebel F. CD3/TCR complex-associated lymphocyte activation gene-3 molecules inhibit CD3/TCR signaling. J Immunol. 1998;161(8):4058-65.Workman CJ, Dugger KJ, Vignali DA. Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J Immunol. 2002;169(10):5392-5.Huard B, Tournier M, Hercend T, Triebel F, Faure F. Lymphocyte-activation gene 3/major histocompatibility complex class II interaction modulates the antigenic response of CD4+ T lymphocytes. Eur J Immunol. 1994;24(12):3216-21.Goldberg MV, Drake CG. LAG-3 in Cancer Immunotherapy. Curr Top Microbiol Immunol. 2011;344:269-78.Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10(1):29-37.Bijker MS, van den Eeden SJ, Franken KL, Melief CJ, van der Burg SH, Offringa R. Superior induction of anti-tumor CTL immunity by extended peptide vaccines involves prolonged, DC-focused antigen presentation. Eur J Immunol. 2008;38(4):1033-42.Gutierrez-Martinez E, Planes R, Anselmi G, Reynolds M, Menezes S, Adiko AC, et al. Cross-Presentation of Cell-Associated Antigens by MHC Class I in Dendritic Cell Subsets. Front Immunol. 2015;6:363.Alloatti A, Kotsias F, Magalhaes JG, Amigorena S. Dendritic cell maturation and crosspresentation: timing matters! Immunol Rev. 2016;272(1):97-108.Menager J, Ebstein F, Oger R, Hulin P, Nedellec S, Duverger E, et al. 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(2):e89897.Tang-Huau TL, Gueguen P, Goudot C, Durand M, Bohec M, Baulande S, et al. Human in vivogenerated monocyte-derived dendritic cells and macrophages cross-present antigens through a vacuolar pathway. Nat Commun. 2018;9(1):2570.Fonteneau JF, Kavanagh DG, Lirvall M, Sanders C, Cover TL, Bhardwaj N, et al. Characterization of the MHC class I cross-presentation pathway for cell-associated antigens by human dendritic cells. Blood. 2003;102(13):4448-55.Du J MA, Widlund HR, Horstmann MA, Ramaswamy S, Fisher DE. MLANA/MART1 and SILV/PMEL17/GP100 are transcriptionally regulated by MITF in melanocytes and melanoma. Am J Pathol. 2003(Jul;163(1):333-43.).Li J, Song JS, Bell RJ, Tran TN, Haq R, Liu H, et al. YY1 regulates melanocyte development and function by cooperating with MITF. PLoS Genet. 2012;8(5):e1002688.Vidard L. 4-1BB and cytokines trigger human NK, gammadelta T, and CD8(+) T cell proliferation and activation, but are not required for their effector functions. Immun Inflamm Dis. 2023;11(1):e749.Almunia C, Bretaudeau M, Held G, Babon A, Marchetti C, Castelli FA, et al. Bee Venom Phospholipase A2, a Good "Chauffeur" for Delivering Tumor Antigen to the MHC I and MHC II Peptide-Loading Compartments of the Dendritic Cells: The Case of NY-ESO-1. PLoS One. 2013;8(6):e67645.Levy F, Muehlethaler K, Salvi S, Peitrequin AL, Lindholm CK, Cerottini JC, et al. Ubiquitylation of a melanosomal protein by HECT-E3 ligases serves as sorting signal for lysosomal degradation. Mol Biol Cell. 2005;16(4):1777-87.Sabbatino F, Wang Y, Scognamiglio G, Favoino E, Feldman SA, Villani V, et al. Antitumor Activity of BRAF Inhibitor and IFNalpha Combination in BRAF-Mutant Melanoma. J Natl Cancer Inst. 2016;108(7).Suriano R, Rajoria S, A LG, Geliebter J, Wallack M, Tiwari RK. Ex vivo derived primary melanoma cells: implications for immunotherapeutic vaccines. J Cancer. 2013;4(5):371-82.Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 1994;179(4):1109-18.Jonuleit H, Kuhn U, Muller G, Steinbrink K, Paragnik L, Schmitt E, et al. Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. Eur J Immunol. 1997;27(12):3135-42.Martinuzzi E, Afonso G, Gagnerault MC, Naselli G, Mittag D, Combadiere B, et al. acDCs enhance human antigen-specific T-cell responses. Blood. 2011;118(8):2128-37.Aspord C, Leloup C, Reche S, Plumas J. pDCs efficiently process synthetic long peptides to induce functional virus- and tumour-specific T-cell responses. Eur J Immunol. 2014;44(10):2880-92.Wada H, Isobe M, Kakimi K, Mizote Y, Eikawa S, Sato E, et al. Vaccination with NY-ESO-1 overlapping peptides mixed with Picibanil OK-432 and montanide ISA-51 in patients with cancers expressing the NY-ESO-1 antigen. J Immunother. 2014;37(2):84-92.Martínez-Enríquez L. Identificación y caracterización de linfocitos T neoantígeno específicos de donantes sanos con fines de inmunoterapia en cáncer: Universidad Nacional de Colombia; 2022.Weekes MP, Wills MR, Mynard K, Carmichael AJ, Sissons JG. The memory cytotoxic Tlymphocyte (CTL) response to human cytomegalovirus infection contains individual peptide-specific CTL clones that have undergone extensive expansion in vivo. J Virol. 1999;73(3):2099-108.Solache A, Morgan CL, Dodi AI, Morte C, Scott I, Baboonian C, et al. Identification of three HLA-A*0201-restricted cytotoxic T cell epitopes in the cytomegalovirus protein pp65 that are conserved between eight strains of the virus. J Immunol. 1999;163(10):5512-8.Bruggner RV, Bodenmiller B, Dill DL, Tibshirani RJ, Nolan GP. Automated identification of stratifying signatures in cellular subpopulations. Proc Natl Acad Sci U S A. 2014;111(26):E2770-7.Carmichael J, DeGraff WG, Gazdar AF, Minna JD, Mitchell JB. Evaluation of a tetrazoliumbased semiautomated colorimetric assay: assessment of chemosensitivity testing. Cancer Res. 1987;47(4):936-42.Infante-Crúz A. Activación de respuesta inmune innata a partir de la inducción de muerte tumoral inmunogénica.: Universidad Nacional de Colombia; 2016.Yadav B, Wennerberg K, Aittokallio T, Tang J. Searching for Drug Synergy in Complex Dose-Response Landscapes Using an Interaction Potency Model. Comput Struct Biotechnol J. 2015;13:504-13.Altomonte M, Gloghini A, Bertola G, Gasparollo A, Carbone A, Ferrone S, et al. Differential expression of cell adhesion molecules CD54/CD11a and CD58/CD2 by human melanoma cells and functional role in their interaction with cytotoxic cells. Cancer Res. 1993;53(14):3343-8.Bernal Estévez D. Evaluación de la capacidad inmuno-estimulante de la terapia neoadyuvante con Doxorrubicina y Ciclofosfamida en pacientes con cáncer de mama: Universidad Nacional de Colombia Sede Bogotá 2017.Mariotti S, Nisini R. Generation of human T cell clones. Methods Mol Biol. 2009;514:65-93.Klapper JA, Thomasian AA, Smith DM, Gorgas GC, Wunderlich JR, Smith FO, et al. Single-pass, closed-system rapid expansion of lymphocyte cultures for adoptive cell therapy. J Immunol Methods. 2009;345(1-2):90-9.Rimoldi D, Muehlethaler K, Salvi S, Valmori D, Romero P, Cerottini JC, et al. Subcellular localization of the melanoma-associated protein Melan-AMART-1 influences the processing of its HLA-A2-restricted epitope. J Biol Chem. 2001;276(46):43189-96.Busam KJ JA. Melan-A, a new melanocytic differentiation marker. Adv Anat Pathol. 1999(Jan;6(1):12-8.).Blass E, Ott PA. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat Rev Clin Oncol. 2021;18(4):215-29.Wahida A, Buschhorn L, Frohling S, Jost PJ, Schneeweiss A, Lichter P, et al. The coming decade in precision oncology: six riddles. Nat Rev Cancer. 2023;23(1):43-54.Olivier T, Haslam A, Tuia J, Prasad V. Eligibility for Human Leukocyte Antigen-Based Therapeutics by Race and Ethnicity. JAMA Netw Open. 2023;6(10):e2338612.Welten SPM, Baumann NS, Oxenius A. Fuel and brake of memory T cell inflation. Med Microbiol Immunol. 2019;208(3-4):329-38.van der Leun AM, Thommen DS, Schumacher TN. CD8(+) T cell states in human cancer: insights from single-cell analysis. Nat Rev Cancer. 2020;20(4):218-32.Rodriguez IJ, Bernal-Estevez DA, Llano-Leon M, Bonilla CE, Parra-Lopez CA. Neoadjuvant chemotherapy modulates exhaustion of T cells in breast cancer patients. PLoS One. 2023;18(2):e0280851.Zhou LJ, Schwarting R, Smith HM, Tedder TF. A novel cell-surface molecule expressed by human interdigitating reticulum cells, Langerhans cells, and activated lymphocytes is a new member of the Ig superfamily. J Immunol. 1992;149(2):735-42.Zhou LJ, Tedder TF. Human blood dendritic cells selectively express CD83, a member of the immunoglobulin superfamily. J Immunol. 1995;154(8):3821-35.Lechmann M, Berchtold S, Hauber J, Steinkasserer A. CD83 on dendritic cells: more than just a marker for maturation. Trends Immunol. 2002;23(6):273-5.Li Z, Ju X, Silveira PA, Abadir E, Hsu WH, Hart DNJ, et al. CD83: Activation Marker for Antigen Presenting Cells and Its Therapeutic Potential. Front Immunol. 2019;10:1312.Trepiakas R, Pedersen AE, Met O, Hansen MH, Berntsen A, Svane IM. Comparison of alpha-Type-1 polarizing and standard dendritic cell cytokine cocktail for maturation of therapeutic monocyte-derived dendritic cell preparations from cancer patients. Vaccine. 2008;26(23):2824-32.Li JG, Du YM, Yan ZD, Yan J, Zhuansun YX, Chen R, et al. CD80 and CD86 knockdown in dendritic cells regulates Th1/Th2 cytokine production in asthmatic mice. Exp Ther Med. 2016;11(3):878-84.Dustin ML, Rothlein R, Bhan AK, Dinarello CA, Springer TA. Induction by IL 1 and interferongamma: tissue distribution, biochemistry, and function of a natural adherence molecule (ICAM-1). J Immunol. 1986;137(1):245-54.Carrasco YR, Fleire SJ, Cameron T, Dustin ML, Batista FD. LFA-1/ICAM-1 interaction lowers the threshold of B cell activation by facilitating B cell adhesion and synapse formation. Immunity. 2004;20(5):589-99.Sheikh NA, Jones LA. CD54 is a surrogate marker of antigen presenting cell activation. Cancer Immunol Immunother. 2008;57(9):1381-90.Josephs TM, Grant EJ, Gras S. Molecular challenges imposed by MHC-I restricted long epitopes on T cell immunity. Biol Chem. 2017;398(9):1027-36.Jimenez-Fernandez M, de la Fuente H, Martin P, Cibrian D, Sanchez-Madrid F. Unraveling CD69 signaling pathways, ligands and laterally associated molecules. EXCLI J. 2023;22:334-51.Hosono M, de Boer OJ, van der Wal AC, van der Loos CM, Teeling P, Piek JJ, et al. Increased expression of T cell activation markers (CD25, CD26, CD40L and CD69) in atherectomy specimens of patients with unstable angina and acute myocardial infarction. Atherosclerosis. 2003;168(1):73-80.Engelhard VH. Structure of peptides associated with class I and class II MHC molecules. Annu Rev Immunol. 1994;12:181-207.Wang M, Larsen MV, Nielsen M, Harndahl M, Justesen S, Dziegiel MH, et al. HLA class I binding 9mer peptides from influenza A virus induce CD4 T cell responses. PLoS One. 2010;5(5):e10533.Wang M, Tang ST, Stryhn A, Justesen S, Larsen MV, Dziegiel MH, et al. Identification of MHC class II restricted T-cell-mediated reactivity against MHC class I binding Mycobacterium tuberculosis peptides. Immunology. 2011;132(4):482-91.Bioley G, Jandus C, Tuyaerts S, Rimoldi D, Kwok WW, Speiser DE, et al. Melan-A/MART-1-specific CD4 T cells in melanoma patients: identification of new epitopes and ex vivo visualization of specific T cells by MHC class II tetramers. J Immunol. 2006;177(10):6769-79.Jandus C, Bioley G, Dojcinovic D, Derre L, Baitsch L, Wieckowski S, et al. Tumor antigenspecific FOXP3+ CD4 T cells identified in human metastatic melanoma: peptide vaccination results in selective expansion of Th1-like counterparts. Cancer Res. 2009;69(20):8085-93.Gross S, Lennerz V, Gallerani E, Mach N, Bohm S, Hess D, et al. Short Peptide Vaccine Induces CD4+ T Helper Cells in Patients with Different Solid Cancers. Cancer Immunol Res. 2016;4(1):18-25.Hemmer B, Kondo T, Gran B, Pinilla C, Cortese I, Pascal J, et al. Minimal peptide length requirements for CD4(+) T cell clones--implications for molecular mimicry and T cell survival. Int Immunol. 2000;12(3):375-83.Meeuwsen MH, Wouters AK, Hagedoorn RS, Kester MGD, Remst DFG, van der Steen DM, et al. Cutting Edge: Unconventional CD8(+) T Cell Recognition of a Naturally Occurring HLA-A*02:01- Restricted 20mer Epitope. J Immunol. 2022;208(8):1851-6.Jimenez-Fernandez M, Rodriguez-Sinovas C, Canes L, Ballester-Servera C, Vara A, Requena S, et al. CD69-oxLDL ligand engagement induces Programmed Cell Death 1 (PD-1) expression in human CD4 + T lymphocytes. Cell Mol Life Sci. 2022;79(8):468.Hu ZW, Sun W, Wen YH, Ma RQ, Chen L, Chen WQ, et al. CD69 and SBK1 as potential predictors of responses to PD-1/PD-L1 blockade cancer immunotherapy in lung cancer and melanoma. Front Immunol. 2022;13:952059.Maruhashi T, Okazaki IM, Sugiura D, Takahashi S, Maeda TK, Shimizu K, et al. LAG-3 inhibits the activation of CD4(+) T cells that recognize stable pMHCII through its conformation-dependent recognition of pMHCII. Nat Immunol. 2018;19(12):1415-26.MacLachlan BJ, Mason GH, Greenshields-Watson A, Triebel F, Gallimore A, Cole DK, et al. Molecular characterization of HLA class II binding to the LAG-3 T cell co-inhibitory receptor. Eur J Immunol. 2021;51(2):331-41.Liu W, Tang L, Zhang G, Wei H, Cui Y, Guo L, et al. Characterization of a novel C-type lectin-like gene, LSECtin: demonstration of carbohydrate binding and expression in sinusoidal endothelial cells of liver and lymph node. J Biol Chem. 2004;279(18):18748-58.Kouo T, Huang L, Pucsek AB, Cao M, Solt S, Armstrong T, et al. Galectin-3 Shapes Antitumor Immune Responses by Suppressing CD8+ T Cells via LAG-3 and Inhibiting Expansion of Plasmacytoid Dendritic Cells. Cancer Immunol Res. 2015;3(4):412-23.Lichtenegger FS, Rothe M, Schnorfeil FM, Deiser K, Krupka C, Augsberger C, et al. Targeting LAG-3 and PD-1 to Enhance T Cell Activation by Antigen-Presenting Cells. Front Immunol. 2018;9:385.Andreae S, Piras F, Burdin N, Triebel F. Maturation and activation of dendritic cells induced by lymphocyte activation gene-3 (CD223). J Immunol. 2002;168(8):3874-80.Tiago M, de Oliveira EM, Brohem CA, Pennacchi PC, Paes RD, Haga RB, et al. Fibroblasts protect melanoma cells from the cytotoxic effects of doxorubicin. Tissue Eng Part A. 2014;20(17-18):2412-21.Mehraj U, Mir IA, Hussain MU, Alkhanani M, Wani NA, Mir MA. Adapalene and Doxorubicin Synergistically Promote Apoptosis of TNBC Cells by Hyperactivation of the ERK1/2 Pathway Through ROS Induction. Front Oncol. 2022;12:938052.Licarete E, Rauca VF, Luput L, Drotar D, Stejerean I, Patras L, et al. Overcoming Intrinsic Doxorubicin Resistance in Melanoma by Anti-Angiogenic and Anti-Metastatic Effects of Liposomal Prednisolone Phosphate on Tumor Microenvironment. Int J Mol Sci. 2020;21(8).Bernard S, Poon AC, Tam PM, Mutsaers AJ. Investigation of the effects of mTOR inhibitors rapamycin and everolimus in combination with carboplatin on canine malignant melanoma cells. BMC Vet Res. 2021;17(1):382.Liebmann JE, Cook JA, Lipschultz C, Teague D, Fisher J, Mitchell JB. Cytotoxic studies of paclitaxel (Taxol) in human tumour cell lines. Br J Cancer. 1993;68(6):1104-9.Fucikova J, Kepp O, Kasikova L, Petroni G, Yamazaki T, Liu P, et al. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020;11(11):1013.Kim DY, Pyo A, Yun M, Thangam R, You SH, Zhang Y, et al. Imaging Calreticulin for Early Detection of Immunogenic Cell Death During Anticancer Treatment. J Nucl Med. 2021;62(7):956-60.Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13(1):54-61.Liu P, Zhao L, Kepp O, Kroemer G. Quantitation of calreticulin exposure associated with immunogenic cell death. Methods Enzymol. 2020;632:1-13.Mistarz A, Graczyk M, Winkler M, Singh PK, Cortes E, Miliotto A, et al. Induction of cell death in ovarian cancer cells by doxorubicin and oncolytic vaccinia virus is associated with CREB3L1 activation. Mol Ther Oncolytics. 2021;23:38-50.Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology. 2014;3:e28518.Schaer DA, Geeganage S, Amaladas N, Lu ZH, Rasmussen ER, Sonyi A, et al. The Folate Pathway Inhibitor Pemetrexed Pleiotropically Enhances Effects of Cancer Immunotherapy. Clin Cancer Res. 2019;25(23):7175-88.Lau TS, Chan LKY, Man GCW, Wong CH, Lee JHS, Yim SF, et al. Paclitaxel Induces Immunogenic Cell Death in Ovarian Cancer via TLR4/IKK2/SNARE-Dependent Exocytosis. Cancer Immunol Res. 2020;8(8):1099-111.Arandjelovic S, Ravichandran KS. Phagocytosis of apoptotic cells in homeostasis. Nat Immunol. 2015;16(9):907-17.Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-Ullrich JE, et al. Cellsurface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell. 2005;123(2):321-34.Liu X, Li J, Liu Y, Ding J, Tong Z, Liu Y, et al. Calreticulin acts as an adjuvant to promote dendritic cell maturation and enhances antigen-specific cytotoxic T lymphocyte responses against non-small cell lung cancer cells. Cell Immunol. 2016;300:46-53.Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology. 2014;3(9):e955691.Universidad Nacional de ColombiaMinisterio de Ciencia Tecnología e InnovaciónEstudiantesInvestigadoresLICENSElicense.txtlicense.txttext/plain; charset=utf-85879https://repositorio.unal.edu.co/bitstream/unal/86203/1/license.txteb34b1cf90b7e1103fc9dfd26be24b4aMD51ORIGINAL1026272802.2024.pdf1026272802.2024.pdfTesis de Maestría en Inmunologíaapplication/pdf3143082https://repositorio.unal.edu.co/bitstream/unal/86203/2/1026272802.2024.pdf6f54f9ece62a46be6843d08a1742e2a2MD52THUMBNAIL1026272802.2024.pdf.jpg1026272802.2024.pdf.jpgGenerated Thumbnailimage/jpeg4460https://repositorio.unal.edu.co/bitstream/unal/86203/3/1026272802.2024.pdf.jpg8aad89a8c243229ee65c6be73b9238c9MD53unal/86203oai:repositorio.unal.edu.co:unal/862032024-06-04 23:04:31.827Repositorio Institucional Universidad Nacional de Colombiarepositorio_nal@unal.edu.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