1
|
Middelburg J, Schaap G, Sluijter M, Lloyd K, Ovcinnikovs V, Schuurman J, van der Burg SH, Kemper K, van Hall T. Cancer vaccines compensate for the insufficient induction of protective tumor-specific immunity of CD3 bispecific antibody therapy. J Immunother Cancer 2025; 13:e010331. [PMID: 39800374 PMCID: PMC11749218 DOI: 10.1136/jitc-2024-010331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/23/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND CD3 bispecific antibody (CD3 bsAb) therapy has become an established treatment modality for some cancer types and exploits endogenous T cells irrespective of their specificity. However, durable clinical responses are hampered by immune escape through loss of tumor target antigen expression. Induction of long-lasting tumor-specific immunity might therefore improve therapeutic efficacy, but has not been studied in detail yet for CD3 bsAbs. Here, we examined multiple combination strategies aiming to improve survival rates in solid tumors and, simultaneously, install endogenous immunity capable of protection to tumor rechallenge. METHODS Two syngeneic mouse tumor models were employed: The immunologically "cold" B16F10 melanoma and the immunologically "hot" MC38.TRP1 colon carcinoma model. Mice were treated with CD3xTRP1 bsAb (murine Fc-inert immunoglobulin G2a) as monotherapy, or in combination with agonistic costimulatory antibodies, Fc-active tumor-opsonizing antibodies, or tumor-(non)specific vaccines. Treatment efficacy of primary tumors and protection from rechallenge was monitored, as well as induction of tumor-specific T-cell responses. RESULTS In the immunologically "cold" B16F10 model, all combination therapies improved antitumor activity compared with CD3 bsAb monotherapy and induced systemic tumor-specific T-cell responses. However, this endogenous T-cell immunity swiftly waned and failed to protect mice from subsequent tumor rechallenge, except for combination therapy with tumor-specific vaccination. These vaccines strongly improved the therapeutic efficacy of CD3 bsAb against primary tumors and led to long-term immunological protection. In the immunologically "hot" MC38.TRP1 model, CD3 bsAb combined with only the vaccine adjuvant was sufficient to generate protective T-cell immunity and, moreover, prevented tumor escape via antigen loss. CONCLUSIONS These results demonstrate the impact of tumor antigenicity on the induction of protective endogenous antitumor immunity during CD3 bsAb treatment and, importantly, show that the combination with tumor-specific vaccines improves therapeutic efficacy and installs long-term immunological memory in both "hot" and "cold" tumors.
Collapse
Affiliation(s)
- Jim Middelburg
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Gaby Schaap
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjolein Sluijter
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Katy Lloyd
- Genmab BV, Utrecht, Utrecht, The Netherlands
| | | | | | - Sjoerd H van der Burg
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Thorbald van Hall
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Middelburg J, Ghaffari S, Schoufour TAW, Sluijter M, Schaap G, Göynük B, Sala BM, Al-Tamimi L, Scheeren F, Franken KLMC, Akkermans JJLL, Cabukusta B, Joosten SA, Derksen I, Neefjes J, van der Burg SH, Achour A, Wijdeven RHM, Weidanz J, van Hall T. The MHC-E peptide ligands for checkpoint CD94/NKG2A are governed by inflammatory signals, whereas LILRB1/2 receptors are peptide indifferent. Cell Rep 2023; 42:113516. [PMID: 38048225 DOI: 10.1016/j.celrep.2023.113516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/23/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023] Open
Abstract
The immune checkpoint NKG2A/CD94 is a promising target for cancer immunotherapy, and its ligand major histocompatibility complex E (MHC-E) is frequently upregulated in cancer. NKG2A/CD94-mediated inhibition of lymphocytes depends on the presence of specific leader peptides in MHC-E, but when and where they are presented in situ is unknown. We apply a nanobody specific for the Qdm/Qa-1b complex, the NKG2A/CD94 ligand in mouse, and find that presentation of Qdm peptide depends on every member of the endoplasmic reticulum-resident peptide loading complex. With a turnover rate of 30 min, the Qdm peptide reflects antigen processing capacity in real time. Remarkably, Qdm/Qa-1b complexes require inflammatory signals for surface expression in situ, despite the broad presence of Qa-1b molecules in homeostasis. Furthermore, we identify LILRB1 as a functional inhibition receptor for MHC-E in steady state. These data provide a molecular understanding of NKG2A blockade in immunotherapy and assign MHC-E as a convergent ligand for multiple immune checkpoints.
Collapse
Affiliation(s)
- Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Soroush Ghaffari
- Department of Biology, College of Science, The University of Texas at Arlington, Arlington, TX, USA
| | - Tom A W Schoufour
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Marjolein Sluijter
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Gaby Schaap
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Büsra Göynük
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Benedetta M Sala
- Science for Life Laboratory, Department of Medicine, Karolinska Institute & Division of Infectious Diseases, Karolinska University Hospital, 171 65 Solna, Sweden
| | - Lejla Al-Tamimi
- Science for Life Laboratory, Department of Medicine, Karolinska Institute & Division of Infectious Diseases, Karolinska University Hospital, 171 65 Solna, Sweden
| | - Ferenc Scheeren
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Jimmy J L L Akkermans
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Birol Cabukusta
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Ian Derksen
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine, Karolinska Institute & Division of Infectious Diseases, Karolinska University Hospital, 171 65 Solna, Sweden
| | - Ruud H M Wijdeven
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Jon Weidanz
- Abexxa Biologics, Inc., Arlington, TX, USA; College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, USA
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
3
|
Kumar S, Schoonderwoerd MJA, Kroonen JS, de Graaf IJ, Sluijter M, Ruano D, González-Prieto R, Verlaan-de Vries M, Rip J, Arens R, de Miranda NFCC, Hawinkels LJAC, van Hall T, Vertegaal ACO. Targeting pancreatic cancer by TAK-981: a SUMOylation inhibitor that activates the immune system and blocks cancer cell cycle progression in a preclinical model. Gut 2022; 71:2266-2283. [PMID: 35074907 PMCID: PMC9554032 DOI: 10.1136/gutjnl-2021-324834] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 12/29/2021] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) has the characteristics of high-density desmoplastic stroma, a distinctive immunosuppressive microenvironment and is profoundly resistant to all forms of chemotherapy and immunotherapy, leading to a 5-year survival rate of 9%. Our study aims to add novel small molecule therapeutics for the treatment of PDAC. DESIGN We have studied whether TAK-981, a novel highly selective and potent small molecule inhibitor of the small ubiquitin like modifier (SUMO) activating enzyme E1 could be used to treat a preclinical syngeneic PDAC mouse model and we have studied the mode of action of TAK-981. RESULTS We found that SUMOylation, a reversible post-translational modification required for cell cycle progression, is increased in PDAC patient samples compared with normal pancreatic tissue. TAK-981 decreased SUMOylation in PDAC cells at the nanomolar range, thereby causing a G2/M cell cycle arrest, mitotic failure and chromosomal segregation defects. TAK-981 efficiently limited tumour burden in the KPC3 syngeneic mouse model without evidence of systemic toxicity. In vivo treatment with TAK-981 enhanced the proportions of activated CD8 T cells and natural killer (NK) cells but transiently decreased B cell numbers in tumour, peripheral blood, spleen and lymph nodes. Single cell RNA sequencing revealed activation of the interferon response on TAK-981 treatment in lymphocytes including T, B and NK cells. TAK-981 treatment of CD8 T cells ex vivo induced activation of STAT1 and interferon target genes. CONCLUSION Our findings indicate that pharmacological inhibition of the SUMO pathway represents a potential strategy to target PDAC via a dual mechanism: inhibiting cancer cell cycle progression and activating anti-tumour immunity by inducing interferon signalling.
Collapse
Affiliation(s)
- Sumit Kumar
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jessie S Kroonen
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ilona J de Graaf
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dina Ruano
- Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Román González-Prieto
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jasper Rip
- Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ramon Arens
- Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alfred C O Vertegaal
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
4
|
Agathangelidis A, Vlachonikola E, Davi F, Langerak AW, Chatzidimitriou A. High-Throughput immunogenetics for precision medicine in cancer. Semin Cancer Biol 2021; 84:80-88. [PMID: 34757183 DOI: 10.1016/j.semcancer.2021.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 01/20/2023]
Abstract
Cancer is characterized by an extremely complex biological background, which hinders personalized therapeutic interventions. Precision medicine promises to overcome this obstacle through integrating information from different 'subsystems', including the host, the external environment, the tumor itself and the tumor micro-environment. Immunogenetics is an essential tool that allows dissecting both lymphoid cancer ontogeny at both a cell-intrinsic and a cell-extrinsic level, i.e. through characterizing micro-environmental interactions, with a view to precision medicine. This is particularly thanks to the introduction of powerful, high-throughput approaches i.e. next generation sequencing, which allow the comprehensive characterization of immune repertoires. Indeed, NGS immunogenetic analysis (Immune-seq) has emerged as key to both understanding cancer pathogenesis and improving the accuracy of clinical decision making in oncology. Immune-seq has applications in lymphoid malignancies, assisting in the diagnosis e.g. through differentiating from reactive conditions, as well as in disease monitoring through accurate assessment of minimal residual disease. Moreover, Immune-seq facilitates the study of T cell receptor clonal dynamics in critical clinical contexts, including transplantation as well as innovative immunotherapy for solid cancers. The clinical utility of Immune-seq represents the focus of the present contribution, where we highlight what can be achieved but also what must be addressed in order to maximally realize the promise of Immune-seq in precision medicine in cancer.
Collapse
Affiliation(s)
- Andreas Agathangelidis
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece; Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Elisavet Vlachonikola
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece; Department of Genetics and Molecular Biology, Faculty of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Frederic Davi
- Department of Hematology, APHP, Hôpital Pitié-Salpêtrière and Sorbonne University, Paris, France
| | - Anton W Langerak
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, the Netherlands
| | - Anastasia Chatzidimitriou
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75236, Sweden.
| |
Collapse
|
5
|
Hamblin MR, Abrahamse H. Factors Affecting Photodynamic Therapy and Anti-Tumor Immune Response. Anticancer Agents Med Chem 2021; 21:123-136. [PMID: 32188394 DOI: 10.2174/1871520620666200318101037] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/15/2020] [Accepted: 01/29/2020] [Indexed: 11/22/2022]
Abstract
Photodynamic Therapy (PDT) is a cancer therapy involving the systemic injection of a Photosensitizer (PS) that localizes to some extent in a tumor. After an appropriate time (ranging from minutes to days), the tumor is irradiated with red or near-infrared light either as a surface spot or by interstitial optical fibers. The PS is excited by the light to form a long-lived triplet state that can react with ambient oxygen to produce Reactive Oxygen Species (ROS) such as singlet oxygen and/or hydroxyl radicals, that kill tumor cells, destroy tumor blood vessels, and lead to tumor regression and necrosis. It has long been realized that in some cases, PDT can also stimulate the host immune system, leading to a systemic anti-tumor immune response that can also destroy distant metastases and guard against tumor recurrence. The present paper aims to cover some of the factors that can affect the likelihood and efficiency of this immune response. The structure of the PS, drug-light interval, rate of light delivery, mode of cancer cell death, expression of tumor-associated antigens, and combinations of PDT with various adjuvants all can play a role in stimulating the host immune system. Considering the recent revolution in tumor immunotherapy triggered by the success of checkpoint inhibitors, it appears that the time is ripe for PDT to be investigated in combination with other approaches in clinical scenarios.
Collapse
Affiliation(s)
- Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| |
Collapse
|
6
|
León-Letelier RA, Castro-Medina DI, Badillo-Godinez O, Tepale-Segura A, Huanosta-Murillo E, Aguilar-Flores C, De León-Rodríguez SG, Mantilla A, Fuentes-Pananá EM, López-Macías C, Bonifaz LC. Induction of Progenitor Exhausted Tissue-Resident Memory CD8 + T Cells Upon Salmonella Typhi Porins Adjuvant Immunization Correlates With Melanoma Control and Anti-PD-1 Immunotherapy Cooperation. Front Immunol 2020; 11:583382. [PMID: 33240271 PMCID: PMC7682137 DOI: 10.3389/fimmu.2020.583382] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/14/2020] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy has improved the clinical response in melanoma patients, although a relevant percentage of patients still cannot be salvaged. The search for the immune populations that provide the best tumor control and that can be coaxed by immunotherapy strategies is a hot topic in cancer research nowadays. Tumor-infiltrating TCF-1+ progenitor exhausted CD8+ T cells seem to grant the best melanoma prognosis and also efficiently respond to anti-PD-1 immunotherapy, giving rise to a TIM-3+ terminally exhausted population with heightened effector activity. We tested Porins from Salmonella Typhi as a pathogen associated molecular pattern adjuvant of natural or model antigen in prophylactic and therapeutic immunization approaches against murine melanoma. Porins induced protection against melanomas, even upon re-challenging of tumor-free mice. Porins efficiently expanded IFN-γ-producing CD8+ T cells and induced central and effector memory in lymph nodes and tissue-resident (Trm) T cells in the skin and tumors. Porins induced TCF-1+ PD-1+ CD8+ Trm T cells in the tumor stroma and the presence of this population correlated with melanoma growth protection in mice. Porins immunization also cooperated with anti-PD-1 immunotherapy to hamper melanoma growth. Importantly, the potentially protective Trm populations induced by Porins in the murine model were also observed in melanoma patients in which their presence also correlated with disease control. Our data support the use of cancer vaccination to sculpt the tumor stroma with efficient and lasting Trm T cells with effector activities, highlighting the use of Porins as an adjuvant. Furthermore, our data place CD8+ Trm T cells with a progenitor exhausted phenotype as an important population for melanoma control, either independently or in cooperation with anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Ricardo A León-Letelier
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.,Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Daniel I Castro-Medina
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Oscar Badillo-Godinez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Araceli Tepale-Segura
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Enrique Huanosta-Murillo
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Cristina Aguilar-Flores
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Saraí G De León-Rodríguez
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandra Mantilla
- Servicio de Patología, Hospital de Oncología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Ezequiel M Fuentes-Pananá
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Laura C Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
7
|
van Montfoort N, Borst L, Korrer MJ, Sluijter M, Marijt KA, Santegoets SJ, van Ham VJ, Ehsan I, Charoentong P, André P, Wagtmann N, Welters MJP, Kim YJ, Piersma SJ, van der Burg SH, van Hall T. NKG2A Blockade Potentiates CD8 T Cell Immunity Induced by Cancer Vaccines. Cell 2018; 175:1744-1755.e15. [PMID: 30503208 PMCID: PMC6354585 DOI: 10.1016/j.cell.2018.10.028] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/14/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023]
Abstract
Tumor-infiltrating CD8 T cells were found to frequently express the inhibitory receptor NKG2A, particularly in immune-reactive environments and after therapeutic cancer vaccination. High-dimensional cluster analysis demonstrated that NKG2A marks a unique immune effector subset preferentially co-expressing the tissue-resident CD103 molecule, but not immune checkpoint inhibitors. To examine whether NKG2A represented an adaptive resistance mechanism to cancer vaccination, we blocked the receptor with an antibody and knocked out its ligand Qa-1b, the conserved ortholog of HLA-E, in four mouse tumor models. The impact of therapeutic vaccines was greatly potentiated by disruption of the NKG2A/Qa-1b axis even in a PD-1 refractory mouse model. NKG2A blockade therapy operated through CD8 T cells, but not NK cells. These findings indicate that NKG2A-blocking antibodies might improve clinical responses to therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Nadine van Montfoort
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Linda Borst
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Michael J Korrer
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Koen A Marijt
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Saskia J Santegoets
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Vanessa J van Ham
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Ilina Ehsan
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Pornpimol Charoentong
- Department of Medical Oncology, National Center for Tumor diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | | | | | - Marij J P Welters
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Young J Kim
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sytse J Piersma
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands.
| |
Collapse
|
8
|
Benonisson H, Altıntaş I, Sluijter M, Verploegen S, Labrijn AF, Schuurhuis DH, Houtkamp MA, Verbeek JS, Schuurman J, van Hall T. CD3-Bispecific Antibody Therapy Turns Solid Tumors into Inflammatory Sites but Does Not Install Protective Memory. Mol Cancer Ther 2018; 18:312-322. [PMID: 30381448 DOI: 10.1158/1535-7163.mct-18-0679] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/01/2018] [Accepted: 10/24/2018] [Indexed: 11/16/2022]
Abstract
Immunotherapy of cancer with CD3-targeting bispecific antibodies (CD3 bsAb) is a fast developing field, and multiple tumor-associated antigens (TAA) are evaluated for hematologic and solid malignancies. The efficacy of these CD3 bsAb is usually examined in xenograft mouse tumor models with human T cells or in genetically engineered mouse models, where human TAA are introduced. These models often fail to fully recapitulate the natural tumor environment, especially for solid cancers, because of interspecies differences. Here, we investigated the systemic and intratumoral effects of a mouse CD3 bsAb in a fully immune-competent mouse melanoma model. Systemic administration of 0.5 mg/kg antibody induced a brief overall T-cell activation that was selectively sustained in the tumor microenvironment for several days. A fast subsequent influx of inflammatory macrophages into the tumor microenvironment was observed, followed by an increase in the number of CD4+ and CD8+ T cells. Although the capacity to directly kill melanoma cells in vitro was very modest, optimal tumor elimination was observed in vivo, even in the absence of CD8+ T cells, implying a redundancy in T-cell subsets for therapeutic efficacy. Finally, we took advantage of the full immune competence of our mouse model and tested immune memory induction. Despite a strong initial immunity against melanoma, treatment with the CD3 bsAb did not install protective memory responses. The observed mechanisms of action revealed in this immune-competent mouse model might form a rational basis for combinatorial approaches.
Collapse
Affiliation(s)
- Hreinn Benonisson
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
9
|
Pilla L, Ferrone S, Maccalli C. Methods for improving the immunogenicity and efficacy of cancer vaccines. Expert Opin Biol Ther 2018; 18:765-784. [PMID: 29874943 PMCID: PMC8670419 DOI: 10.1080/14712598.2018.1485649] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Cancer vaccines represent one of the oldest immunotherapy strategies. A variety of tumor-associated antigens have been exploited to investigate their immunogenicity as well as multiple strategies for vaccine administration. These efforts have led to the development of several clinical trials in tumors with different histological origins to test the clinical efficacy of cancer vaccines. However, suboptimal clinical results have been reported mainly due to the lack of optimized strategies to induce strong and sustained systemic tumor antigen-specific immune responses. AREAS COVERED We provide an overview of different types of cancer vaccines that have been developed and used in the context of clinical studies. Moreover, we review different preclinical and clinical strategies pursued to enhance the immunogenicity, stability, and targeting at tumor site of cancer vaccines. EXPERT OPINION Additional and appropriate preclinical studies are warranted to optimize the immunogenicity and delivery of cancer vaccines. The appropriate choice of target antigens is challenging; however, the exploitation of neoantigens generated from somatic mutations of tumor cells represents a promising approach to target highly immunogenic tumor-specific antigens. Remarkably, the investigation of the combination of cancer vaccines with immunomodulating agents able to skew the tumor microenvironment from immunosuppressive to immunostimulating will dramatically improve their clinical efficacy.
Collapse
Affiliation(s)
- Lorenzo Pilla
- Medical Oncology Unit, San Gerardo Hospital, Monza, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Maccalli
- Clinical Research Center, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| |
Collapse
|
10
|
Adoptive cell therapy with CD4 + T helper 1 cells and CD8 + cytotoxic T cells enhances complete rejection of an established tumour, leading to generation of endogenous memory responses to non-targeted tumour epitopes. Clin Transl Immunology 2017; 6:e160. [PMID: 29114389 PMCID: PMC5671987 DOI: 10.1038/cti.2017.37] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 06/15/2017] [Accepted: 06/15/2017] [Indexed: 01/31/2023] Open
Abstract
The results of adoptive T-cell therapies (ACTs) are very encouraging and show clinical evidence that ACT can provide a cure for patients with metastatic disease. However, various response rates and long-term cancer remission have been observed in different ACT trials. The types of T cells, prior treatment with chemotherapy and co-administration of other immune-target therapies have been found to influence the efficacy of ACT. In this study, we investigate the ability of ACT using CD4+ T helper 1 (Th1) cells and CD8+ cytotoxic T lymphocytes (CTLs) to reject the growth of established B16-ovalbumin (OVA) melanoma. CD8+ CTLs were found to be the main effector T cells that mediated tumour regression. However, low tumour-free survival rates were observed in ACT with CD8+ CTLs only. Co-transferring CD4+ Th1 cells and CD8+ CTLs has been observed to induce a synergistic antitumour response, resulting in complete regression in 80% of the tumour-bearing mice. We also examined a prior Dacarbazine (DTIC) and after virus-like particle (VLP)-OVA vaccine treatment to enhance ACT, but no therapeutic benefit was observed during primary B16-OVA tumour growth. Nevertheless, the ACT-mediated antitumour response was able to generate memory responses to both B16-OVA and B16-gp33 tumours. VLP-OVA vaccination following ACT enhances the memory responses to tumours that express a heterogenic population of both B16-OVA and B16-gp33 cells; however, it abolished the memory response to tumours consisting of only gp33-expressing cells. These findings provide important information for designing therapeutic treatments for patients with metastatic disease and cancer relapse to achieve durable cancer remission.
Collapse
|
11
|
Calì B, Molon B, Viola A. Tuning cancer fate: the unremitting role of host immunity. Open Biol 2017; 7:rsob.170006. [PMID: 28404796 PMCID: PMC5413907 DOI: 10.1098/rsob.170006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/14/2017] [Indexed: 12/12/2022] Open
Abstract
Host immunity plays a central and complex role in dictating tumour progression. Solid tumours are commonly infiltrated by a large number of immune cells that dynamically interact with the surrounding microenvironment. At first, innate and adaptive immune cells successfully cooperate to eradicate microcolonies of transformed cells. Concomitantly, surviving tumour clones start to proliferate and harness immune responses by specifically hijacking anti-tumour effector mechanisms and fostering the accumulation of immunosuppressive immune cell subsets at the tumour site. This pliable interplay between immune and malignant cells is a relentless process that has been concisely organized in three different phases: elimination, equilibrium and escape. In this review, we aim to depict the distinct immune cell subsets and immune-mediated responses characterizing the tumour landscape throughout the three interconnected phases. Importantly, the identification of key immune players and molecules involved in the dynamic crosstalk between tumour and immune system has been crucial for the introduction of reliable prognostic factors and effective therapeutic protocols against cancers.
Collapse
Affiliation(s)
- B Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy .,Venetian Institute of Molecular Medicine, Padua, Italy
| | - B Molon
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine, Padua, Italy
| | - A Viola
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
12
|
Potential use of lymph node-derived HPV-specific T cells for adoptive cell therapy of cervical cancer. Cancer Immunol Immunother 2016; 65:1451-1463. [PMID: 27619514 PMCID: PMC5099359 DOI: 10.1007/s00262-016-1892-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022]
Abstract
Adoptive transfer of tumor-specific T cells, expanded from tumor-infiltrating lymphocytes or from peripheral blood, is a promising immunotherapeutic approach for the treatment of cancer. Here, we studied whether the tumor-draining lymph nodes (TDLN) of patients with human papillomavirus (HPV)-induced cervical cancer can be used as a source for ACT. The objectives were to isolate lymph node mononuclear cells (LNMC) from TDLN and optimally expand HPV-specific CD4+ and CD8+ T cells under clinical grade conditions. TDLN were isolated from 11 patients with early-stage cervical cancer during radical surgery. Isolated lymphocytes were expanded in the presence of HPV16 E6 and E7 clinical grade synthetic long peptides and IL-2 for 22 days and then analyzed for HPV16 specificity by proliferation assay, multiparameter flow cytometry and cytokine analysis as well as for CD25 and FoxP3 expression. Stimulation of LNMC resulted in expansion of polyclonal HPV-specific T cells in all patients. On average a 36-fold expansion of a CD4+ and/or CD8+ HPV16-specific T cell population was observed, which maintained its capacity for secondary expansion. The T helper type 1 cytokine IFNγ was produced in all cell cultures and in some cases also the Th2 cytokines IL-10 and IL-5. The procedure was highly reproducible, as evidenced by complete repeats of the stimulation procedures under research and under full good manufacturing practice conditions. In conclusion, TDLN represent a rich source of polyclonal HPV16 E6- and E7-specific T cells, which can be expanded under clinical grade conditions for adoptive immunotherapy in patients with cervical cancer.
Collapse
|
13
|
Redeker A, Arens R. Improving Adoptive T Cell Therapy: The Particular Role of T Cell Costimulation, Cytokines, and Post-Transfer Vaccination. Front Immunol 2016; 7:345. [PMID: 27656185 PMCID: PMC5011476 DOI: 10.3389/fimmu.2016.00345] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/24/2016] [Indexed: 12/22/2022] Open
Abstract
Adoptive cellular therapy (ACT) is a form of immunotherapy whereby antigen-specific T cells are isolated or engineered, expanded ex vivo, and transferred back to patients. Clinical benefit after ACT has been obtained in treatment of infection, various hematological malignancies, and some solid tumors; however, due to poor functionality and persistence of the transferred T cells, the efficacy of ACT in the treatment of most solid tumors is often marginal. Hence, much effort is undertaken to improve T cell function and persistence in ACT and significant progress is being made. Herein, we will review strategies to improve ACT success rates in the treatment of cancer and infection. We will deliberate on the most favorable phenotype for the tumor-specific T cells that are infused into patients and on how to obtain T cells bearing this phenotype by applying novel ex vivo culture methods. Moreover, we will discuss T cell function and persistence after transfer into patients and how these factors can be manipulated by means of providing costimulatory signals, cytokines, blocking antibodies to inhibitory molecules, and vaccination. Incorporation of these T cell stimulation strategies and combinations of the different treatment modalities are likely to improve clinical response rates further.
Collapse
Affiliation(s)
- Anke Redeker
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden , Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
14
|
van der Burg SH, Arens R, Ossendorp F, van Hall T, Melief CJM. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer 2016; 16:219-33. [PMID: 26965076 DOI: 10.1038/nrc.2016.16] [Citation(s) in RCA: 510] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Therapeutic vaccines preferentially stimulate T cells against tumour-specific epitopes that are created by DNA mutations or oncogenic viruses. In the setting of premalignant disease, carcinoma in situ or minimal residual disease, therapeutic vaccination can be clinically successful as monotherapy; however, in established cancers, therapeutic vaccines will require co-treatments to overcome immune evasion and to become fully effective. In this Review, we discuss the progress that has been made in overcoming immune evasion controlled by tumour cell-intrinsic factors and the tumour microenvironment. We summarize how therapeutic benefit can be maximized in patients with established cancers by improving vaccine design and by using vaccines to increase the effects of standard chemotherapies, to establish and/or maintain tumour-specific T cells that are re-energized by checkpoint blockade and other therapies, and to sustain the antitumour response of adoptively transferred T cells.
Collapse
Affiliation(s)
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | - Cornelis J M Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- ISA Pharmaceuticals, J. H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| |
Collapse
|
15
|
van Poelgeest MIE, Welters MJP, Vermeij R, Stynenbosch LFM, Loof NM, Berends-van der Meer DMA, Löwik MJG, Hamming ILE, van Esch EMG, Hellebrekers BWJ, van Beurden M, Schreuder HW, Kagie MJ, Trimbos JBMZ, Fathers LM, Daemen T, Hollema H, Valentijn ARPM, Oostendorp J, Oude Elberink JHNG, Fleuren GJ, Bosse T, Kenter GG, Stijnen T, Nijman HW, Melief CJM, van der Burg SH. Vaccination against Oncoproteins of HPV16 for Noninvasive Vulvar/Vaginal Lesions: Lesion Clearance Is Related to the Strength of the T-Cell Response. Clin Cancer Res 2016; 22:2342-50. [PMID: 26813357 DOI: 10.1158/1078-0432.ccr-15-2594] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/03/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Therapeutic vaccination with human papillomavirus type 16 (HPV16) E6 and E7 synthetic long peptides (SLP) is effective against HPV16-induced high-grade vulvar and vaginal intraepithelial neoplasia (VIN/VaIN). However, clinical nonresponders displayed weak CD8(+) T-cell reactivity. Here, we studied if imiquimod applied at the vaccine site could improve CD8(+) T-cell reactivity, clinical efficacy, and safety of HPV16-SLP (ISA101). EXPERIMENTAL DESIGN A multicenter open-label, randomized controlled trial was conducted in patients with HPV16(+) high-grade VIN/VaIN. Patients received ISA101 vaccination with or without application of 5% imiquimod at the vaccine site. The primary objective was the induction of a directly ex vivo detectable HPV16-specific CD8(+) T-cell response. The secondary objectives were clinical responses (lesion size, histology, and virology) and their relation with the strength of vaccination-induced immune responses. RESULTS Forty-three patients were assigned to either ISA101 with imiquimod (n = 21) or ISA101 only (n = 22). Imiquimod did not improve the outcomes of vaccination. However, vaccine-induced clinical responses were observed in 18 of 34 (53%; 95% CI, 35.1-70.2) patients at 3 months and in 15 of 29 (52%; 95% CI, 32.5-70.6) patients, 8 of whom displayed a complete histologic response, at 12 months after the last vaccination. All patients displayed vaccine-induced T-cell responses, which were significantly stronger in patients with complete responses. Importantly, viral clearance occurred in all but one of the patients with complete histologic clearance. CONCLUSIONS This new study confirms that clinical efficacy of ISA101 vaccination is related to the strength of vaccine-induced HPV16-specific T-cell immunity and is an effective therapy for HPV16-induced high-grade VIN/VaIN. Clin Cancer Res; 22(10); 2342-50. ©2016 AACRSee related commentary by Karaki et al., p. 2317.
Collapse
Affiliation(s)
| | - Marij J P Welters
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Renee Vermeij
- Department of Gynaecology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Linda F M Stynenbosch
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Nikki M Loof
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Margriet J G Löwik
- Department of Gynaecology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Ineke L E Hamming
- Department of Gynaecology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Edith M G van Esch
- Department of Gynaecology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Bart W J Hellebrekers
- Department of Obstetrics and Gynaecology, Haga Teaching Hospital, the Hague, the Netherlands
| | | | - Henk W Schreuder
- Department of Obstetrics and Gynaecology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Marjolein J Kagie
- Department of Obstetrics and Gynaecology, Medical Centre Haaglanden, the Hague, the Netherlands
| | - J Baptist M Z Trimbos
- Department of Gynaecology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Lorraine M Fathers
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Toos Daemen
- Department of Medical Microbiology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Harry Hollema
- Department of Pathology and Medical Biology, University Medical Centre Groningen, Groningen, the Netherlands
| | - A Rob P M Valentijn
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jaap Oostendorp
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - J Hanneke N G Oude Elberink
- Department of Allergy, Internal Medicine, Groningen Research Institute for Asthma and COPD, University Medical Centre Groningen, Groningen, the Netherlands
| | - Gertjan J Fleuren
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Gemma G Kenter
- Department of Gynaecology, Academic Medical Centre, Amsterdam, the Netherlands
| | - Theo Stijnen
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Centre, Leiden, the Netherlands
| | - Hans W Nijman
- Department of Gynaecology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Cornelis J M Melief
- ISA Pharmaceuticals, Leiden, the Netherlands. Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
16
|
Hofmann S, Mead A, Malinovskis A, Hardwick NR, Guinn BA. Analogue peptides for the immunotherapy of human acute myeloid leukemia. Cancer Immunol Immunother 2015; 64:1357-67. [PMID: 26438084 PMCID: PMC11029593 DOI: 10.1007/s00262-015-1762-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 09/27/2015] [Indexed: 12/14/2022]
Abstract
The use of peptide vaccines, enhanced by adjuvants, has shown some efficacy in clinical trials. However, responses are often short-lived and rarely induce notable memory responses. The reason is that self-antigens have already been presented to the immune system as the tumor develops, leading to tolerance or some degree of host tumor cell destruction. To try to break tolerance against self-antigens, one of the methods employed has been to modify peptides at the anchor residues to enhance their ability to bind major histocompatibility complex molecules, extending their exposure to the T-cell receptor. These modified or analogue peptides have been investigated as stimulators of the immune system in patients with different cancers with variable but sometimes notable success. In this review we describe the background and recent developments in the use of analogue peptides for the immunotherapy of acute myeloid leukemia describing knowledge useful for the application of analogue peptide treatments for other malignancies.
Collapse
Affiliation(s)
- Susanne Hofmann
- Third Clinic for Internal Medicine, University of Ulm, Ulm, Germany
| | - Andrew Mead
- Department of Life Sciences, University of Bedfordshire, Park Square, Luton, LU1 3JU, UK
| | - Aleksandrs Malinovskis
- Department of Life Sciences, University of Bedfordshire, Park Square, Luton, LU1 3JU, UK
| | - Nicola R Hardwick
- Division of Translational Vaccine Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
- Department of Haematological Medicine, Guy's, King's & St. Thomas' School of Medicine, The Rayne Institute, King's College London, 123 Coldharbour Lane, London, UK
| | - Barbara-Ann Guinn
- Department of Life Sciences, University of Bedfordshire, Park Square, Luton, LU1 3JU, UK.
- Department of Haematological Medicine, Guy's, King's & St. Thomas' School of Medicine, The Rayne Institute, King's College London, 123 Coldharbour Lane, London, UK.
- Cancer Sciences Unit, Southampton University Hospitals Trust, University of Southampton, Southampton, UK.
| |
Collapse
|
17
|
Garapaty A, Champion JA. Biomimetic and synthetic interfaces to tune immune responses. Biointerphases 2015; 10:030801. [PMID: 26178262 PMCID: PMC4506308 DOI: 10.1116/1.4922798] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/06/2015] [Accepted: 06/10/2015] [Indexed: 01/05/2023] Open
Abstract
Organisms depend upon complex intercellular communication to initiate, maintain, or suppress immune responses during infection or disease. Communication occurs not only between different types of immune cells, but also between immune cells and nonimmune cells or pathogenic entities. It can occur directly at the cell-cell contact interface, or indirectly through secreted signals that bind cell surface molecules. Though secreted signals can be soluble, they can also be particulate in nature and direct communication at the cell-particle interface. Secreted extracellular vesicles are an example of native particulate communication, while viruses are examples of foreign particulates. Inspired by communication at natural immunological interfaces, biomimetic materials and designer molecules have been developed to mimic and direct the type of immune response. This review describes the ways in which native, biomimetic, and designer materials can mediate immune responses. Examples include extracellular vesicles, particles that mimic immune cells or pathogens, and hybrid designer molecules with multiple signaling functions, engineered to target and bind immune cell surface molecules. Interactions between these materials and immune cells are leading to increased understanding of natural immune communication and function, as well as development of immune therapeutics for the treatment of infection, cancer, and autoimmune disease.
Collapse
Affiliation(s)
- Anusha Garapaty
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Dr. NW, Atlanta, Georgia 30332
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Dr. NW, Atlanta, Georgia 30332
| |
Collapse
|
18
|
Anzengruber F, Avci P, de Freitas LF, Hamblin MR. T-cell mediated anti-tumor immunity after photodynamic therapy: why does it not always work and how can we improve it? Photochem Photobiol Sci 2015; 14:1492-1509. [PMID: 26062987 PMCID: PMC4547550 DOI: 10.1039/c4pp00455h] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Photodynamic therapy (PDT) uses the combination of non-toxic photosensitizers and harmless light to generate reactive oxygen species that destroy tumors by a combination of direct tumor cell killing, vascular shutdown, and activation of the immune system. It has been shown in some animal models that mice that have been cured of cancer by PDT, may exhibit resistance to rechallenge. The cured mice can also possess tumor specific T-cells that recognize defined tumor antigens, destroy tumor cells in vitro, and can be adoptively transferred to protect naïve mice from cancer. However, these beneficial outcomes are the exception rather than the rule. The reasons for this lack of consistency lie in the ability of many tumors to suppress the host immune system and to actively evade immune attack. The presence of an appropriate tumor rejection antigen in the particular tumor cell line is a requisite for T-cell mediated immunity. Regulatory T-cells (CD25+, Foxp3+) are potent inhibitors of anti-tumor immunity, and their removal by low dose cyclophosphamide can potentiate the PDT-induced immune response. Treatments that stimulate dendritic cells (DC) such as CpG oligonucleotide can overcome tumor-induced DC dysfunction and improve PDT outcome. Epigenetic reversal agents can increase tumor expression of MHC class I and also simultaneously increase expression of tumor antigens. A few clinical reports have shown that anti-tumor immunity can be generated by PDT in patients, and it is hoped that these combination approaches may increase tumor cures in patients.
Collapse
Affiliation(s)
- Florian Anzengruber
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Pinar Avci
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Dermatooncology and Venerology, Semmelweis University School of Medicine, Budapest, 1085, Hungary
| | - Lucas Freitas de Freitas
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Programa de Pos Graduacao Interunidades Bioengenharia – USP – Sao Carlos, Brazil
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Correspondence to: Michael R Hamblin, PhD, Wellman Center for Photomedicine, Massachusetts General Hospital, 50 Blossom Street, Boston, MA 02114, USA.
| |
Collapse
|
19
|
Obeid J, Hu Y, Slingluff CL. Vaccines, Adjuvants, and Dendritic Cell Activators--Current Status and Future Challenges. Semin Oncol 2015; 42:549-61. [PMID: 26320060 DOI: 10.1053/j.seminoncol.2015.05.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cancer vaccines offer a low-toxicity approach to induce anticancer immune responses. They have shown promise for clinical benefit with one cancer vaccine approved in the United States for advanced prostate cancer. As other immune therapies are now clearly effective for treatment of advanced cancers of many histologies, there is renewed enthusiasm for optimizing cancer vaccines for use to prevent recurrence in early-stage cancers and/or to combine with other immune therapies for therapy of advanced cancers. Future advancements in vaccine therapy will involve the identification and selection of effective antigen formulations, optimization of adjuvants, dendritic cell (DC) activation, and combination therapies. In this summary we present the current practice, the broad collection of challenges, and the promising future directions of vaccine therapy for cancer.
Collapse
Affiliation(s)
- Joseph Obeid
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Yinin Hu
- Department of Surgery, University of Virginia, Charlottesville, VA
| | | |
Collapse
|
20
|
Cannas G, Thomas X. Supportive care in patients with acute leukaemia: historical perspectives. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2015; 13:205-20. [PMID: 25369611 PMCID: PMC4385068 DOI: 10.2450/2014.0080-14] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 06/24/2014] [Indexed: 01/09/2023]
Affiliation(s)
- Giovanna Cannas
- Haemovigilance Unit, Edouard Herriot Hospital and Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France
| | - Xavier Thomas
- Leukaemia Unit, Haematology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
21
|
Garg AD, Dudek AM, Ferreira GB, Verfaillie T, Vandenabeele P, Krysko DV, Mathieu C, Agostinis P. ROS-induced autophagy in cancer cells assists in evasion from determinants of immunogenic cell death. Autophagy 2014; 9:1292-307. [DOI: 10.4161/auto.25399] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
22
|
Chung MJ, Park JY, Bang S, Park SW, Song SY. Phase II clinical trial of ex vivo-expanded cytokine-induced killer cells therapy in advanced pancreatic cancer. Cancer Immunol Immunother 2014; 63:939-946. [PMID: 24916038 PMCID: PMC11029701 DOI: 10.1007/s00262-014-1566-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/30/2014] [Indexed: 12/11/2022]
Abstract
Second-line chemotherapy in patients with gemcitabine-refractory advanced pancreatic cancer has shown disappointing survival outcomes due to rapid disease progression and performance deterioration. The aim of this phase II trial was to evaluate the efficacy and safety of adoptive immunotherapy using ex vivo-expanded, cytokine-induced killer (CIK) cells in gemcitabine-refractory advanced pancreatic cancer. Patients with advanced pancreatic cancer who showed disease progression during gemcitabine-based chemotherapy were enrolled in this study. For generation of CIK cells, peripheral blood samples were collected from each patient and cultured with anti-CD3 monoclonal antibody and IL-2. Patients received CIK cells intravenously 10 times, every week for 5 weeks and then every other week for 10 weeks. Twenty patients were enrolled between November 2009 and September 2010. The disease control rate was 25 % (4/16 patients). The median progression-free survival (PFS) was 11.0 weeks (95 % CI 8.8-13.2), and the median overall survival (OS) was 26.6 weeks (95 % CI 8.6-44.6). Grade 3 toxicities included general weakness in two patients and thrombocytopenia in one patient. Grade 4 hematologic or non-hematologic toxicity was not observed. Patients showed improvement in pancreatic pain, gastrointestinal distress, jaundice, body image alterations, altered bowel habits, health satisfaction, and sexuality when assessing quality of life (QoL). Adoptive immunotherapy using CIK cells showed comparable PFS and OS to survival data of previous trials that assessed conventional chemotherapies while maintaining tolerability and showing encouraging results in terms of patient QoL in gemcitabine-refractory advanced pancreatic cancer (clinicalTrials.gov number NCT00965718).
Collapse
Affiliation(s)
- Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei Institute of Gastroenterology, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 120-752 Republic of Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei Institute of Gastroenterology, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 120-752 Republic of Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Yonsei Institute of Gastroenterology, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 120-752 Republic of Korea
| | - Seung Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei Institute of Gastroenterology, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 120-752 Republic of Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Yonsei Institute of Gastroenterology, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 120-752 Republic of Korea
| |
Collapse
|
23
|
Sluijter M, van der Sluis TC, van der Velden PA, Versluis M, West BL, van der Burg SH, van Hall T. Inhibition of CSF-1R supports T-cell mediated melanoma therapy. PLoS One 2014; 9:e104230. [PMID: 25110953 PMCID: PMC4128661 DOI: 10.1371/journal.pone.0104230] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/11/2014] [Indexed: 12/25/2022] Open
Abstract
Tumor associated macrophages (TAM) can promote angiogenesis, invasiveness and immunosuppression. The cytokine CSF-1 (or M-CSF) is an important factor of TAM recruitment and differentiation and several pharmacological agents targeting the CSF-1 receptor (CSF-1R) have been developed to regulate TAM in solid cancers. We show that the kinase inhibitor PLX3397 strongly dampened the systemic and local accumulation of macrophages driven by B16F10 melanomas, without affecting Gr-1+ myeloid derived suppressor cells. Removal of intratumoral macrophages was remarkably efficient and a modest, but statistically significant, delay in melanoma outgrowth was observed. Importantly, CSF-1R inhibition strongly enhanced tumor control by immunotherapy using tumor-specific CD8 T cells. Elevated IFNγ production by T cells was observed in mice treated with the combination of PLX3397 and immunotherapy. These results support the combined use of CSF-1R inhibition with CD8 T cell immunotherapy, especially for macrophage-stimulating tumors.
Collapse
Affiliation(s)
- Marjolein Sluijter
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tetje C. van der Sluis
- Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Mieke Versluis
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - Brian L. West
- Plexxikon Inc., Berkeley, California, United States of America
| | - Sjoerd H. van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Thorbald van Hall
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
- * E-mail:
| |
Collapse
|
24
|
Sahdev P, Ochyl LJ, Moon JJ. Biomaterials for nanoparticle vaccine delivery systems. Pharm Res 2014; 31:2563-82. [PMID: 24848341 DOI: 10.1007/s11095-014-1419-y] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/12/2014] [Indexed: 01/11/2023]
Abstract
Subunit vaccination benefits from improved safety over attenuated or inactivated vaccines, but their limited capability to elicit long-lasting, concerted cellular and humoral immune responses is a major challenge. Recent studies have demonstrated that antigen delivery via nanoparticle formulations can significantly improve immunogenicity of vaccines due to either intrinsic immunostimulatory properties of the materials or by co-entrapment of molecular adjuvants such as Toll-like receptor agonists. These studies have collectively shown that nanoparticles designed to mimic biophysical and biochemical cues of pathogens offer new exciting opportunities to enhance activation of innate immunity and elicit potent cellular and humoral immune responses with minimal cytotoxicity. In this review, we present key research advances that were made within the last 5 years in the field of nanoparticle vaccine delivery systems. In particular, we focus on the impact of biomaterials composition, size, and surface charge of nanoparticles on modulation of particle biodistribution, delivery of antigens and immunostimulatory molecules, trafficking and targeting of antigen presenting cells, and overall immune responses in systemic and mucosal tissues. This review describes recent progresses in the design of nanoparticle vaccine delivery carriers, including liposomes, lipid-based particles, micelles and nanostructures composed of natural or synthetic polymers, and lipid-polymer hybrid nanoparticles.
Collapse
Affiliation(s)
- Preety Sahdev
- Department of Pharmaceutical Sciences, College of Pharmacy University of Michigan, 2800 Plymouth Road NCRC, Ann Arbor, Michigan, 48109, USA
| | | | | |
Collapse
|
25
|
Quakkelaar ED, Fransen MF, van Maren WWC, Vaneman J, Loof NM, van Heiningen SH, Verbeek JS, Ossendorp F, Melief CJM. IgG-mediated anaphylaxis to a synthetic long peptide vaccine containing a B cell epitope can be avoided by slow-release formulation. THE JOURNAL OF IMMUNOLOGY 2014; 192:5813-20. [PMID: 24813207 DOI: 10.4049/jimmunol.1302337] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Synthetic long peptides (SLP) are a promising vaccine modality to induce therapeutic T cell responses in patients with chronic infections and tumors. We studied different vaccine formulations in mice using SLP derived from carcinoembryonic Ag. We discovered that one of the SLP contains a linear Ab epitope in combination with a CD4 epitope. Repeated vaccination with this carcinoembryonic Ag SLP in mice shows improved T cell responses and simultaneously induced high titers of peptide-specific Abs. These Abs resulted in unexpected anaphylaxis after a third or subsequent vaccinations with the SLP when formulated in saline. Administration of low SLP doses in the slow-release vehicle IFA prevented the anaphylaxis after repeated vaccination. This study underscores both the immunogenicity of SLP vaccination, for inducing T cell as well as B cell responses, and the necessity of safe administration routes.
Collapse
Affiliation(s)
- Esther D Quakkelaar
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Wendy W C van Maren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Joost Vaneman
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Nikki M Loof
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Sandra H van Heiningen
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; and
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; and
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Cornelis J M Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; ISA Pharmaceuticals, 2333 CH Leiden, The Netherlands
| |
Collapse
|
26
|
Bot A, Marincola F, Smith KA. Repositioning therapeutic cancer vaccines in the dawning era of potent immune interventions. Expert Rev Vaccines 2013; 12:1219-34. [PMID: 24099049 DOI: 10.1586/14760584.2013.836908] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Based on lessons learned with various immune interventions, this review aims to provide a constructive framework for repositioning therapeutic cancer vaccination. Intensive research throughout the past decade has identified key hurdles interfering with the efficacy of cancer vaccines. The vaccination concept still holds promise if positioned appropriately in minimal residual disease and select early disease stage cancer indications. However, in advanced cancer, it must be integrated with complementary immune interventions to ensure reconstruction of a functional immune repertoire and simultaneous blockade of immune inhibiting mechanisms. Vaccination could render complex and integrative immune interventions simpler, safer and more effective. The near future will witness an explosion of activities in the cancer immunotherapy arena, witnessing a rational repositioning of vaccines rather than their extinction.
Collapse
|
27
|
Arens R, van Hall T, van der Burg SH, Ossendorp F, Melief CJM. Prospects of combinatorial synthetic peptide vaccine-based immunotherapy against cancer. Semin Immunol 2013; 25:182-90. [PMID: 23706598 DOI: 10.1016/j.smim.2013.04.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/10/2013] [Accepted: 04/19/2013] [Indexed: 01/15/2023]
Abstract
The insight that the immune system is involved in tumor resistance is gaining momentum and this has led to the development of immunotherapeutic strategies aiming at enhancement of immune-mediated tumor destruction. Although some of these strategies have moderate clinical benefit, most stand-alone therapies fail to significantly affect progressive disease and survival or do so only in a minority of patients. Research on the mechanisms underlying the generation of immune responses against tumors and the immune evasion by tumors has emphasized that various mechanisms simultaneously prevent effective immunity against cancer including inefficient presentation of tumor antigens by dendritic cells and induction of negative immune regulation by regulatory T-cells (Tregs) and myeloid derived suppressor cells (MDSCs). Thus the design of therapies that simultaneously improve effective tumor immunity and counteract immune evasion by tumors seems most desirable for clinical efficacy. As it is unlikely that a single immunotherapeutic strategy addresses all necessary requirements, combinatorial strategies that act synergistically need to be developed. Here we discuss the current knowledge and prospects of treatment with synthetic peptide vaccines that stimulate tumor-specific T-cell responses combined with adjuvants, immune modulating antibodies, cytokines and chemotherapy. We conclude that combinatorial approaches have the best potency to accomplish the most significant tumor destruction but further research is required to optimize such approaches.
Collapse
Affiliation(s)
- Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
28
|
Engels B, Engelhard VH, Sidney J, Sette A, Binder DC, Liu RB, Kranz DM, Meredith SC, Rowley DA, Schreiber H. Relapse or eradication of cancer is predicted by peptide-major histocompatibility complex affinity. Cancer Cell 2013; 23:516-26. [PMID: 23597565 PMCID: PMC3658176 DOI: 10.1016/j.ccr.2013.03.018] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 02/21/2013] [Accepted: 03/19/2013] [Indexed: 01/24/2023]
Abstract
Cancers often relapse after adoptive therapy, even though specific T cells kill cells from the same cancer efficiently in vitro. We found that tumor eradication by T cells required high affinities of the targeted peptides for major histocompatibility complex (MHC) class I. Affinities of at least 10 nM were required for relapse-free regression. Only high-affinity peptide-MHC interactions led to efficient cross-presentation of antigen, thereby stimulating cognate T cells to secrete cytokines. These findings highlight the importance of targeting peptides with high affinity for MHC class I when designing T cell-based immunotherapy.
Collapse
Affiliation(s)
- Boris Engels
- Department of Pathology, Committee on Immunology and Committee on Cancer Biology, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Hailemichael Y, Dai Z, Jaffarzad N, Ye Y, Medina MA, Huang XF, Dorta-Estremera SM, Greeley NR, Nitti G, Peng W, Liu C, Lou Y, Wang Z, Ma W, Rabinovich B, Sowell RT, Schluns KS, Davis RE, Hwu P, Overwijk WW. Persistent antigen at vaccination sites induces tumor-specific CD8⁺ T cell sequestration, dysfunction and deletion. Nat Med 2013; 19:465-72. [PMID: 23455713 PMCID: PMC3618499 DOI: 10.1038/nm.3105] [Citation(s) in RCA: 356] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 01/25/2013] [Indexed: 12/22/2022]
Abstract
To understand why cancer vaccine-induced T cells often fail to eradicate tumors, we studied immune responses in mice vaccinated with gp100 melanoma peptide in incomplete Freund’s adjuvant (IFA), commonly used in clinical cancer vaccine trials. Peptide/IFA vaccination primed tumor-specific CD8+ T cells, which accumulated not in tumors but at the persisting, antigen-rich vaccination site. Once there, primed T cells became dysfunctional and underwent antigen-driven, Interferon-γ (IFN-γ) and Fas ligand (FasL)-mediated apoptosis, resulting in hyporesponsiveness to subsequent vaccination. Provision of anti-CD40 antibody, Toll-like receptor 7 (TLR7) agonist and interleukin-2 (IL-2) reduced T cell apoptosis but did not prevent vaccination site sequestration. A non-persisting vaccine formulation shifted T cell localization towards tumors, inducing superior anti-tumor activity while reducing systemic T cell dysfunction and promoting memory formation. Persisting peptide/IFA vaccine depots can induce specific T cell sequestration, dysfunction and deletion at vaccination sites; short-lived formulations may overcome these limitations and result in greater therapeutic efficacy of peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Yared Hailemichael
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Yorty JL. Short-term corticosterone treatment decreases the early CD8+ T cell response to simian virus 40 tumor antigen but has no impact on the late CD8+ T cell response. Brain Behav Immun 2013; 28:139-48. [PMID: 23164951 DOI: 10.1016/j.bbi.2012.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/09/2012] [Accepted: 11/09/2012] [Indexed: 10/27/2022] Open
Abstract
CD8+ T cells (T(CD8)) help control tumor growth in vivo through recognition of distinct tumor antigens and cytolysis of tumor cells. The T(CD8) immune response in C57BL/6 mice to the Simian Virus 40 oncoprotein, large tumor antigen (Tag), targets multiple epitopes and is well-characterized. Epitope IV, an H-2K(b)-restricted epitope, is immunodominant while epitope I, an H-2D(b)-restricted epitope is subdominant. GCs alter many aspects of T cell function. Indeed, the current studies demonstrate that exposure of mice to the immunosuppressive GC, corticosterone (CORT), over the entire course of the primary immune response limits activation of endogenous Tag-specific T(CD8). Even short-term CORT treatment from day -1 to day +2 post-immunization significantly reduced splenic size and the absolute number of Tag-specific T(CD8) on day 6 post-immunization. In vivo killing activity was also reduced. However, by day 10 post-immunization, the peak of the immune response, the absolute number of Tag-specific T(CD8) and their in vivo killing of epitope I or epitope IV-expressing target cells had recovered in CORT treated mice. Adoptive transfer of transgenic T cells post-CORT removal demonstrated that CORT decreased the ability of the endogenous antigen-presenting cells to induce proliferation of the exogenous transgenic T cells. Combined, these studies have implications about the timing of clinical steroid treatment relative to immunization or adoptive transfer for cancer immunotherapy.
Collapse
Affiliation(s)
- Jodi L Yorty
- Department of Biology, Elizabethtown College, One Alpha Drive, Elizabethtown, PA 17022, United States.
| |
Collapse
|
31
|
Melief CJM. Selective activation of oxygen-deprived tumor-infiltrating lymphocytes through local intratumoral delivery of CD137 monoclonal antibodies. Cancer Discov 2012; 2:586-7. [PMID: 22787088 DOI: 10.1158/2159-8290.cd-12-0229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypoxia-inducing transcription factor-1α (HIF-1α) in hypoxic tumors induces the TNF receptor family member CD137 on tumor-infiltrating lymphocytes. This can be exploited for intratumoral low-dose injection of effective systemic immunotherapy with agonist CD137-specific monoclonal antibodies that induce circulation of systemic tumor-specific effector T cells capable of eradicating distant metastases.
Collapse
Affiliation(s)
- Cornelis J M Melief
- Immune System Activation, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
32
|
Ly LV, Sluijter M, van der Burg SH, Jager MJ, van Hall T. Effective cooperation of monoclonal antibody and peptide vaccine for the treatment of mouse melanoma. THE JOURNAL OF IMMUNOLOGY 2012. [PMID: 23203930 DOI: 10.4049/jimmunol.1200135] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
mAbs binding to tumor-associated surface Ags are therapeutically applied in a range of malignancies. Therapeutic vaccination only recently met with clinical success, and the first cancer vaccine received U.S. Food and Drug Administration approval last year. To improve current protocols, we combined peptide vaccines with mAb to the tyrosinase-related protein (TRP)-1 surface Ag for the treatment of B16F10 skin melanoma. Vaccine formulations with synthetic long peptides failed to elicit strong CD8 T cell responses to self-differentiation Ags gp100 and TRP-2, whereas altered peptide sequences recruited gp100-specific CD8 T cells from the endogenous repertoire with frequencies of 40%. However, these high frequencies were reached too late; large, progressively growing melanomas had already emerged. Addition of the TRP-1-directed mAb TA99 to the treatment protocol mediated eradication of s.c. lesions. The mode of action of the Ab did not depend on complement factor C3 and did not lead to improved Ag presentation and CD8 T cell immunity; rather, it recruited FcγR-bearing innate immune cells during early tumor control, thereby creating a window of time for the generation of protective cellular immunity. These data support the concept of combination therapy, in which passive transfer of mAbs is supplemented with cancer peptide vaccines. Moreover, we advocate that tumor Ag-specific T cell immunity directed against self-proteins can be exploited from the endogenous repertoire.
Collapse
Affiliation(s)
- Long V Ly
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
33
|
Stern PL, van der Burg SH, Hampson IN, Broker TR, Fiander A, Lacey CJ, Kitchener HC, Einstein MH. Therapy of human papillomavirus-related disease. Vaccine 2012; 30 Suppl 5:F71-82. [PMID: 23199967 PMCID: PMC4155500 DOI: 10.1016/j.vaccine.2012.05.091] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 04/10/2012] [Accepted: 05/03/2012] [Indexed: 12/18/2022]
Abstract
This chapter reviews the current treatment of chronic and neoplastic human papillomavirus (HPV)-associated conditions and the development of novel therapeutic approaches. Surgical excision of HPV-associated lower genital tract neoplasia is very successful but largely depends on secondary prevention programmes for identification of disease. Only high-risk HPV-driven chronic, pre-neoplastic lesions and some very early cancers cannot be successfully treated by surgical procedures alone. Chemoradiation therapy of cervical cancer contributes to the 66-79% cervical cancer survival at 5 years. Outlook for those patients with persistent or recurrent cervical cancer following treatment is very poor. Topical agents such as imiquimod (immune response modifier), cidofovir (inhibition of viral replication; induction apoptosis) or photodynamic therapy (direct damage of tumour and augmentation of anti-tumour immunity) have all shown some useful efficacy (~50-60%) in treatment of high grade vulvar intraepithelial neoplasia (VIN). Provider administered treatments of genital warts include cryotherapy, trichloracetic acid, or surgical removal which has the highest primary clearance rate. Patient applied therapies include podophyllotoxin and imiquimod. Recurrence after "successful" treatment is 30-40%. Further improvements could derive from a rational combination of current therapy with new drugs targeting molecular pathways mediated by HPV in cancer. Small molecule inhibitors targeting the DNA binding activities of HPV E1/E2 or the anti-apoptotic consequences of E6/E7 oncogenes are in preclinical development. Proteasome and histone deacetylase inhibitors, which can enhance apoptosis in HPV positive tumour cells, are being tested in early clinical trials. Chronic high-risk HPV infection/neoplasia is characterised by systemic and/or local immune suppressive regulatory or escape factors. Recently two E6/E7 vaccines have shown some clinical efficacy in high grade VIN patients and this correlated with strong and broad systemic HPV-specific T cell response and modulation of key local immune factors. Treatments that can shift the balance of immune effectors locally in combination with vaccination are now being tested. This article forms part of a special supplement entitled "Comprehensive Control of HPV Infections and Related Diseases" Vaccine Volume 30, Supplement 5, 2012.
Collapse
Affiliation(s)
- Peter L Stern
- Paterson Institute for Cancer Research, University of Manchester, Manchester M20 4BX, UK.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
van Luijn MM, van de Loosdrecht AA, Lampen MH, van Veelen PA, Zevenbergen A, Kester MGD, de Ru AH, Ossenkoppele GJ, van Hall T, van Ham SM. Promiscuous binding of invariant chain-derived CLIP peptide to distinct HLA-I molecules revealed in leukemic cells. PLoS One 2012; 7:e34649. [PMID: 22563374 PMCID: PMC3338516 DOI: 10.1371/journal.pone.0034649] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Accepted: 03/05/2012] [Indexed: 01/20/2023] Open
Abstract
Antigen presentation by HLA class I (HLA-I) and HLA class II (HLA-II) complexes is achieved by proteins that are specific for their respective processing pathway. The invariant chain (Ii)-derived peptide CLIP is required for HLA-II-mediated antigen presentation by stabilizing HLA-II molecules before antigen loading through transient and promiscuous binding to different HLA-II peptide grooves. Here, we demonstrate alternative binding of CLIP to surface HLA-I molecules on leukemic cells. In HLA-II-negative AML cells, we found plasma membrane display of the CLIP peptide. Silencing Ii in AML cells resulted in reduced HLA-I cell surface display, which indicated a direct role of CLIP in the HLA-I antigen presentation pathway. In HLA-I-specific peptide eluates from B-LCLs, five Ii-derived peptides were identified, of which two were from the CLIP region. In vitro peptide binding assays strikingly revealed that the eluted CLIP peptide RMATPLLMQALPM efficiently bound to four distinct HLA-I supertypes (-A2, -B7, -A3, -B40). Furthermore, shorter length variants of this CLIP peptide also bound to these four supertypes, although in silico algorithms only predicted binding to HLA-A2 or -B7. Immunization of HLA-A2 transgenic mice with these peptides did not induce CTL responses. Together these data show a remarkable promiscuity of CLIP for binding to a wide variety of HLA-I molecules. The found participation of CLIP in the HLA-I antigen presentation pathway could reflect an aberrant mechanism in leukemic cells, but might also lead to elucidation of novel processing pathways or immune escape mechanisms.
Collapse
Affiliation(s)
- Marvin M van Luijn
- Department of Hematology, VU Institute for Cancer and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Jotereau F, Gervois N, Labarrière N. Adoptive transfer with high-affinity TCR to treat human solid tumors: how to improve the feasibility? Target Oncol 2012; 7:3-14. [PMID: 22350487 DOI: 10.1007/s11523-012-0207-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 01/12/2012] [Indexed: 01/05/2023]
Abstract
The adoptive transfer of tumor antigen-specific T cells recently achieved clinical efficacy for a fraction of melanoma patients refractory to other therapies. Unfortunately, the application of this strategy to the remaining melanoma and most other cancer patients is hampered by the difficulty to generate high-affinity tumor-reactive T cells. Two strategies are currently developed to extend the feasibility of this therapeutic approach: clinical grade tool production for MHC-peptide multimer-driven sorting of antigen-specific T cells from the endogenous peripheral T cell repertoire and de novo engineering of the missing repertoire by genetic transfer of cloned specific T cell receptor (TCR) into T cells. The expected multiplication of adoptive transfer treatments, by these strategies, and their careful evaluation should enable the cure of a number of otherwise compromised cancer patients and to gain insight into the characteristics of transferred T cells best fitted to eradicate tumor cells, in terms of antigen specificities, phenotype, and functions. In particular, identification of tumor-rejection antigens by this approach would improve the design and efficacy of all immunotherapeutic approaches.
Collapse
|
36
|
van Hall T, van der Burg SH. Mechanisms of peptide vaccination in mouse models: tolerance, immunity, and hyperreactivity. Adv Immunol 2012; 114:51-76. [PMID: 22449778 DOI: 10.1016/b978-0-12-396548-6.00003-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The development of synthetic peptide vaccines capable of inducing strong and protective T-cell immunity has taken more than 20 years. Peptide vaccines come in many flavors and although their design is simple, their use is more complicated as the success of a particular peptide vaccine is influenced by many parameters. In fact, peptide vaccination may lead to tolerance, immunity or even hyper-reactivity causing death of the animals. Here we systematically dissect the parameters that influence the final outcome of peptide vaccines as examined in mouse models and this will guide the rational design of new vaccines in the future.
Collapse
Affiliation(s)
- Thorbald van Hall
- Department of Clinical Oncology, Experimental Cancer Immunology and Therapy, Leiden University Medical Center, Leiden, Netherlands
| | | |
Collapse
|
37
|
Aurora kinase A-specific T-cell receptor gene transfer redirects T lymphocytes to display effective antileukemia reactivity. Blood 2011; 119:368-76. [PMID: 22025529 DOI: 10.1182/blood-2011-06-360354] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Aurora kinase A (AURKA) is overexpressed in leukemias. Previously, we demonstrated that AURKA-specific CD8(+) T cells specifically and selectively lysed leukemia cells, indicating that AURKA is an excellent target for immunotherapy. In this study, we examined the feasibility of adoptive therapy using redirected T cells expressing an HLA-A*0201-restricted AURKA(207-215)-specific T-cell receptor (TCR). Retrovirally transduced T cells recognized relevant peptide-pulsed but not control target cells. Furthermore, TCR-redirected CD8(+) T cells lysed AURKA-overexpressing human leukemic cells in an HLA-A*0201-restricted manner, but did not kill HLA-A*0201(+) normal cells, including hematopoietic progenitors. In addition, AURKA(207-215)-specific TCR-transduced CD4(+) T cells displayed target-responsive Th1 cytokine production. Finally, AURKA(207-215)-specific TCR-transduced CD8(+) T cells displayed antileukemia efficacy in a xenograft mouse model. Collectively, these data demonstrate the feasibility of redirected T cell-based AURKA-specific immunotherapy for the treatment of human leukemia.
Collapse
|
38
|
Novel adoptive T-cell immunotherapy using a WT1-specific TCR vector encoding silencers for endogenous TCRs shows marked antileukemia reactivity and safety. Blood 2011; 118:1495-503. [DOI: 10.1182/blood-2011-02-337089] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Abstract
Adoptive T-cell therapy for malignancies using redirected T cells genetically engineered by tumor antigen-specific T-cell receptor (TCR) gene transfer is associated with mispairing between introduced and endogenous TCR chains with unknown specificity. Therefore, deterioration of antitumor reactivity and serious autoimmune reactivity are major concerns. To address this problem, we have recently established a novel retroviral vector system encoding siRNAs for endogenous TCR genes (siTCR vector). In this study, to test the clinical application of siTCR gene therapy for human leukemia, we examined in detail the efficacy and safety of WT1-siTCR–transduced T cells. Compared with conventional WT1-TCR (WT1-coTCR) gene-transduced T cells, these cells showed significant enhancement of antileukemia reactivity resulting from stronger expression of the introduced WT1-specific TCR with inhibition of endogenous TCRs. Notably, WT1-siTCR gene-transduced T cells were remarkably expandable after repetitive stimulation with WT1 peptide in vitro, without any deterioration of antigen specificity. WT1-siTCR gene–transduced T cells from leukemia patients successfully lysed autologous leukemia cells, but not normal hematopoietic progenitor cells. In a mouse xenograft model, adoptively transferred WT1-siTCR gene-transduced T cells exerted distinct antileukemia efficacy but did not inhibit human hematopoiesis. Our results suggest that gene-immunotherapy for leukemia using this WT1-siTCR system holds considerable promise.
Collapse
|
39
|
Melief CJM, O'Shea JJ, Stroncek DF. Summit on cell therapy for cancer: The importance of the interaction of multiple disciplines to advance clinical therapy. J Transl Med 2011; 9:107. [PMID: 21740581 PMCID: PMC3160382 DOI: 10.1186/1479-5876-9-107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 07/08/2011] [Indexed: 01/01/2023] Open
Abstract
The field of cellular therapy of cancer is moving quickly and the issues involved with its advancement are complex and wide ranging. The growing clinical applications and success of adoptive cellular therapy of cancer has been due to the rapid evolution of immunology, cancer biology, gene therapy and stem cell biology and the translation of advances in these fields from the research laboratory to the clinic. The continued development of this field is dependent on the exchange of ideas across these diverse disciplines, the testing of new ideas in the research laboratory and in animal models, the development of new cellular therapies and GMP methods to produce these therapies, and the testing of new adoptive cell therapies in clinical trials. The Summit on Cell Therapy for Cancer to held on November 1 and 2, 2011 at the National Institutes of Health (NIH) campus will include a mix of perspectives, concepts and ideas related to adoptive cellular therapy that are not normally presented together at any single meeting. This novel assembly will generate new ideas and new collaborations and possibly increase the rate of advancement of this field.
Collapse
Affiliation(s)
- Cornelis J M Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | | | | |
Collapse
|
40
|
Klebanoff CA, Gattinoni L, Palmer DC, Muranski P, Ji Y, Hinrichs CS, Borman ZA, Kerkar SP, Scott CD, Finkelstein SE, Rosenberg SA, Restifo NP. Determinants of successful CD8+ T-cell adoptive immunotherapy for large established tumors in mice. Clin Cancer Res 2011; 17:5343-52. [PMID: 21737507 DOI: 10.1158/1078-0432.ccr-11-0503] [Citation(s) in RCA: 231] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE Adoptive cell transfer (ACT) of tumor infiltrating or genetically engineered T cells can cause durable responses in patients with metastatic cancer. Multiple clinically modifiable parameters can comprise this therapy, including cell dose and phenotype, in vivo antigen restimulation, and common gamma-chain (γ(c)) cytokine support. However, the relative contributions of each these individual components to the magnitude of the antitumor response have yet to be quantified. EXPERIMENTAL DESIGN To systematically and quantitatively appraise each of these variables, we employed the Pmel-1 mouse model treating large, established B16 melanoma tumors. In addition to cell dose and magnitude of in vivo antigen restimulation, we also evaluated the relative efficacy of central memory (T(CM)), effector memory (T(EM)), and stem cell memory (T(SCM)) subsets on the strength of tumor regression as well as the dose and type of clinically available γ(c) cytokines, including IL-2, IL-7, IL-15, and IL-21. RESULTS We found that cell dose, T-cell differentiation status, and viral vaccine titer each were correlated strongly and significantly with the magnitude of tumor regression. Surprisingly, although the total number of IL-2 doses was correlated with tumor regression, no significant benefit to prolonged (≥6 doses) administration was observed. Moreover, the specific type and dose of γ(c) cytokine only moderately correlated with response. CONCLUSION Collectively, these findings elucidate some of the key determinants of successful ACT immunotherapy for the treatment of cancer in mice and further show that γ(c) cytokines offer a similar ability to effectively drive antitumor T-cell function in vivo.
Collapse
Affiliation(s)
- Christopher A Klebanoff
- Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Correction: Peptide Vaccination after T-Cell Transfer Causes Massive Clonal Expansion, Tumor Eradication, and Manageable Cytokine Storm. Cancer Res 2011. [DOI: 10.1158/0008-5472.can-11-1279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
42
|
Li A, Xiong S, Lin Y, Liu R, Chu Y. A high-affinity T-helper epitope enhances peptide-pulsed dendritic cell-based vaccine. DNA Cell Biol 2011; 30:883-92. [PMID: 21612399 DOI: 10.1089/dna.2011.1222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The NV epitope, a dominant helper determinant from the circumsporozoite antigen of Plasmodium falciparum, is strongly immunogenic and can provide help for cytotoxic T-lymphocyte (CTL) activation. In this study, we evaluated whether the addition of NV peptide can augment the efficacy of peptide-pulsed dendritic cell (DC) immunization in vivo. Using B16 melanoma as tumor model, we demonstrated that DCs pulsed with both NV and gp100 (a melanoma-specific antigen) peptide enhanced immune priming and protection from tumor challenge in vivo. Further, we showed the mechanisms of the NV epitope that help CTL activation; MHC-II-restricted NV peptide induced dramatically more effective helper cells, with a higher level of CD40L expression and IFN-γ production, which, in turn, more effectively conditioned DCs for CTL activation. The improved helper cells also induced greater IL-12 production by DCs, accounting for the reciprocal T-helper polarization to Th1, and increased the expression of costimulatory molecules. Collectively, these findings demonstrate that NV peptide in addition to tumor antigen-pulsed DC immunizations augment helper cell activation, which in turn promotes maturation of DC, and enhance in vivo antitumor activity.
Collapse
Affiliation(s)
- Ang Li
- Key Laboratory of Molecular Medicine of Ministry of Education, Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, Shanghai, P.R. China
| | | | | | | | | |
Collapse
|
43
|
Jager MJ, Ly LV, El Filali M, Madigan MC. Macrophages in uveal melanoma and in experimental ocular tumor models: Friends or foes? Prog Retin Eye Res 2011; 30:129-46. [DOI: 10.1016/j.preteyeres.2010.11.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 11/16/2010] [Accepted: 11/22/2010] [Indexed: 01/05/2023]
|
44
|
Gilham DE. Effective adoptive T-cell therapy for cancer in the absence of host lymphodepletion. Immunotherapy 2011; 3:177-9. [DOI: 10.2217/imt.10.115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Evaluation of: Ly LV, Sluijter M, Versluis M et al.: Peptide vaccination after T-cell transfer causes massive clonal expansion, tumor eradication and manageable cytokine storm. Cancer Res. 70(21), 8339–8346 (2010). Adoptive T-cell transfer (ACT) to treat cancer, autoimmunity and infectious disease holds great promise. In the cancer field, current dogma suggests that achieving a high frequency of circulating, transferred T cells is critical for therapeutic success. Achieving this high level of T-cell engraftment currently requires preconditioning of the patient. In effect, this means the eradication of the patient’s own immune system, thereby creating ‘space’ for the adoptively transferred T cells to populate in the absence of host-cell competition. While different forms of preconditioning are employed, each carries a significant level of toxicity itself. In the paper being evaluated, Ly et al. demonstrate that the combination of ACT with vaccination using long peptides, a Toll-like receptor-7 ligand and cytokine support in the form of IL-2 can drive the expansion of adoptively transferred antigen-specific T cells in the absence of preconditioning regimens. This paper infers that reduced intensity regimens may be suitable for ACT clinical protocols.
Collapse
Affiliation(s)
- David E Gilham
- Clinical & Experimental Immunotherapy, Department of Medical Oncology, Paterson Institute for Cancer Research, The University of Machester, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| |
Collapse
|