251
|
Cannon MJ, Block MS, Morehead LC, Knutson KL. The evolving clinical landscape for dendritic cell vaccines and cancer immunotherapy. Immunotherapy 2019; 11:75-79. [PMID: 30730268 DOI: 10.2217/imt-2018-0129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Martin J Cannon
- Department of Microbiology & Immunology, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205, USA
| | - Matthew S Block
- Department of Oncology, & Department of Immunology, Mayo Clinic, 200 First St, SW Rochester, MN 55905, USA
| | - Lauren C Morehead
- Department of Microbiology & Immunology, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205, USA
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| |
Collapse
|
252
|
Wickström SL, Lövgren T, Volkmar M, Reinhold B, Duke-Cohan JS, Hartmann L, Rebmann J, Mueller A, Melief J, Maas R, Ligtenberg M, Hansson J, Offringa R, Seliger B, Poschke I, Reinherz EL, Kiessling R. Cancer Neoepitopes for Immunotherapy: Discordance Between Tumor-Infiltrating T Cell Reactivity and Tumor MHC Peptidome Display. Front Immunol 2019; 10:2766. [PMID: 31921104 PMCID: PMC6918724 DOI: 10.3389/fimmu.2019.02766] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/12/2019] [Indexed: 12/22/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TIL) are considered enriched for T cells recognizing shared tumor antigens or mutation-derived neoepitopes. We performed exome sequencing and HLA-A*02:01 epitope prediction from tumor cell lines from two HLA-A2-positive melanoma patients whose TIL displayed strong tumor reactivity. The potential neoepitopes were screened for recognition using autologous TIL by immunological assays and presentation on tumor major histocompatibility complex class I (MHC-I) molecules by Poisson detection mass spectrometry (MS). TIL from the patients recognized 5/181 and 3/49 of the predicted neoepitopes, respectively. MS screening detected 3/181 neoepitopes on tumor MHC-I from the first patient but only one was also among those recognized by TIL. Consequently, TIL enriched for neoepitope specificity failed to recognize tumor cells, despite being activated by peptides. For the second patient, only after IFN-γ treatment of the tumor cells was one of 49 predicted neoepitopes detected by MS, and this coincided with recognition by TIL sorted for the same specificity. Importantly, specific T cells could be expanded from patient and donor peripheral blood mononuclear cells (PBMC) for all neoepitopes recognized by TIL and/or detected on tumor MHC-I. In summary, stimulating the appropriate inflammatory environment within tumors may promote neoepitope MHC presentation while expanding T cells in blood may circumvent lack of specific TIL. The discordance in detection between physical and functional methods revealed here can be rationalized and used to improve neoantigen-targeted T cell immunotherapy.
Collapse
Affiliation(s)
- Stina L Wickström
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Tanja Lövgren
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Michael Volkmar
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bruce Reinhold
- Laboratory of Immunobiology, Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Jonathan S Duke-Cohan
- Laboratory of Immunobiology, Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Laura Hartmann
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Janina Rebmann
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Mueller
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Jeroen Melief
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Roeltje Maas
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Maarten Ligtenberg
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Johan Hansson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Rienk Offringa
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Isabel Poschke
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany.,DKTK Immune Monitoring Unit, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Ellis L Reinherz
- Laboratory of Immunobiology, Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Rolf Kiessling
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
253
|
Ubachs J, Ziemons J, Minis‐Rutten IJ, Kruitwagen RF, Kleijnen J, Lambrechts S, Olde Damink SW, Rensen SS, Van Gorp T. Sarcopenia and ovarian cancer survival: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle 2019; 10:1165-1174. [PMID: 31389674 PMCID: PMC6903439 DOI: 10.1002/jcsm.12468] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/24/2019] [Accepted: 06/12/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Sarcopenia is the loss of skeletal muscle mass and function that occurs with advancing age and certain diseases. It is thought to have a negative impact on survival in cancer patients. Routine computed tomography imaging is often used to quantify skeletal muscle in cancer patients. Sarcopenia is defined by a low skeletal muscle index (SMI). Skeletal muscle radiation attenuation (SMRA) is used to define muscle quality. The primary aim of this meta-analysis was to study the association between sarcopenia or SMRA and overall survival (OS) or complications in patients with ovarian cancer. METHODS Medline, Embase, CINAHL, and PEDro databases were searched from inception to 15 February 2019. Studies evaluating the prognostic effect of SMI and SMRA on ovarian cancer survival or surgical complications were included. Risk of bias and study quality were evaluated with the Quality in Prognosis Studies Instrument (QUIPS) according to the modified Grading of Recommendations Assessment, Development, and Evaluation (GRADE) framework. RESULTS The search strategy yielded 4262 hits in all four databases combined. Ten and eight studies were included for qualitative and quantitative analysis, respectively. Meta-analysis revealed a significant association between the SMI and OS [0.007; hazard ratio (HR): 1.11, 95% confidence interval (CI): 1.03-1.20]. SMRA was also significantly associated with OS (P < 0.001; HR: 1.14, 95% CI: 1.08-1.20). Association between the SMI and surgical complications had borderline statistical significance (0.05; HR: 1.23, 95% CI: 1.00-1.52). The risk of bias assessed with QUIPS was high in all studies. The quality of the evidence was very low. CONCLUSIONS Whereas our meta-analysis indicated that a low SMI and low SMRA are associated with survival in ovarian cancer patients, the low quality of the source data precludes drawing definitive conclusions.
Collapse
Affiliation(s)
- Jorne Ubachs
- Department of Obstetrics and GynecologyMaastricht University Medical CentreMaastrichtThe Netherlands
- GROW—School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
- Department of SurgeryMaastricht University Medical CentreMaastrichtThe Netherlands
- NUTRIM, School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtThe Netherlands
| | - Janine Ziemons
- Department of SurgeryMaastricht University Medical CentreMaastrichtThe Netherlands
| | | | - Roy F.P.M. Kruitwagen
- Department of Obstetrics and GynecologyMaastricht University Medical CentreMaastrichtThe Netherlands
- GROW—School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Jos Kleijnen
- CAPHRI, School for Public Health and Primary CareMaastricht UniversityMaastrichtThe Netherlands
| | - Sandrina Lambrechts
- Department of Obstetrics and GynecologyMaastricht University Medical CentreMaastrichtThe Netherlands
- GROW—School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Steven W.M. Olde Damink
- Department of SurgeryMaastricht University Medical CentreMaastrichtThe Netherlands
- NUTRIM, School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtThe Netherlands
- Department of Visceral and Transplantation SurgeryRWTH Aachen UniversityAachenGermany
| | - Sander S. Rensen
- Department of SurgeryMaastricht University Medical CentreMaastrichtThe Netherlands
- NUTRIM, School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtThe Netherlands
| | - Toon Van Gorp
- Department of Obstetrics and Gynecology, Division of Gynaecological Oncology, Leuven Cancer InstituteUniversity Hospitals LeuvenLeuvenBelgium
| |
Collapse
|
254
|
Perez CR, De Palma M. Engineering dendritic cell vaccines to improve cancer immunotherapy. Nat Commun 2019; 10:5408. [PMID: 31776331 PMCID: PMC6881351 DOI: 10.1038/s41467-019-13368-y] [Citation(s) in RCA: 289] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/06/2019] [Indexed: 12/19/2022] Open
Abstract
At the interface between the innate and adaptive immune system, dendritic cells (DCs) play key roles in tumour immunity and hold a hitherto unrealized potential for cancer immunotherapy. Here we review the role of distinct DC subsets in the tumour microenvironment, with special emphasis on conventional type 1 DCs. Integrating new knowledge of DC biology and advancements in cell engineering, we provide a blueprint for the rational design of optimized DC vaccines for personalized cancer medicine. Dendritic cells (DCs) have been explored as a promising strategy for cancer immunotherapy. In this Perspective, the authors discuss the different types of DCs and their therapeutic potential in the context of vaccines for personalized cancer therapy.
Collapse
Affiliation(s)
- Caleb R Perez
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland.
| |
Collapse
|
255
|
Sarivalasis A, Boudousquié C, Balint K, Stevenson BJ, Gannon PO, Iancu EM, Rossier L, Martin Lluesma S, Mathevet P, Sempoux C, Coukos G, Dafni U, Harari A, Bassani-Sternberg M, Kandalaft LE. A Phase I/II trial comparing autologous dendritic cell vaccine pulsed either with personalized peptides (PEP-DC) or with tumor lysate (OC-DC) in patients with advanced high-grade ovarian serous carcinoma. J Transl Med 2019; 17:391. [PMID: 31771601 PMCID: PMC6880492 DOI: 10.1186/s12967-019-02133-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/09/2019] [Indexed: 02/07/2023] Open
Abstract
Background Most ovarian cancer patients are diagnosed at a late stage with 85% of them relapsing after surgery and standard chemotherapy; for this reason, new treatments are urgently needed. Ovarian cancer has become a candidate for immunotherapy by reason of their expression of shared tumor-associated antigens (TAAs) and private mutated neoantigens (NeoAgs) and the recognition of the tumor by the immune system. Additionally, the presence of intraepithelial tumor infiltrating lymphocytes (TILs) is associated with improved progression-free and overall survival of patients with ovarian cancer. The aim of active immunotherapy, including vaccination, is to generate a new anti-tumor response and amplify an existing immune response. Recently developed NeoAgs-based cancer vaccines have the advantage of being more tumor specific, reducing the potential for immunological tolerance, and inducing robust immunogenicity. Methods We propose a randomized phase I/II study in patients with advanced ovarian cancer to compare the immunogenicity and to assess safety and feasibility of two personalized DC vaccines. After standard of care surgery and chemotherapy, patients will receive either a novel vaccine consisting of autologous DCs pulsed with up to ten peptides (PEP-DC), selected using an agnostic, yet personalized, epitope discovery algorithm, or a sequential combination of a DC vaccine loaded with autologous oxidized tumor lysate (OC-DC) prior to an equivalent PEP-DC vaccine. All vaccines will be administered in combination with low-dose cyclophosphamide. This study is the first attempt to compare the two approaches and to use NeoAgs-based vaccines in ovarian cancer in the adjuvant setting. Discussion The proposed treatment takes advantage of the beneficial effects of pre-treatment with OC-DC prior to PEP-DC vaccination, prompting immune response induction against a wide range of patient-specific antigens, and amplification of pre-existing NeoAgs-specific T cell clones. Trial registration This trial is already approved by Swissmedic (Ref.: 2019TpP1004) and will be registered at http://www.clinicaltrials.gov before enrollment opens.
Collapse
Affiliation(s)
- Apostolos Sarivalasis
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Caroline Boudousquié
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Klara Balint
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Philippe O Gannon
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Emanuela Marina Iancu
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Laetitia Rossier
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Silvia Martin Lluesma
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Patrice Mathevet
- Women-Mother-Child Department, Service of Gynecology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Christine Sempoux
- Department of Pathology, University Hospital of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Urania Dafni
- Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian, University of Athens, Athens, Greece
| | - Alexandre Harari
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lana E Kandalaft
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
256
|
Dendritic Cells Loaded with Heat Shock-Conditioned Ovarian Epithelial Carcinoma Cell Lysates Elicit T Cell-Dependent Antitumor Immune Responses In Vitro. J Immunol Res 2019; 2019:9631515. [PMID: 31886313 PMCID: PMC6899292 DOI: 10.1155/2019/9631515] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 09/08/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
Ovarian epithelial carcinoma (OEC) is the most frequent ovarian tumor, characterized by a high mortality in advanced stages where conventional therapies are not effective. Based on the role of the immune system in the progression of this disease, immunotherapy using checkpoint blockade has been considered as a therapeutic alternative. Nevertheless, its results do not match up to the positive results in entities like melanoma and other malignancies, suggesting the need to find other therapies to be used alone or in combination. Dendritic cell- (DC-) based vaccines have shown promising results in several types of cancer, such as melanoma, prostate, and lung cancers, due to the essential role played by DCs in the activation of specific T cells, thus using other ways of activating the immune response than immune checkpoint blockade. During the last decade, we have used DC-based vaccines loaded with an allogeneic heat shock-conditioned melanoma cell lysate in the treatment of advanced stage patients in a series of clinical trials. In these studies, 60% of treated patients showed immunological responses which correlated positively with improved survival. Considering the relevance of ovarian cancer and the promising results of our DC-based vaccine, we show here that heat shock-conditioned cell lysates derived from ovarian epithelial carcinoma cell lines have the potential to induce the phenotypic and functional maturation of human DC, which in turn, is able to induce an efficient CD4+ and CD8+ T cell-mediated immune responses against ovarian cancer cell lines in vitro. In summary, OEC heat shock-conditioned cell lysate-loaded DCs may be considered for future combined immunotherapy approaches against ovarian tumors.
Collapse
|
257
|
Immunological ignorance is an enabling feature of the oligo-clonal T cell response to melanoma neoantigens. Proc Natl Acad Sci U S A 2019; 116:23662-23670. [PMID: 31685621 DOI: 10.1073/pnas.1906026116] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The impact of intratumoral heterogeneity (ITH) and the resultant neoantigen landscape on T cell immunity are poorly understood. ITH is a widely recognized feature of solid tumors and poses distinct challenges related to the development of effective therapeutic strategies, including cancer neoantigen vaccines. Here, we performed deep targeted DNA sequencing of multiple metastases from melanoma patients and observed ubiquitous sharing of clonal and subclonal single nucleotide variants (SNVs) encoding putative HLA class I-restricted neoantigen epitopes. However, spontaneous antitumor CD8+ T cell immunity in peripheral blood and tumors was restricted to a few clonal neoantigens featuring an oligo-/monoclonal T cell-receptor (TCR) repertoire. Moreover, in various tumors of the 4 patients examined, no neoantigen-specific TCR clonotypes were identified despite clonal neoantigen expression. Mature dendritic cell (mDC) vaccination with tumor-encoded amino acid-substituted (AAS) peptides revealed diverse neoantigen-specific CD8+ T responses, each composed of multiple TCR clonotypes. Isolation of T cell clones by limiting dilution from tumor-infiltrating lymphocytes (TILs) permitted functional validation regarding neoantigen specificity. Gene transfer of TCRαβ heterodimers specific for clonal neoantigens confirmed correct TCR clonotype assignments based on high-throughput TCRBV CDR3 sequencing. Our findings implicate immunological ignorance of clonal neoantigens as the basis for ineffective T cell immunity to melanoma and support the concept that therapeutic vaccination, as an adjunct to checkpoint inhibitor treatment, is required to increase the breadth and diversity of neoantigen-specific CD8+ T cells.
Collapse
|
258
|
Benvenuto M, Focaccetti C, Izzi V, Masuelli L, Modesti A, Bei R. Tumor antigens heterogeneity and immune response-targeting neoantigens in breast cancer. Semin Cancer Biol 2019; 72:65-75. [PMID: 31698088 DOI: 10.1016/j.semcancer.2019.10.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/30/2019] [Indexed: 12/25/2022]
Abstract
Breast cancer is both the most common type of cancer and the most frequent cause of cancer mortality in women, mainly because of its heterogeneity and limited immunogenicity. The aim of specific active cancer immunotherapy is to stimulate the host's immune response against cancer cells directly using a vaccine platform carrying one or more tumor antigens. In particular, the ideal tumor antigen should be able to elicit T cell and B cell responses, be specific for the tumor and be expressed at high levels on cancer cells. Neoantigens are ideal targets for immunotherapy because they are exclusive to individual patient's tumors, are absent in healthy tissues and are not subject to immune tolerance mechanisms. Thus, neoantigens should generate a specific reaction towards tumors since they constitute the largest fraction of targets of tumor-infiltrating T cells. In this review, we describe the technologies used for neoantigen discovery, the heterogeneity of neoantigens in breast cancer and recent studies of breast cancer immunotherapy targeting neoantigens.
Collapse
Affiliation(s)
- Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy; Saint Camillus International University of Health and Medical Sciences, via di Sant'Alessandro 8, 00131, Rome, Italy.
| | - Chiara Focaccetti
- Department of Human Science and Promotion of the Quality of Life, University San Raffaele Rome, Via di Val Cannuta 247, 00166, Rome, Italy.
| | - Valerio Izzi
- Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7C, FI-90230, Oulu, Finland.
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy.
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
259
|
A systems approach to clinical oncology uses deep phenotyping to deliver personalized care. Nat Rev Clin Oncol 2019; 17:183-194. [DOI: 10.1038/s41571-019-0273-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2019] [Indexed: 02/06/2023]
|
260
|
Scheetz L, Park KS, Li Q, Lowenstein PR, Castro MG, Schwendeman A, Moon JJ. Engineering patient-specific cancer immunotherapies. Nat Biomed Eng 2019; 3:768-782. [PMID: 31406259 PMCID: PMC6783331 DOI: 10.1038/s41551-019-0436-x] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
Research into the immunological processes implicated in cancer has yielded a basis for the range of immunotherapies that are now considered the fourth pillar of cancer treatment (alongside surgery, radiotherapy and chemotherapy). For some aggressive cancers, such as advanced non-small-cell lung carcinoma, combination immunotherapies have resulted in unprecedented treatment efficacy for responding patients, and have become frontline therapies. Individualized immunotherapy, enabled by the identification of patient-specific mutations, neoantigens and biomarkers, and facilitated by advances in genomics and proteomics, promises to broaden the responder patient population. In this Perspective, we give an overview of immunotherapies leveraging engineering approaches, including the design of biomaterials, delivery strategies and nanotechnology solutions, for the realization of individualized cancer treatments such as nanoparticle vaccines customized with neoantigens, cell therapies based on patient-derived dendritic cells and T cells, and combinations of theranostic strategies. Developments in precision cancer immunotherapy will increasingly rely on the adoption of engineering principles.
Collapse
Affiliation(s)
- Lindsay Scheetz
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Kyung Soo Park
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Qiao Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
261
|
Castiello L, Aricò E, D'Agostino G, Santodonato L, Belardelli F. In situ Vaccination by Direct Dendritic Cell Inoculation: The Coming of Age of an Old Idea? Front Immunol 2019; 10:2303. [PMID: 31611878 PMCID: PMC6773832 DOI: 10.3389/fimmu.2019.02303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
For more than 25 years, dendritic cell (DC) based vaccination has flashily held promises to represent a therapeutic approach for cancer treatment. While the vast majority of studies has focused on the use of antigen loaded DC, the intratumoral delivery of unloaded DC aiming at in situ vaccination has gained much less attention. Such approach grounds on the ability of inoculated DC to internalize and process antigens directly released by tumor (usually in combination with cell-death-inducing agents) to activate broad patient-specific antitumor T cell response. In this review, we highlight the recent studies in both solid and hematological tumors showing promising clinical results and discuss the main pitfalls and advantages of this approach for endogenous cancer vaccination. Lastly, we discuss how in situ vaccination by DC inoculation may fit with current immunotherapy approaches to expand and prolong patient response.
Collapse
Affiliation(s)
- Luciano Castiello
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Eleonora Aricò
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Laura Santodonato
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Filippo Belardelli
- Consiglio Nazionale Delle Ricerche, Institute of Translational Pharmacology, Rome, Italy
| |
Collapse
|
262
|
Current Possibilities of Gynecologic Cancer Treatment with the Use of Immune Checkpoint Inhibitors. Int J Mol Sci 2019; 20:ijms20194705. [PMID: 31547532 PMCID: PMC6801535 DOI: 10.3390/ijms20194705] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 12/17/2022] Open
Abstract
Despite the ongoing progress in cancer research, the global cancer burden has increased to 18.1 million new cases and 9.6 million deaths in 2018. Gynecological cancers, such as ovarian, endometrial, and cervical cancers, considerably contribute to global cancer burden, leading to $5,862.6, $2,945.7, and $1,543.9 million of annual costs of cancer care, respectively. Thus, the development of effective therapies against gynecological cancers is still a largely unmet medical need. One of the novel therapeutic approaches is to induce anti-cancer immunity by the inhibition of the immune checkpoint pathways using monoclonal antibodies. The molecular targets for monoclonal antibodies are cytotoxic T lymphocyte-associated protein-4 (CTLA-4), programmed cell death protein-1 (PD-1), and programmed death-ligand 1 (PD-L1). The rationale for the use of immune checkpoint inhibitors in patients with gynecological cancers was based on the immunohistological studies showing high expression levels of PD-1 and PD-L1 in those cancers. Currently available immune checkpoint inhibitors include nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, and ipilimumab. The efficacy and safety of these inhibitors, used as monotherapy and with combination with chemotherapy, is being currently evaluated in several clinical studies. As the results are promising, more clinical trials are being planned, which may lead to the development of efficient therapies for gynecological cancer patients.
Collapse
|
263
|
Corradetti B, Pisano S, Conlan RS, Ferrari M. Nanotechnology and Immunotherapy in Ovarian Cancer: Tracing New Landscapes. J Pharmacol Exp Ther 2019; 370:636-646. [PMID: 30737357 PMCID: PMC6806629 DOI: 10.1124/jpet.118.254979] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/28/2019] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer (OC) is the seventh most common cancer in women worldwide. Standard therapeutic treatments involve debulking surgery combined with platinum-based chemotherapies. Of the patients with advanced-stage cancer who initially respond to current treatments, 50%-75% relapse. Immunotherapy-based approaches aimed at boosting antitumor immunity have recently emerged as promising tools to challenge tumor progression. Treatments with inhibitors of immune checkpoint molecules have shown impressive results in other types of tumors. However, only 15% of checkpoint inhibitors evaluated have proven successful in OC due to the immunosuppressive environment of the tumor and the transport barriers. This limits the efficacy of the existing immunotherapies. Nanotechnology-based delivery systems hold the potential to overcome such limitations. Various nanoformulations including polymeric, liposomes, and lipid-polymer hybrid nanoparticles have already been proposed to improve the biodistribution and targeting capabilities of drugs against tumor-associated immune cells, including dendritic cells and macrophages. In this review, we examine the impact of immunotherapeutic approaches that are currently under consideration for the treatment of OC. In this review, we also provide a comprehensive analysis of the existing nanoparticle-based synthetic strategies and their limitations and advantages over standard treatments. Furthermore, we discuss how the strength of the combination of nanotechnology with immunotherapy may help to overcome the current therapeutic limitations associated with their individual application and unravel a new paradigm in the treatment of this malignancy.
Collapse
Affiliation(s)
- Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas (B.C., S.P., R.S.C., M.F.); Swansea University Medical School, Singleton Park, Swansea, United Kingdom (B.C., S.P., R.S.C.); and Department of Medicine, Weill Cornell Medical College, New York, New York (M.F.)
| | - Simone Pisano
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas (B.C., S.P., R.S.C., M.F.); Swansea University Medical School, Singleton Park, Swansea, United Kingdom (B.C., S.P., R.S.C.); and Department of Medicine, Weill Cornell Medical College, New York, New York (M.F.)
| | - Robert Steven Conlan
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas (B.C., S.P., R.S.C., M.F.); Swansea University Medical School, Singleton Park, Swansea, United Kingdom (B.C., S.P., R.S.C.); and Department of Medicine, Weill Cornell Medical College, New York, New York (M.F.)
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas (B.C., S.P., R.S.C., M.F.); Swansea University Medical School, Singleton Park, Swansea, United Kingdom (B.C., S.P., R.S.C.); and Department of Medicine, Weill Cornell Medical College, New York, New York (M.F.)
| |
Collapse
|
264
|
Shae D, Baljon JJ, Wehbe M, Becker KW, Sheehy TL, Wilson JT. At the bench: Engineering the next generation of cancer vaccines. J Leukoc Biol 2019; 108:1435-1453. [PMID: 31430398 DOI: 10.1002/jlb.5bt0119-016r] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 06/29/2019] [Accepted: 07/25/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer vaccines hold promise as an immunotherapeutic modality based on their potential to generate tumor antigen-specific T cell responses and long-lived antitumor responses capable of combating metastatic disease and recurrence. However, cancer vaccines have historically failed to deliver significant therapeutic benefit in the clinic, which we maintain is due in part to drug delivery challenges that have limited vaccine immunogenicity and efficacy. In this review, we examine some of the known and putative failure mechanisms of common first-generation clinical cancer vaccines, and describe how the rational design of materials engineered for vaccine delivery and immunomodulation can address these shortcomings. First, we outline vaccine design principles for augmenting cellular immunity to tumor antigens and describe how well-engineered materials can improve vaccine efficacy, highlighting recent innovations in vaccine delivery technology that are primed for integration into neoantigen vaccine development pipelines. We also discuss the importance of sequencing, timing, and kinetics in mounting effective immune responses to cancer vaccines, and highlight examples of materials that potentiate antitumor immunity through spatiotemporal control of immunomodulation. Furthermore, we describe several engineering strategies for improving outcomes of in situ cancer vaccines, which leverage local, intratumoral delivery to stimulate systemic immunity. Finally, we highlight recent innovations leveraging nanotechnology for increasing the immunogenicity of the tumor microenvironment (TME), which is critical to enhancing tumor infiltration and function of T cells elicited in response to cancer vaccines. These immunoengineering strategies and tools complement ongoing advances in cancer vaccines as they reemerge as an important component of the immunotherapeutic armamentarium.
Collapse
Affiliation(s)
- Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jessalyn J Baljon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Mohamed Wehbe
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Kyle W Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Taylor L Sheehy
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - John Tanner Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
265
|
Kandalaft LE, Odunsi K, Coukos G. Immunotherapy in Ovarian Cancer: Are We There Yet? J Clin Oncol 2019; 37:2460-2471. [PMID: 31403857 DOI: 10.1200/jco.19.00508] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Lana E Kandalaft
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| | - Kunle Odunsi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - George Coukos
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
266
|
Matulonis UA, Shapira-Frommer R, Santin AD, Lisyanskaya AS, Pignata S, Vergote I, Raspagliesi F, Sonke GS, Birrer M, Provencher DM, Sehouli J, Colombo N, González-Martín A, Oaknin A, Ottevanger PB, Rudaitis V, Katchar K, Wu H, Keefe S, Ruman J, Ledermann JA. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE-100 study. Ann Oncol 2019; 30:1080-1087. [PMID: 31046082 DOI: 10.1093/annonc/mdz135] [Citation(s) in RCA: 460] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Advanced recurrent ovarian cancer (ROC) is the leading cause of gynecologic cancer-related death in developed countries and new treatments are needed. Previous studies of immune checkpoint blockade showed low objective response rates (ORR) in ROC with no identified predictive biomarker. PATIENTS AND METHODS This phase II study of pembrolizumab (NCT02674061) examined two patient cohorts with ROC: cohort A received one to three prior lines of treatment with a platinum-free interval (PFI) or treatment-free interval (TFI) between 3 and 12 months and cohort B received four to six prior lines with a PFI/TFI of ≥3 months. Pembrolizumab 200 mg was administered intravenously every 3 weeks until cancer progression, toxicity, or completion of 2 years. Primary end points were ORR by Response Evaluation Criteria in Solid Tumors version 1.1 per blinded independent central review by cohort and by PD-L1 expression measured as combined positive score (CPS). Secondary end points included duration of response (DOR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and safety. RESULTS Cohort A enrolled 285 patients; the first 100 served as the training set for PD-L1 biomarker analysis. Cohort B enrolled 91 patients. ORR was 7.4% for cohort A and 9.9% for cohort B. Median DOR was 8.2 months for cohort A and not reached for cohort B. DCR was 37.2% and 37.4%, respectively, in cohorts A and B. Based on the training set analysis, CPS 1 and 10 were selected for evaluation in the confirmation set. In the confirmation set, ORR was 4.1% for CPS <1, 5.7% CPS ≥1, and 10.0% for CPS ≥10. PFS was 2.1 months for both cohorts. Median OS was not reached for cohort A and was 17.6 months for cohort B. Toxicities were consistent with other single-agent pembrolizumab trials. CONCLUSIONS Single-agent pembrolizumab showed modest activity in patients with ROC. Higher PD-L1 expression was correlated with higher response. CLINICAL TRIAL NUMBER Clinicaltrials.gov, NCT02674061.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/drug therapy
- Adenocarcinoma, Clear Cell/pathology
- Aged
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Cohort Studies
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/pathology
- Female
- Follow-Up Studies
- Humans
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/pathology
- Prognosis
- Survival Rate
Collapse
Affiliation(s)
- U A Matulonis
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, Boston, USA.
| | - R Shapira-Frommer
- Oncology Institute and Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - A D Santin
- Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, USA
| | - A S Lisyanskaya
- Department of Gynaecological Oncology, City Clinical Oncology Dispensary, Saint Petersburg, Russia
| | - S Pignata
- Department of Urogynaecological Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G Pascale", IRCCS, Naples, Italy
| | - I Vergote
- Department of Obstetrics and Gynaecology and Gynaecologic Oncology, University Hospital Leuven, Leuven, Belgium
| | - F Raspagliesi
- Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - G S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Birrer
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, USA
| | - D M Provencher
- Hôpital Notre-Dame - Pavillon L-C Simard, Centre Hospitalier de L'Université de Montréal (CHUM), Montreal, Canada
| | - J Sehouli
- Gynecology and Obstetrics, Charité-Medical University of Berlin, Berlin, Germany
| | - N Colombo
- Department of Surgical Sciences, University of Milano-Bicocca and European Institute of Oncology, Milano, Italy
| | - A González-Martín
- Medical Oncology, Clinica Universidad de Navarra; formerly of MD Anderson International España, Madrid
| | - A Oaknin
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - P B Ottevanger
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - V Rudaitis
- Clinic of Obstetrics and Gynecology, Vilnius University Institute of Clinical Medicine, Vilnius, Lithuania
| | - K Katchar
- Companion Diagnostics, Merck & Co., Inc, Kenilworth, USA
| | - H Wu
- BARDS, MSD China, Beijing, China
| | - S Keefe
- Clinical Development, Merck & Co., Inc., Kenilworth, USA
| | - J Ruman
- Clinical Development, Merck & Co., Inc., Kenilworth, USA
| | - J A Ledermann
- UCL Cancer Institute and UCL Hospitals, Department of Oncology, University College London, London, UK
| |
Collapse
|
267
|
Liu S, Matsuzaki J, Wei L, Tsuji T, Battaglia S, Hu Q, Cortes E, Wong L, Yan L, Long M, Miliotto A, Bateman NW, Lele SB, Chodon T, Koya RC, Yao S, Zhu Q, Conrads TP, Wang J, Maxwell GL, Lugade AA, Odunsi K. Efficient identification of neoantigen-specific T-cell responses in advanced human ovarian cancer. J Immunother Cancer 2019; 7:156. [PMID: 31221207 PMCID: PMC6587259 DOI: 10.1186/s40425-019-0629-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/30/2019] [Indexed: 12/20/2022] Open
Abstract
Background Efficient identification of neoantigen-specific T-cell responses in epithelial ovarian cancer (EOC) remains a challenge. Existing investigations of spontaneous T-cell response to tumor neoepitope in EOC have taken the approach of comprehensive screening all neoantigen candidates, with a validation rate of 0.5–2%. Methods Whole-exome and transcriptome sequencing analysis of treatment-naive EOC patients were performed to identify neoantigen candidates, and the immunogenicity of prioritized neoantigens was evaluated by analyzing spontaneous neoantigen-specfic CD4+ and CD8+ T-cell responses in the tumor and/or peripheral blood. The biological relevance of neoantigen-specific T-cell lines and clones were analyzed by evaluating the capacity of autologous ovarian tumor recognition. Genetic transfer of T-cell receptor (TCR) from these neoantigen-specific T-cell clones into peripheral blood T-cells was conducted to generate neoepitope-specific T-cells. The molecular signature associated with positive neoantigen T-cell responses was investigated, and the impacts of expression level and lymphocyte source on neoantigen identification were explored. Results Using a small subset of prioritized neoantigen candidates, we were able to detect spontaneous CD4+ and/or CD8+ T-cell responses against neoepitopes from autologous lymphocytes in half of treatment-naïve EOC patients, with a significantly improved validation rate of 19%. Tumors from patients exhibiting neoantigen-specific T-cell responses exhibited a signature of upregulated antigen processing and presentation machinery, which was also associated with favorable patient survival in the TCGA ovarian cohort. T-cells specific against two mutated cancer-associated genes, NUP214 and JAK1, recognized autologous tumors. Gene-engineering with TCR from these neoantigen-specific T-cell clones conferred neoantigen-reactivity to peripheral T-cells. Conclusions Our study demonstrated the feasibility of efficiently identifying both CD4+ and CD8+ neoantigen-specific T-cells in EOC. Autologous lymphocytes genetically engineered with tumor antigen-specific TCR can be used to generate cells for use in the personalized adoptive T-cell transfer immunotherapy. Electronic supplementary material The online version of this article (10.1186/s40425-019-0629-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| | - Junko Matsuzaki
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| | - Lei Wei
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Takemasa Tsuji
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Sebastiano Battaglia
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Eduardo Cortes
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Laiping Wong
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Mark Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Anthony Miliotto
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Nicholas W Bateman
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, John P. Murtha Cancer Center, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA
| | - Shashikant B Lele
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Thinle Chodon
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Richard C Koya
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Qianqian Zhu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Thomas P Conrads
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, John P. Murtha Cancer Center, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA.,Department of Obstetrics and Gynecology, Inova Fairfax Medical Campus, Falls Church, VA, 22003, USA.,Inova Schar Cancer Institute, Falls Church, VA, 22003, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - George L Maxwell
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, John P. Murtha Cancer Center, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA.,Department of Obstetrics and Gynecology, Inova Fairfax Medical Campus, Falls Church, VA, 22003, USA
| | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA. .,Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
268
|
Bast RC, Matulonis UA, Sood AK, Ahmed AA, Amobi AE, Balkwill FR, Wielgos-Bonvallet M, Bowtell DDL, Brenton JD, Brugge JS, Coleman RL, Draetta GF, Doberstein K, Drapkin RI, Eckert MA, Edwards RP, Elias KM, Ennis D, Futreal A, Gershenson DM, Greenberg RA, Huntsman DG, Ji JXY, Kohn EC, Iavarone C, Lengyel ER, Levine DA, Lord CJ, Lu Z, Mills GB, Modugno F, Nelson BH, Odunsi K, Pilsworth JA, Rottapel RK, Powell DJ, Shen L, Shih LM, Spriggs DR, Walton J, Zhang K, Zhang R, Zou L. Critical questions in ovarian cancer research and treatment: Report of an American Association for Cancer Research Special Conference. Cancer 2019; 125:1963-1972. [PMID: 30835824 PMCID: PMC6557260 DOI: 10.1002/cncr.32004] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 12/24/2022]
Abstract
Substantial progress has been made in understanding ovarian cancer at the molecular and cellular level. Significant improvement in 5-year survival has been achieved through cytoreductive surgery, combination platinum-based chemotherapy, and more effective treatment of recurrent cancer, and there are now more than 280,000 ovarian cancer survivors in the United States. Despite these advances, long-term survival in late-stage disease has improved little over the last 4 decades. Poor outcomes relate, in part, to late stage at initial diagnosis, intrinsic drug resistance, and the persistence of dormant drug-resistant cancer cells after primary surgery and chemotherapy. Our ability to accelerate progress in the clinic will depend on the ability to answer several critical questions regarding this disease. To assess current answers, an American Association for Cancer Research Special Conference on "Critical Questions in Ovarian Cancer Research and Treatment" was held in Pittsburgh, Pennsylvania, on October 1-3, 2017. Although clinical, translational, and basic investigators conducted much of the discussion, advocates participated in the meeting, and many presentations were directly relevant to patient care, including treatment with poly adenosine diphosphate ribose polymerase (PARP) inhibitors, attempts to improve immunotherapy by overcoming the immune suppressive effects of the microenvironment, and a better understanding of the heterogeneity of the disease.
Collapse
Affiliation(s)
- Robert C. Bast
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Anil K. Sood
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andrew Futreal
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | | | | - Zhen Lu
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Brad H. Nelson
- University of British Columbia, Canada
- BC Cancer Agency, Canada
| | | | | | | | | | - Li Shen
- Roswell Park Cancer Institute, Buffalo, NY
| | - le-Ming Shih
- Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | | | | | - Lee Zou
- Massachusetts General Hospital, Boston, MD
| |
Collapse
|
269
|
Rossi JF, Céballos P, Lu ZY. Immune precision medicine for cancer: a novel insight based on the efficiency of immune effector cells. Cancer Commun (Lond) 2019; 39:34. [PMID: 31200766 PMCID: PMC6567551 DOI: 10.1186/s40880-019-0379-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/03/2019] [Indexed: 02/06/2023] Open
Abstract
Cancer cell growth is associated with immune surveillance failure. Nowadays, restoring the desired immune response against cancer cells remains a major therapeutic strategy. Due to the recent advances in biological knowledge, efficient therapeutic tools have been developed to support the best bio-clinical approaches for immune precision therapy. One of the most important successes in immune therapy is represented by the applicational use of monoclonal antibodies, particularly the use of rituximab for B-cell lymphoproliferative disorders. More recently, other monoclonal antibodies have been developed, to inhibit immune checkpoints within the tumor microenvironment that limit immune suppression, or to enhance some immune functions with immune adjuvants through different targets such as Toll-receptor agonists. The aim is to inhibit cancer proliferation by the diminishing/elimination of cancer residual cells and clinically improving the response duration with no or few adverse effects. This effect is supported by enhancing the number, functions, and activity of the immune effector cells, including the natural killer (NK) lymphocytes, NKT-lymphocytes, γδ T-lymphocytes, cytotoxic T-lymphocytes, directly or indirectly through vaccines particularly with neoantigens, and by lowering the functions of the immune suppressive cells. Beyond these new therapeutics and their personalized usage, new considerations have to be taken into account, such as epigenetic regulation particularly from microbiota, evaluation of transversal functions, particularly cellular metabolism, and consideration to the clinical consequences at the body level. The aim of this review is to discuss some practical aspects of immune therapy, giving to clinicians the concept of immune effector cells balancing between control and tolerance. Immunological precision medicine is a combination of modern biological knowledge and clinical therapeutic decisions in a global vision of the patient.
Collapse
Affiliation(s)
- Jean-François Rossi
- Institut Sainte Catherine, 84918, Avignon, France. .,Université Montpellier 1, UFR Médecine, 34396, Montpellier, France. .,Département d'Hématologie, CHU de Montpellier, 34295, Montpellier, France.
| | - Patrice Céballos
- Département d'Hématologie, CHU de Montpellier, 34295, Montpellier, France
| | - Zhao-Yang Lu
- Unité de Thérapie Cellulaire, CHU Saint-Eloi, 34295, Montpellier, France
| |
Collapse
|
270
|
Magnin M, Guillaume P, Coukos G, Harari A, Schmidt J. High-throughput identification of human antigen-specific CD8 + and CD4 + T cells using soluble pMHC multimers. Methods Enzymol 2019; 631:21-42. [PMID: 31948548 DOI: 10.1016/bs.mie.2019.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Peptide major histocompatibility complex (pMHC) multimers have been used since decades to identify, isolate and analyze antigen-specific T cells by flow (and more recently mass) cytometry. Yet well established as a standard technology, improvements are still required to face the growing needs of personalized immune monitoring. Here we review the latest developments about (i) the quality of pMHC class I and II monomers, (ii) the importance of the multimeric scaffold, (iii) the staining conditions and (iv) the high-throughput synthesis of pMHC monomers. Finally, innovative multiplexed, combinatorial strategies for parallel detection of antigen-specific T cells in a single sample are discussed.
Collapse
Affiliation(s)
- Morgane Magnin
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Philippe Guillaume
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.
| | - Alexandre Harari
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Julien Schmidt
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| |
Collapse
|
271
|
Abstract
PURPOSE OF REVIEW Despite all efforts, pancreatic ductal adenocarcinoma (PDAC) remains a disease that causes substantial morbidity and mortality, with a 5-year survival rate of 7%. Innovative paradigms for treating PDAC are urgently needed. RECENT FINDINGS We discuss the advances and difficulties in using immunotherapy and developing immunotherapeutic vaccines for PDAC. Current excitement about antigen-specific immunotherapy has been propelled by advances in multiple areas, such as next-generation sequencing to identify neoantigens and manufacturing to produce immunotherapeutic vaccines. Antigen-specific immunotherapy is being actively explored in clinical trials. As the field of immunotherapy matures and as our understanding of the complex interactions between tumor and host develops, we hope to identify new methods for treating and managing PDAC.
Collapse
Affiliation(s)
- Annie A Wu
- Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 488, Baltimore, MD, 21287, USA
| | - Elizabeth Jaffee
- Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 488, Baltimore, MD, 21287, USA
| | - Valerie Lee
- Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 488, Baltimore, MD, 21287, USA.
| |
Collapse
|
272
|
Unleashing Tumour-Dendritic Cells to Fight Cancer by Tackling Their Three A's: Abundance, Activation and Antigen-Delivery. Cancers (Basel) 2019; 11:cancers11050670. [PMID: 31091774 PMCID: PMC6562396 DOI: 10.3390/cancers11050670] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/22/2019] [Accepted: 05/10/2019] [Indexed: 12/24/2022] Open
Abstract
Recent advances in cancer immunotherapy have mainly focused on re-activating T-cell responses against cancer cells. However, both priming and activation of effector T-cell responses against cancer-specific antigens require cross-talk with dendritic cells (DCs), which are responsible for the capturing, processing and presentation of tumour-(neo)antigens to T cells. DCs consequently constitute an essential target in efforts to generate therapeutic immunity against cancer. This review will discuss recent research that is unlocking the cancer-fighting potential of tumour-infiltrating DCs. First, the complexity of DCs in the tumour microenvironment regarding the different subsets and the difficulty of translating mouse data into equivalent human data will be briefly touched upon. Mainly, possible solutions to problems currently faced in DC-based cancer treatments will be discussed, including their infiltration into tumours, activation strategies, and antigen delivery methods. In this way, we hope to put together a broad picture of potential synergistic therapies that could be implemented to harness the full capacity of tumour-infiltrating DCs to stimulate anti-tumour immune responses in patients.
Collapse
|
273
|
Sun X, Zeng L, Huang Y. Transcutaneous delivery of DNA/mRNA for cancer therapeutic vaccination. J Gene Med 2019; 21:e3089. [PMID: 30958606 DOI: 10.1002/jgm.3089] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/17/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
Abstract
Therapeutic vaccination is a promising strategy for the immunotherapy of cancers. It eradicates cancer cells by evoking and strengthening the patient's own immune system. Because of the easy access and sophisticated immune networks, the skin becomes an ideal target organ for vaccination. Genetic vaccines have been widely investigated, with the advantages of the delivery of multiple antigens and a lower cost for production compared to protein/peptide vaccines. This review summarizes the advances made with respect to the transcutaneous delivery of DNA/mRNA for cancer therapeutic vaccination and also gives a brief description of the immunological milieu of the skin and the importance of dendritic cell-targeting in vaccine delivery, as well as the technologies that aim to facilitate antigen delivery and modulate antigen-presenting cells, thus improving cellular responses. The applications of genetic vaccines encoding tumor antigens delivered through the skin route, both in preclinical and clinical trials, are outlined.
Collapse
Affiliation(s)
- Xiaoyi Sun
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Linghui Zeng
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
274
|
Mastelic-Gavillet B, Balint K, Boudousquie C, Gannon PO, Kandalaft LE. Personalized Dendritic Cell Vaccines-Recent Breakthroughs and Encouraging Clinical Results. Front Immunol 2019; 10:766. [PMID: 31031762 PMCID: PMC6470191 DOI: 10.3389/fimmu.2019.00766] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
Abstract
With the advent of combined immunotherapies, personalized dendritic cell (DC)-based vaccination could integrate the current standard of care for the treatment of a large variety of tumors. Due to their proficiency at antigen presentation, DC are key coordinators of the innate and adaptive immune system, and have critical roles in the induction of antitumor immunity. However, despite proven immunogenicity and favorable safety profiles, DC-based immunotherapies have not succeeded at inducing significant objective clinical responses. Emerging data suggest that the combination of DC-based vaccination with other cancer therapies may fully unleash the potential of DC-based cancer vaccines and improve patient survival. In this review, we discuss the recent efforts to develop innovative personalized DC-based vaccines and their use in combined therapies, with a particular focus on ovarian cancer and the promising results of mutanome-based personalized immunotherapies.
Collapse
Affiliation(s)
- Beatris Mastelic-Gavillet
- Department of Oncology, Center for Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Klara Balint
- Department of Oncology, Center for Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Caroline Boudousquie
- Department of Oncology, Center for Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Philippe O Gannon
- Department of Oncology, Center for Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lana E Kandalaft
- Department of Oncology, Center for Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
275
|
Abstract
The phenotype and functionalities of the major immune cell subsets including myeloid cells, macrophages, dendritic cells, and T cells are altered in the ovarian cancer microenvironment. Immunosuppressive networks including inhibitory B7 family members and regulatory T cell-associated adenosine pathway have been defined in human ovarian cancer. In this review, the authors integrate emerging information on immunosuppressive mechanisms and T cell phenotype and discuss strategies of immunotherapeutic and vaccine regimens. Finally, crucial points regarding design of immuno-oncology clinical trials are reviewed.
Collapse
Affiliation(s)
- Weimin Wang
- Department of Surgery, University of Michigan School of Medicine, BSRB 5448, 109 Zina Pitcher Place, Ann Arbor, MI 48109-0669, USA
| | - Janice Rebecca Liu
- Department of Obstetrics and Gynecology, University of Michigan School of Medicine, L4604 WH, 1500 East Medical Center, Ann Arbor, MI 48109, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, BSRB 5071, 109 Zina Pitcher Place, Ann Arbor, MI 48109-0669, USA.
| |
Collapse
|
276
|
Abstract
Effective migration of dendritic cells into the lymphatic system organs is the prerequisite for a functional dendritic cell vaccine. We have previously developed a porous silicon microparticle (PSM)-based therapeutic dendritic cell vaccine (Nano-DC vaccine) where PSM serves both as the vehicle for antigen peptides and an adjuvant. Here, we analyzed parameters that determined dendritic cell uptake of PSM particles and Nano-DC vaccine accumulation in lymphatic tissues in a murine model of HER2-positive breast cancer. Our study revealed a positive correlation between sphericity of the PSM particles and their cellular uptake by circulating dendritic cells. In addition, the intravenously administered vaccines accumulated more in the spleens and inguinal lymph nodes, while the intradermally inoculated vaccines got enriched in the popliteal lymph nodes. Furthermore, mice with large tumors received more vaccines in the lymph nodes than those with small to medium size tumors. Information from this study will provide guidance on design and optimization of future therapeutic cancer vaccines.
Collapse
|
277
|
Corey L, Valente A, Wade K. Personalized Medicine in Gynecologic Cancer: Fact or Fiction? Obstet Gynecol Clin North Am 2019; 46:155-163. [PMID: 30683261 DOI: 10.1016/j.ogc.2018.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Personalized medicine in gynecologic oncology is an evolving field. In recent years, tumor profiling and large databases such as TCGA and NCI-Match have provided us with enormous amounts of molecular data. Several therapies that capitalize on novel genetic and immune discoveries including VEGF inhibitors, PARP inhibitors, and cancer vaccinations are discussed in this article. Additionally, we have seen direct to consumer marketing play an important role in cancer care and prevention as patients have increased ability to access genetic testing. This presents a unique challenge to gynecologic oncology providers as we learn to navigate the world of personalized medicine.
Collapse
Affiliation(s)
- Logan Corey
- Department of Obstetrics and Gynecology, Ochsner Clinic Foundation, 2700 Napoleon Avenue, New Orleans, LA 70115, USA.
| | - Ana Valente
- Department of Obstetrics and Gynecology, Ochsner Clinic Foundation, 2700 Napoleon Avenue, New Orleans, LA 70115, USA
| | - Katrina Wade
- Department of Gynecologic Oncology, Ochsner Clinic Foundation, 2700 Napoleon Avenue, New Orleans, LA 70115, USA
| |
Collapse
|
278
|
Tran TB, Maker VK, Maker AV. Impact of Immunotherapy after Resection of Pancreatic Cancer. J Am Coll Surg 2019; 229:19-27.e1. [PMID: 30742911 DOI: 10.1016/j.jamcollsurg.2019.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 01/28/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Adjuvant immunotherapy has improved outcomes in patients with advanced melanoma; however, the potential benefit for patients with pancreatic ductal adenocarcinoma (PDAC) remains unknown. The aim of this study was to determine the impact of adjuvant chemotherapy and immunotherapy (CTx-IT) compared with CTx alone on patient survival after resection of PDAC. STUDY DESIGN Patients who underwent resection of PDAC from 2004 to 2015 were identified from the National Cancer Database. Univariate and multivariate Cox proportional hazards models were used to determine predictors of overall survival (OS) based on the type of adjuvant therapy received. Patients who received adjuvant immunotherapy were compared with those who received adjuvant CTx alone by propensity score matching. RESULTS Of 21,313 patients who received curative-intent resection for PDAC followed by adjuvant systemic therapy, 269 (1.3%) patients were treated with adjuvant CTx-IT. Propensity score matching resulted in a cohort of 477 patients: (229 CTx only and 248 CTx-IT). The 5-year OS was higher in the CTx-IT group compared with CTx alone (29.2% vs 18.3%; p = 0.0045). On multivariate analysis, the addition of adjuvant immunotherapy was associated was improved overall survival (hazard ratio 0.74; p = 0.007). CONCLUSIONS The addition of adjuvant immunotherapy to chemotherapy is associated with improved survival compared with chemotherapy alone after curative-intent resection of pancreatic adenocarcinoma. Future research is warranted to match specific immunotherapy agents with susceptible patient populations to improve outcomes for this aggressive disease.
Collapse
Affiliation(s)
- Thuy B Tran
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, and the Creticos Cancer Center at Advocate Illinois Masonic Medical Center, Chicago, IL
| | - Vijay K Maker
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, and the Creticos Cancer Center at Advocate Illinois Masonic Medical Center, Chicago, IL
| | - Ajay V Maker
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, and the Creticos Cancer Center at Advocate Illinois Masonic Medical Center, Chicago, IL.
| |
Collapse
|
279
|
Song M, Cubillos-Ruiz JR. Endoplasmic Reticulum Stress Responses in Intratumoral Immune Cells: Implications for Cancer Immunotherapy. Trends Immunol 2019; 40:128-141. [DOI: 10.1016/j.it.2018.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/28/2018] [Accepted: 12/04/2018] [Indexed: 01/08/2023]
|
280
|
Johanns TM, Miller CA, Liu CJ, Perrin RJ, Bender D, Kobayashi DK, Campian JL, Chicoine MR, Dacey RG, Huang J, Fritsch EF, Gillanders WE, Artyomov MN, Mardis ER, Schreiber RD, Dunn GP. Detection of neoantigen-specific T cells following a personalized vaccine in a patient with glioblastoma. Oncoimmunology 2019; 8:e1561106. [PMID: 30906654 PMCID: PMC6422384 DOI: 10.1080/2162402x.2018.1561106] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/27/2018] [Accepted: 12/10/2018] [Indexed: 12/30/2022] Open
Abstract
Neoantigens represent promising targets for personalized cancer vaccine strategies. However, the feasibility of this approach in lower mutational burden tumors like glioblastoma (GBM) remains unknown. We have previously reported the use of an immunogenomics pipeline to identify candidate neoantigens in preclinical models of GBM. Here, we report the application of the same immunogenomics pipeline to identify candidate neoantigens and guide screening for neoantigen-specific T cell responses in a patient with GBM treated with a personalized synthetic long peptide vaccine following autologous tumor lysate DC vaccination. Following vaccination, reactivity to three HLA class I- and five HLA class II-restricted candidate neoantigens were detected by IFN-γ ELISPOT in peripheral blood. A similar pattern of reactivity was observed among isolated post-treatment tumor-infiltrating lymphocytes. Genomic analysis of pre- and post-treatment GBM reflected clonal remodeling. These data demonstrate the feasibility and translational potential of a therapeutic neoantigen-based vaccine approach in patients with primary CNS tumors.
Collapse
Affiliation(s)
- Tanner M Johanns
- Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA.,Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher A Miller
- The McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Connor J Liu
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard J Perrin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Diane Bender
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Dale K Kobayashi
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Jian L Campian
- Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael R Chicoine
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Ralph G Dacey
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jiayi Huang
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - William E Gillanders
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.,Department of Surgery, Section of Endocrine and Oncologic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Maxim N Artyomov
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Elaine R Mardis
- Institute for Genomic Medicine, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - Robert D Schreiber
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gavin P Dunn
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
281
|
Moufarrij S, Dandapani M, Arthofer E, Gomez S, Srivastava A, Lopez-Acevedo M, Villagra A, Chiappinelli KB. Epigenetic therapy for ovarian cancer: promise and progress. Clin Epigenetics 2019; 11:7. [PMID: 30646939 PMCID: PMC6334391 DOI: 10.1186/s13148-018-0602-0] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/19/2018] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer is the deadliest gynecologic malignancy, with a 5-year survival rate of approximately 47%, a number that has remained constant over the past two decades. Early diagnosis improves survival, but unfortunately only 15% of ovarian cancers are diagnosed at an early or localized stage. Most ovarian cancers are epithelial in origin and treatment prioritizes surgery and cytoreduction followed by cytotoxic platinum and taxane chemotherapy. While most tumors will initially respond to this treatment, recurrence is likely to occur within a median of 16 months for patients who present with advanced stage disease. New treatment options separate from traditional chemotherapy that take advantage of advances in understanding of the pathophysiology of ovarian cancer are needed to improve outcomes. Recent work has shown that mutations in genes encoding epigenetic regulators are mutated in ovarian cancer, driving tumorigenesis and resistance to treatment. Several of these epigenetic modifiers have emerged as promising drug targets for ovarian cancer therapy. In this article, we delineate epigenetic abnormalities in ovarian cancer, discuss key scientific advances using epigenetic therapies in preclinical ovarian cancer models, and review ongoing clinical trials utilizing epigenetic therapies in ovarian cancer.
Collapse
Affiliation(s)
- Sara Moufarrij
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- Department of Obstetrics & Gynecology, The George Washington University, Washington, D.C., 20052 USA
- Department of Biochemistry & Molecular Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Monica Dandapani
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- Department of Obstetrics & Gynecology, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Elisa Arthofer
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Stephanie Gomez
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Aneil Srivastava
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Micael Lopez-Acevedo
- Department of Obstetrics & Gynecology, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Alejandro Villagra
- Department of Biochemistry & Molecular Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Katherine B. Chiappinelli
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| |
Collapse
|
282
|
IL-15 and a Two-Step Maturation Process Improve Bone Marrow-Derived Dendritic Cell Cancer Vaccine. Cancers (Basel) 2019; 11:cancers11010040. [PMID: 30621204 PMCID: PMC6356194 DOI: 10.3390/cancers11010040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
In the last 20 years, dendritic cells (DCs) have been largely used as a platform for therapeutic vaccination in cancer patients. However, despite its proven safety and ability to induce cancer specific immune responses, the clinical benefits of DC-based immunotherapy are currently very limited. Thus, novel approaches are still needed to boost its efficacy. Our group recently showed that squaric acid treatment of antigens is an important adjuvant that can increase vaccine-induced downstream immune responses and therapeutic outcomes. Here we further improved this dendritic cell vaccine formulation by developing a new method for differentiating and maturing DCs from their bone marrow precursors. Our data demonstrate that bone marrow-derived DCs differentiated with GM-CSF and IL-15 and matured with a maturation cocktail in two steps present a more mature and immunogenic phenotype, compared to standard DC preparations. Further suppression of the prostaglandin E₂ pathway achieved even more immunogenic DC phenotypes. This vaccine was more potent at delaying tumor growth, improved animal survival and induced a more immunogenic and Th1-skewed T cell response in an ovarian cancer mouse model. These promising results support future efforts for the clinical translation of this approach.
Collapse
|
283
|
Levinson K, Dorigo O, Rubin K, Moore K. Immunotherapy in Gynecologic Cancers: What We Know Now and Where We Are Headed. Am Soc Clin Oncol Educ Book 2019; 39:e126-e140. [PMID: 31099679 DOI: 10.1200/edbk_237967] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Immunotherapy, mainly in the form of immune checkpoint inhibitors (ICIs), has been transformative in both solid tumor and hematologic malignancies. Patients with previously terminal illnesses have experienced profound responses of great durability with these agents, fueling excitement among patients and providers regarding their use. Unfortunately, the gains seen in some solid tumors have not been replicated in a large percentage of patients with gynecologic cancer. This review focuses on the clinical benefits seen to date, toxicities and management when using ICIs, ways to improve prediction of who should receive immunotherapy, and a discussion of next-generation immunotherapy with cellular therapeutics and how these might relate to gynecologic cancers.
Collapse
Affiliation(s)
- Kimberly Levinson
- 1 The Kelly Gynecologic Oncology Service, Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | - Kathleen Moore
- 4 Stephenson Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
284
|
Wolf Y, Samuels Y. Cancer research in the era of immunogenomics. ESMO Open 2018; 3:e000475. [PMID: 30622743 PMCID: PMC6307593 DOI: 10.1136/esmoopen-2018-000475] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 12/14/2022] Open
Abstract
The most meaningful advancement in cancer treatment in recent years has been the emergence of immunotherapy. Checkpoint inhibitor blockade and adoptive T cell therapy have shown remarkable clinical effects in a wide range of tumour types. Despite these advances, many tumours do not respond to these treatments, which raises the need to further investigate how patients can benefit from immunotherapy. This effort can now take advantage of the recent technological progress in single-cell, high-throughput sequencing and computational efforts. In this review, we will discuss advances in different immunotherapies and the principles of cancer immunogenomics, with an emphasis on the detection of cancer neoantigens with human leucocyte antigen peptidomics, and how these principles can be further used for more efficient clinical output.
Collapse
Affiliation(s)
- Yochai Wolf
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
285
|
Jiang L, Fang X, Wang H, Li D, Wang X. Ovarian Cancer-Intrinsic Fatty Acid Synthase Prevents Anti-tumor Immunity by Disrupting Tumor-Infiltrating Dendritic Cells. Front Immunol 2018; 9:2927. [PMID: 30619288 PMCID: PMC6302125 DOI: 10.3389/fimmu.2018.02927] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Fatty acid synthase (FASN), the key metabolic enzyme of de novo lipogenesis, provides proliferative and metastatic capacity directly to cancer cells have been described. However, the impact of aberrant activation of this lipogenic enzyme on host anti-tumor immune milieu remains unknown. In this study, we depicted that elevated FASN expression presented in ovarian cancer with more advanced clinical phenotype and correlated with the immunosuppressive status, which characterized by the lower number and dysfunction of infiltrating T cells. Notably, in a mouse model, we showed that tumor cell-intrinsic FASN drove ovarian cancer (OvCa) progression by blunting anti-tumor immunity. Dendritic cells (DCs) are required to initiate and sustain T cell-dependent anti-tumor immunity. Here, our data showed that constitutive activation of FASN in ovarian cancer cell lead to abnormal lipid accumulation and subsequent inhibition of tumor-infiltrating DCs (TIDCs) capacity to support anti-tumor T cells. Mechanistically, FASN activation in ovarian cancer cell-induced the resulting increase of lipids present at high concentrations in the tumor microenvironment. Dendritic cells educated by FASNhigh OvCa ascites are defective in their ability to present antigens and prime T cells. Accordingly, inhibiting FASN by FASN inhibitor can partly restore the immunostimulatory activity of TIDCs and extended tumor control by evoking protective anti-tumor immune responses. Therefore, our data provide a mechanism by which ovarian cancer-intrinsic FASN oncogenic pathway induce the impaired anti-tumor immune response through lipid accumulation in TIDCs and subsequently T-cells exclusion and dysfunction. These results could further indicate that targeting the FASN oncogenic pathway concomitantly enhance anti-tumor immunity, thus offering a unique approach to ovarian cancer immunotherapy.
Collapse
Affiliation(s)
- Li Jiang
- Department of Gynecology and Obstetrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuhong Fang
- Department of Gynecology and Obstetrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Wang
- Department of Gynecology and Obstetrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Diyou Li
- Department of Gynecology and Obstetrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xipeng Wang
- Department of Gynecology and Obstetrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
286
|
Abstract
Malignant transformation of cells depends on accumulation of DNA damage. Over the past years we have learned that the T cell-based immune system frequently responds to the neoantigens that arise as a consequence of this DNA damage. Furthermore, recognition of neoantigens appears an important driver of the clinical activity of both T cell checkpoint blockade and adoptive T cell therapy as cancer immunotherapies. Here we review the evidence for the relevance of cancer neoantigens in tumor control and the biological properties of these antigens. We discuss recent technological advances utilized to identify neoantigens, and the T cells that recognize them, in individual patients. Finally, we discuss strategies that can be employed to exploit cancer neoantigens in clinical interventions.
Collapse
Affiliation(s)
- Ton N Schumacher
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; , .,Oncode Institute, 3521AL Utrecht, The Netherlands
| | - Wouter Scheper
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; , .,Oncode Institute, 3521AL Utrecht, The Netherlands
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; ,
| |
Collapse
|
287
|
van Gulijk M, Dammeijer F, Aerts JGJV, Vroman H. Combination Strategies to Optimize Efficacy of Dendritic Cell-Based Immunotherapy. Front Immunol 2018; 9:2759. [PMID: 30568653 PMCID: PMC6289976 DOI: 10.3389/fimmu.2018.02759] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/09/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells (APCs) that are essential for the activation of immune responses. In various malignancies, these immunostimulatory properties are exploited by DC-therapy, aiming at the induction of effective anti-tumor immunity by vaccination with ex vivo antigen-loaded DCs. Depending on the type of DC-therapy used, long-term clinical efficacy upon DC-therapy remains restricted to a proportion of patients, likely due to lack of immunogenicity of tumor cells, presence of a stromal compartment, and the suppressive tumor microenvironment (TME), thereby leading to the development of resistance. In order to circumvent tumor-induced suppressive mechanisms and unleash the full potential of DC-therapy, considerable efforts have been made to combine DC-therapy with chemotherapy, radiotherapy or with checkpoint inhibitors. These combination strategies could enhance tumor immunogenicity, stimulate endogenous DCs following immunogenic cell death, improve infiltration of cytotoxic T lymphocytes (CTLs) or specifically deplete immunosuppressive cells in the TME, such as regulatory T-cells and myeloid-derived suppressor cells. In this review, different strategies of combining DC-therapy with immunomodulatory treatments will be discussed. These strategies and insights will improve and guide DC-based combination immunotherapies with the aim of further improving patient prognosis and care.
Collapse
Affiliation(s)
- Mandy van Gulijk
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Floris Dammeijer
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
288
|
Bekeschus S, Clemen R, Metelmann HR. Potentiating anti-tumor immunity with physical plasma. CLINICAL PLASMA MEDICINE 2018. [DOI: 10.1016/j.cpme.2018.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
289
|
Roelofsen T, Wefers C, Gorris MAJ, Textor JC, Massuger LFAG, de Vries IJM, van Altena AM. Spontaneous Regression of Ovarian Carcinoma After Septic Peritonitis; A Unique Case Report. Front Oncol 2018; 8:562. [PMID: 30555799 PMCID: PMC6281979 DOI: 10.3389/fonc.2018.00562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/12/2018] [Indexed: 11/23/2022] Open
Abstract
Despite advances in therapy, ovarian cancer remains the most lethal gynecological malignancy and prognosis has not substantially improved over the past 3 decades. Immunotherapy is a promising new treatment option. However, the immunosuppressive cancer microenvironment must be overcome for immunotherapy to be successful. Here, we present a unique case of spontaneous regression of ovarian carcinoma after septic peritonitis. A 79-year-old woman was diagnosed with stage IIIc ovarian cancer. The omental cake biopsy was complicated by sepsis. Although the patient recovered, her physical condition did not allow further treatment for her ovarian cancer. After 6 months, spontaneous regression of the tumor was observed during surgery. Analysis of the immune infiltrate in the tissues showed a shift from a pro-tumorigenic to an anti-tumorigenic immune response after sepsis. Strong activation of the immune system during sepsis overruled the immunosuppressive tumor microenvironment and allowed for a potent anti-tumor immune response. More understanding of immunological responses in cases with cancer and septic peritonitis might be crucial to identify potential new targets for immunotherapy.
Collapse
Affiliation(s)
- Thijs Roelofsen
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Christina Wefers
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Mark A. J. Gorris
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Johannes C. Textor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Leon F. A. G. Massuger
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Anne M. van Altena
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
290
|
Martin Lluesma S, Graciotti M, Chiang CLL, Kandalaft LE. Does the Immunocompetent Status of Cancer Patients Have an Impact on Therapeutic DC Vaccination Strategies? Vaccines (Basel) 2018; 6:E79. [PMID: 30477198 PMCID: PMC6313858 DOI: 10.3390/vaccines6040079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/09/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022] Open
Abstract
Although different types of therapeutic vaccines against established cancerous lesions in various indications have been developed since the 1990s, their clinical benefit is still very limited. This observed lack of effectiveness in cancer eradication may be partially due to the often deficient immunocompetent status of cancer patients, which may facilitate tumor development by different mechanisms, including immune evasion. The most frequently used cellular vehicle in clinical trials are dendritic cells (DCs), thanks to their crucial role in initiating and directing immune responses. Viable vaccination options using DCs are available, with a positive toxicity profile. For these reasons, despite their limited therapeutic outcomes, DC vaccination is currently considered an additional immunotherapeutic option that still needs to be further explored. In this review, we propose potential actions aimed at improving DC vaccine efficacy by counteracting the detrimental mechanisms recognized to date and implicated in establishing a poor immunocompetent status in cancer patients.
Collapse
Affiliation(s)
- Silvia Martin Lluesma
- Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Michele Graciotti
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| | - Cheryl Lai-Lai Chiang
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| |
Collapse
|
291
|
Deniger DC, Pasetto A, Robbins PF, Gartner JJ, Prickett TD, Paria BC, Malekzadeh P, Jia L, Yossef R, Langhan MM, Wunderlich JR, Danforth DN, Somerville RPT, Rosenberg SA. T-cell Responses to TP53 "Hotspot" Mutations and Unique Neoantigens Expressed by Human Ovarian Cancers. Clin Cancer Res 2018; 24:5562-5573. [PMID: 29853601 PMCID: PMC6239943 DOI: 10.1158/1078-0432.ccr-18-0573] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/18/2018] [Accepted: 05/24/2018] [Indexed: 12/30/2022]
Abstract
Purpose: This was a study prospectively evaluating intratumoral T-cell responses to autologous somatic mutated neoepitopes expressed by human metastatic ovarian cancers.Patients and Methods: Tumor-infiltrating lymphocytes (TIL) were expanded from resected ovarian cancer metastases, which were analyzed by whole-exome and transcriptome sequencing to identify autologous somatic mutations. All mutated neoepitopes, independent of prediction algorithms, were expressed in autologous antigen-presenting cells and then cocultured with TIL fragment cultures. Secretion of IFNγ or upregulation of 41BB indicated a T-cell response.Results: Seven women with metastatic ovarian cancer were evaluated, and 5 patients had clear, dominant T-cell responses to mutated neoantigens, which were corroborated by comparison with the wild-type sequence, identification of the minimal epitope, human leukocyte antigen (HLA) restriction element(s), and neoantigen-specific T-cell receptor(s). Mutated neoantigens were restricted by HLA-B, -C, -DP, -DQ, and/or -DR alleles and appeared to principally arise from random, somatic mutations unique to each patient. We established that TP53 "hotspot" mutations (c.659A>G; p.Y220C and c.733G>A; p.G245S) expressed by two different patients' tumors were both immunogenic in the context of HLA-DRB3*02:02.Conclusions: Mutation-reactive T cells infiltrated ovarian cancer metastases at sufficient frequencies to warrant their investigation as adoptive cell therapy. In addition, transfer of TP53 "hotspot" mutation-reactive T-cell receptors into peripheral blood T cells could be evaluated as a gene therapy for a diverse range of tumor histologies. Clin Cancer Res; 24(22); 5562-73. ©2018 AACR See related commentary by McNeish, p. 5493.
Collapse
MESH Headings
- Amino Acid Sequence
- Antigens, Neoplasm/immunology
- Cell Line, Tumor
- Epitope Mapping
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- HLA Antigens/immunology
- Histones/genetics
- Histones/immunology
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Mutation
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/pathology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/immunology
- Ubiquitin Thiolesterase/genetics
- Ubiquitin Thiolesterase/immunology
Collapse
Affiliation(s)
- Drew C Deniger
- Surgery Branch, National Cancer Institute, Bethesda, Maryland
| | - Anna Pasetto
- Surgery Branch, National Cancer Institute, Bethesda, Maryland
| | - Paul F Robbins
- Surgery Branch, National Cancer Institute, Bethesda, Maryland
| | - Jared J Gartner
- Surgery Branch, National Cancer Institute, Bethesda, Maryland
| | - Todd D Prickett
- Surgery Branch, National Cancer Institute, Bethesda, Maryland
| | - Biman C Paria
- Surgery Branch, National Cancer Institute, Bethesda, Maryland
| | | | - Li Jia
- Surgery Branch, National Cancer Institute, Bethesda, Maryland
| | - Rami Yossef
- Surgery Branch, National Cancer Institute, Bethesda, Maryland
| | | | | | | | | | | |
Collapse
|
292
|
Tanyi JL, George E. Personalized vaccination against ovarian cancer: what are the possibilities? Expert Rev Vaccines 2018; 17:955-958. [PMID: 30362844 DOI: 10.1080/14760584.2018.1541743] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Janos L Tanyi
- a Department of Obstetrics and Gynecology , University of Pennsylvania Perelman School of Medicine , Philadelphia , USA
| | - Erin George
- a Department of Obstetrics and Gynecology , University of Pennsylvania Perelman School of Medicine , Philadelphia , USA
| |
Collapse
|
293
|
Weber ANR, Cardona Gloria Y, Çınar Ö, Reinhardt HC, Pezzutto A, Wolz OO. Oncogenic MYD88 mutations in lymphoma: novel insights and therapeutic possibilities. Cancer Immunol Immunother 2018; 67:1797-1807. [PMID: 30203262 PMCID: PMC11028221 DOI: 10.1007/s00262-018-2242-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023]
Abstract
Oncogenic MYD88 mutations, most notably the Leu 265 Pro (L265P) mutation, were recently identified as potential driver mutations in various B-cell non-Hodgkin Lymphomas (NHLs). The L265P mutation is now thought to be common to virtually all NHLs and occurs in between 4 and 90% of cases, depending on the entity. Since it is tumor-specific, the mutation, and the pathways it regulates, might serve as advantageous therapeutic targets for both conventional chemotherapeutic intervention, as well as immunotherapeutic strategies. Here, we review recent progress on elucidating the molecular and cellular processes affected by the L265P mutation of MYD88, describe a new in vivo model for MyD88 L265P-mediated oncogenesis, and summarize how these findings could be exploited therapeutically by specific targeting of signaling pathways. In addition, we summarize current and explore future possibilities for conceivable immunotherapeutic approaches, such as L265P-derived peptide vaccination, adoptive transfer of L265P-restricted T cells, and use of T-cell receptor-engineered T cells. With clinical trials regarding their efficacy rapidly expanding to NHLs, we also discuss potential combinations of immune checkpoint inhibitors with the described targeted chemotherapies of L265P signaling networks, and/or with the above immunological approaches as potential ways of targeting MYD88-mutated lymphomas in the future.
Collapse
Affiliation(s)
- Alexander N R Weber
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
| | - Yamel Cardona Gloria
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Özcan Çınar
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - H Christian Reinhardt
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Antonio Pezzutto
- Berlin Institute for Health Charité and Max-Delbrück Center, Campus Buch, Building 42-53, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité Medical School, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Olaf-Oliver Wolz
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
| |
Collapse
|
294
|
Empowering dendritic cell cancer vaccination: the role of combinatorial strategies. Cytotherapy 2018; 20:1309-1323. [PMID: 30360963 DOI: 10.1016/j.jcyt.2018.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs) are bone marrow-derived immune cells that play a crucial role in inducing the adaptive immunity and supporting the innate immune response independently from T cells. In the last decade, DCs have become a hopeful instrument for cancer vaccines that aims at re-educating the immune system, leading to a potent anti-cancer immune response able to overcome the immunosuppressive tumor microenvironment (TME). Although several studies have indicated that DC-based vaccines are feasible and safe, the clinical advantages of DC vaccination as monotherapy for most of the neoplasms remain a distant target. Recently, many reports and clinical trials have widely used innovative combinatorial therapeutic strategies to normalize the immune function in the TME and synergistically enhance DC function. This review will describe the most relevant and updated evidence of the anti-cancer combinatorial approaches to boost the clinical potency of DC-based vaccines.
Collapse
|
295
|
Chodon T, Lugade AA, Battaglia S, Odunsi K. Emerging Role and Future Directions of Immunotherapy in Advanced Ovarian Cancer. Hematol Oncol Clin North Am 2018; 32:1025-1039. [PMID: 30390758 DOI: 10.1016/j.hoc.2018.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Clinical progress in cancer immunotherapy has been slow; however, within the last 5 years, breakthrough successes have brought immunotherapy to the forefront in cancer therapy. Promising results have been observed in solid tumors and hematologic malignancies. Most treatment modalities have shown limited efficacy as monotherapy. The complex nature of cancer and the immunosuppressive microenvironment emphasizes the need to personalize immunotherapy by manipulating the patient's own immune system. For successful and long-lasting cure of cancer, a multimodal approach is essential, combining antitumor cell therapy with manipulation of multiple pathways in the tumor microenvironment to ameliorate tumor-induced immunosuppression.
Collapse
Affiliation(s)
- Thinle Chodon
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Sebastiano Battaglia
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
296
|
Rodriguez J, Castañón E, Perez-Gracia JL, Rodriguez I, Viudez A, Alfaro C, Oñate C, Perez G, Rotellar F, Inogés S, López-Diaz de Cerio A, Resano L, Ponz-Sarvise M, Rodriguez-Ruiz ME, Chopitea A, Vera R, Melero I. A randomized phase II clinical trial of dendritic cell vaccination following complete resection of colon cancer liver metastasis. J Immunother Cancer 2018; 6:96. [PMID: 30268156 PMCID: PMC6164167 DOI: 10.1186/s40425-018-0405-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022] Open
Abstract
Surgically resectable synchronic and metachronic liver metastases of colon cancer have high risk of relapse in spite of standard-of-care neoadjuvant and adjuvant chemotherapy regimens. Dendritic cell vaccines loaded with autologous tumor lysates were tested for their potential to avoid or delay disease relapses (NCT01348256). Patients with surgically amenable liver metastasis of colon adenocarcinoma (n = 19) were included and underwent neoadjuvant chemotherapy, surgery and adjuvant chemotherapy. Fifteen patients with disease-free resection margins were randomized 1:1 to receive two courses of four daily doses of dendritic cell intradermal vaccinations versus observation. The trial had been originally designed to include 56 patients but was curtailed due to budgetary restrictions. Follow-up of the patients indicates a clear tendency to fewer and later relapses in the vaccine arm (median disease free survival –DFS-) 25.26 months, 95% CI 8.74-n.r) versus observation arm (median DFS 9.53 months, 95% CI 5.32–18.88).
Collapse
Affiliation(s)
- Javier Rodriguez
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain
| | - Eduardo Castañón
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain
| | - Jose Luis Perez-Gracia
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain.,CIBERONC, Madrid, Spain.,Instituto de investigación de Navarra, IDISNA, Pamplona, Spain
| | - Inmaculada Rodriguez
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain.,CIBERONC, Madrid, Spain
| | - Antonio Viudez
- Complejo Hospitalario de Navarra, Avenida Irunlarrea 5, 31008, Pamplona, Spain.,Instituto de investigación de Navarra, IDISNA, Pamplona, Spain
| | - Carlos Alfaro
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain.,CIBERONC, Madrid, Spain
| | - Carmen Oñate
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain
| | - Guiomar Perez
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain
| | - Fernando Rotellar
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain
| | - Susana Inogés
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain.,Instituto de investigación de Navarra, IDISNA, Pamplona, Spain
| | - Ascensión López-Diaz de Cerio
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain.,Instituto de investigación de Navarra, IDISNA, Pamplona, Spain
| | - Leyre Resano
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain
| | - Mariano Ponz-Sarvise
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain.,Centro de Investigacion Medica Aplicada, CIMA, Avenida Pio XII, 36, 31008, Pamplona, Spain.,Instituto de investigación de Navarra, IDISNA, Pamplona, Spain
| | - Maria E Rodriguez-Ruiz
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain.,Centro de Investigacion Medica Aplicada, CIMA, Avenida Pio XII, 36, 31008, Pamplona, Spain.,CIBERONC, Madrid, Spain.,Instituto de investigación de Navarra, IDISNA, Pamplona, Spain
| | - Ana Chopitea
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain
| | - Ruth Vera
- Complejo Hospitalario de Navarra, Avenida Irunlarrea 5, 31008, Pamplona, Spain.,Instituto de investigación de Navarra, IDISNA, Pamplona, Spain
| | - Ignacio Melero
- Clinica Universidad de Navarra, Avenida Pio XII, 36, 31008, Pamplona, Spain. .,Centro de Investigacion Medica Aplicada, CIMA, Avenida Pio XII, 36, 31008, Pamplona, Spain. .,CIBERONC, Madrid, Spain. .,Instituto de investigación de Navarra, IDISNA, Pamplona, Spain. .,, Pamplona, Spain.
| |
Collapse
|
297
|
Liu M, Guo F. Recent updates on cancer immunotherapy. PRECISION CLINICAL MEDICINE 2018; 1:65-74. [PMID: 30687562 PMCID: PMC6333045 DOI: 10.1093/pcmedi/pby011] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/23/2018] [Accepted: 08/01/2018] [Indexed: 02/05/2023] Open
Abstract
Traditional cancer therapies include surgery, radiation, and chemotherapy, all of which are typically non-specific approaches. Cancer immunotherapy is a type of cancer treatment that helps the immune system fight cancer. Cancer immunotherapy represents a standing example of precision medicine: immune checkpoint inhibitors precisely target the checkpoints; tumor infiltrating lymphocytes, TCR T cells, and CAR T cells precisely kill cancer cells through tumor antigen recognition; and cancer vaccines are made from patient-derived dendritic cells, tumor cell DNA, or RNA, or oncolytic viruses, thus offering a type of personalized medicine. This review will highlight up-to-date advancement in most, if not all, of the immunotherapy strategies.
Collapse
Affiliation(s)
- Ming Liu
- Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
298
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018; 10:E302. [PMID: 30200478 PMCID: PMC6162424 DOI: 10.3390/cancers10090302] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
299
|
Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
|
300
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|