151
|
Martincorena I, Raine KM, Gerstung M, Dawson KJ, Haase K, Van Loo P, Davies H, Stratton MR, Campbell PJ. Universal Patterns of Selection in Cancer and Somatic Tissues. Cell 2017; 171:1029-1041.e21. [PMID: 29056346 PMCID: PMC5720395 DOI: 10.1016/j.cell.2017.09.042] [Citation(s) in RCA: 817] [Impact Index Per Article: 116.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/09/2017] [Accepted: 09/22/2017] [Indexed: 01/17/2023]
Abstract
Cancer develops as a result of somatic mutation and clonal selection, but quantitative measures of selection in cancer evolution are lacking. We adapted methods from molecular evolution and applied them to 7,664 tumors across 29 cancer types. Unlike species evolution, positive selection outweighs negative selection during cancer development. On average, <1 coding base substitution/tumor is lost through negative selection, with purifying selection almost absent outside homozygous loss of essential genes. This allows exome-wide enumeration of all driver coding mutations, including outside known cancer genes. On average, tumors carry ∼4 coding substitutions under positive selection, ranging from <1/tumor in thyroid and testicular cancers to >10/tumor in endometrial and colorectal cancers. Half of driver substitutions occur in yet-to-be-discovered cancer genes. With increasing mutation burden, numbers of driver mutations increase, but not linearly. We systematically catalog cancer genes and show that genes vary extensively in what proportion of mutations are drivers versus passengers.
Collapse
Affiliation(s)
| | - Keiran M Raine
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridgeshire, UK
| | - Moritz Gerstung
- European Molecular Biology Laboratory, European Bioinformatics Institute EMBL-EBI, Hinxton CB10 1SD, UK
| | - Kevin J Dawson
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridgeshire, UK
| | | | - Peter Van Loo
- The Francis Crick Institute, London NW1 1AT, UK; Department of Human Genetics, University of Leuven, Leuven 3000, Belgium
| | - Helen Davies
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridgeshire, UK
| | | | - Peter J Campbell
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridgeshire, UK; Department of Haematology, University of Cambridge, Cambridge CB2 2XY, UK.
| |
Collapse
|
152
|
Bjerregaard AM, Nielsen M, Jurtz V, Barra CM, Hadrup SR, Szallasi Z, Eklund AC. An Analysis of Natural T Cell Responses to Predicted Tumor Neoepitopes. Front Immunol 2017; 8:1566. [PMID: 29187854 PMCID: PMC5694748 DOI: 10.3389/fimmu.2017.01566] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/01/2017] [Indexed: 12/30/2022] Open
Abstract
Personalization of cancer immunotherapies such as therapeutic vaccines and adoptive T-cell therapy may benefit from efficient identification and targeting of patient-specific neoepitopes. However, current neoepitope prediction methods based on sequencing and predictions of epitope processing and presentation result in a low rate of validation, suggesting that the determinants of peptide immunogenicity are not well understood. We gathered published data on human neopeptides originating from single amino acid substitutions for which T cell reactivity had been experimentally tested, including both immunogenic and non-immunogenic neopeptides. Out of 1,948 neopeptide-HLA (human leukocyte antigen) combinations from 13 publications, 53 were reported to elicit a T cell response. From these data, we found an enrichment for responses among peptides of length 9. Even though the peptides had been pre-selected based on presumed likelihood of being immunogenic, we found using NetMHCpan-4.0 that immunogenic neopeptides were predicted to bind significantly more strongly to HLA compared to non-immunogenic peptides. Investigation of the HLA binding strength of the immunogenic peptides revealed that the vast majority (96%) shared very strong predicted binding to HLA and that the binding strength was comparable to that observed for pathogen-derived epitopes. Finally, we found that neopeptide dissimilarity to self is a predictor of immunogenicity in situations where neo- and normal peptides share comparable predicted binding strength. In conclusion, these results suggest new strategies for prioritization of mutated peptides, but new data will be needed to confirm their value.
Collapse
Affiliation(s)
- Anne-Mette Bjerregaard
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Morten Nielsen
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark.,Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Vanessa Jurtz
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Carolina M Barra
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Sine Reker Hadrup
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Zoltan Szallasi
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark.,Computational Health Informatics Program (CHIP), Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Aron Charles Eklund
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
153
|
Determining T-cell specificity to understand and treat disease. Nat Biomed Eng 2017; 1:784-795. [DOI: 10.1038/s41551-017-0143-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023]
|
154
|
van Attekum MHA, van Bruggen JAC, Slinger E, Lebre MC, Reinen E, Kersting S, Eldering E, Kater AP. CD40 signaling instructs chronic lymphocytic leukemia cells to attract monocytes via the CCR2 axis. Haematologica 2017; 102:2069-2076. [PMID: 28971904 PMCID: PMC5709106 DOI: 10.3324/haematol.2016.157206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/22/2017] [Indexed: 01/23/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) cells are provided with essential survival and proliferative signals in the lymph node microenvironment. Here, CLL cells engage in various interactions with bystander cells such as T cells and macrophages. Phenotypically distinct types of tumor infiltrating macrophages can either be tumor supportive (M2) or play a role in tumor immune surveillance (M1). Although recent in vitro findings suggest a protective role for macrophages in CLL, the actual balance between these macrophage subsets in CLL lymphoid tissue is still unclear. Furthermore, the mechanism of recruitment of monocytes towards the CLL lymph node is currently unknown. Both questions are addressed in this paper. Immunofluorescence staining of lymph node samples showed macrophage skewing towards an M2 tumor-promoting phenotype. This polarization likely results from CLL-secreted soluble factors, as both patient serum and CLL-conditioned medium recapitulated the skewing effect. Considering that CLL cell cytokine secretion is affected by adjacent T cells, we next studied CLL-mediated monocyte recruitment in the presence or absence of T-cell signals. While unstimulated CLL cells were inactive, T cell-stimulated CLL cells actively recruited monocytes. This correlated with secretion of various chemokines such as C-C-motif-ligand-2,3,4,5,7,24, C-X-C-motif-ligand-5,10, and Interleukin-10. We also identified CD40L as the responsible T-cell factor that mediated recruitment, and showed that recruitment critically depended on the C-C-motif-chemokine-receptor-2 axis. These studies show that the shaping of a tumor supportive microenvironment depends on cytokinome alterations (including C-C-motif-ligand-2) that occur after interactions between CLL, T cells and monocytes. Therefore, targeted inhibition of CD40L or C-C-motif-chemokine-receptor-2 may be relevant therapeutic options.
Collapse
Affiliation(s)
- Martijn H A van Attekum
- Department of Hematology, Academic Medical Center, University of Amsterdam; the Netherlands.,Department of Experimental Immunology, Academic Medical Center, University of Amsterdam; the Netherlands
| | - Jaco A C van Bruggen
- Department of Hematology, Academic Medical Center, University of Amsterdam; the Netherlands.,Department of Experimental Immunology, Academic Medical Center, University of Amsterdam; the Netherlands
| | - Erik Slinger
- Department of Hematology, Academic Medical Center, University of Amsterdam; the Netherlands.,Department of Experimental Immunology, Academic Medical Center, University of Amsterdam; the Netherlands
| | - M Cristina Lebre
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam; the Netherlands
| | - Emilie Reinen
- Department of Hematology, Academic Medical Center, University of Amsterdam; the Netherlands.,Department of Experimental Immunology, Academic Medical Center, University of Amsterdam; the Netherlands
| | - Sabina Kersting
- Department of Hematology, Haga Teaching Hospital, The Hague, the Netherlands
| | - Eric Eldering
- Department of Hematology, Haga Teaching Hospital, The Hague, the Netherlands.,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), the Netherlands
| | - Arnon P Kater
- Department of Hematology, Academic Medical Center, University of Amsterdam; the Netherlands .,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), the Netherlands
| |
Collapse
|
155
|
Kalina JL, Neilson DS, Lin YY, Hamilton PT, Comber AP, Loy EMH, Sahinalp SC, Collins CC, Hach F, Lum JJ. Mutational Analysis of Gene Fusions Predicts Novel MHC Class I-Restricted T-Cell Epitopes and Immune Signatures in a Subset of Prostate Cancer. Clin Cancer Res 2017; 23:7596-7607. [PMID: 28954787 DOI: 10.1158/1078-0432.ccr-17-0618] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/25/2017] [Accepted: 09/20/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Gene fusions are frequently found in prostate cancer and may result in the formation of unique chimeric amino acid sequences (CASQ) that span the breakpoint of two fused gene products. This study evaluated the potential for fusion-derived CASQs to be a source of tumor neoepitopes, and determined their relationship to patterns of immune signatures in prostate cancer patients.Experimental Design: A computational strategy was used to identify CASQs and their corresponding predicted MHC class I epitopes using RNA-Seq data from The Cancer Genome Atlas of prostate tumors. In vitro peptide-specific T-cell expansion was performed to identify CASQ-reactive T cells. A multivariate analysis was used to relate patterns of in silico-predicted tumor-infiltrating immune cells with prostate tumors harboring these mutational events.Results: Eighty-seven percent of tumors contained gene fusions with a mean of 12 per tumor. In total, 41% of fusion-positive tumors were found to encode CASQs. Within these tumors, 87% gave rise to predicted MHC class I-binding epitopes. This observation was more prominent when patients were stratified into low- and intermediate/high-risk categories. One of the identified CASQ from the recurrent TMPRSS2:ERG type VI fusion contained several high-affinity HLA-restricted epitopes. These peptides bound HLA-A*02:01 in vitro and were recognized by CD8+ T cells. Finally, the presence of fusions and CASQs were associated with expression of immune cell infiltration.Conclusions: Mutanome analysis of gene fusion-derived CASQs can give rise to patient-specific predicted neoepitopes. Moreover, these fusions predicted patterns of immune cell infiltration within a subgroup of prostate cancer patients. Clin Cancer Res; 23(24); 7596-607. ©2017 AACR.
Collapse
Affiliation(s)
- Jennifer L Kalina
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - David S Neilson
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada.,Department of Biochemistry & Microbiology, University of Victoria, Victoria, Canada
| | - Yen-Yi Lin
- School of Computing Science, Simon Fraser University, Burnaby, Canada
| | - Phineas T Hamilton
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Alexandra P Comber
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Emma M H Loy
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada.,Department of Biochemistry & Microbiology, University of Victoria, Victoria, Canada
| | - S Cenk Sahinalp
- School of Computing Science, Simon Fraser University, Burnaby, Canada.,Vancouver Prostate Centre, Vancouver, Canada.,School of Informatics & Computing, Indiana University, Bloomington, Indiana
| | | | - Faraz Hach
- School of Computing Science, Simon Fraser University, Burnaby, Canada. .,Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Julian J Lum
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada. .,Department of Biochemistry & Microbiology, University of Victoria, Victoria, Canada
| |
Collapse
|
156
|
Miller A, Asmann Y, Cattaneo L, Braggio E, Keats J, Auclair D, Lonial S, Russell SJ, Stewart AK. High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma. Blood Cancer J 2017; 7:e612. [PMID: 28937974 PMCID: PMC5709757 DOI: 10.1038/bcj.2017.94] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 06/29/2017] [Indexed: 12/11/2022] Open
Abstract
Tumor-specific mutations can result in immunogenic neoantigens, both of which have been correlated with responsiveness to immune checkpoint inhibitors in highly mutagenic cancers. However, early results of single-agent checkpoint inhibitors in multiple myeloma (MM) have been underwhelming. Therefore, we sought to understand the relationship between mutation and neoantigen landscape of MM patients and responsiveness to therapies. Somatic mutation burden, neoantigen load, and response to therapy were determined using interim data from the MMRF CoMMpass study (NCT01454297) on 664 MM patients. In this population, the mean somatic and missense mutation loads were 405.84(s=608.55) and 63.90(s=95.88) mutations per patient, respectively. There was a positive linear relationship between mutation and neoantigen burdens (R2=0.862). The average predicted neoantigen load was 23.52(s=52.14) neoantigens with an average of 9.40(s=26.97) expressed neoantigens. Survival analysis revealed significantly shorter progression-free survival (PFS) in patients with greater than average somatic missense mutation load (N=163, 0.493 vs 0.726 2-year PFS, P=0.0023) and predicted expressed neoantigen load (N=214, 0.555 vs 0.729 2-year PFS, P=0.0028). This pattern is maintained when stratified by disease stage and cytogenetic abnormalities. Therefore, high mutation and neoantigen load are clinically relevant risk factors that negatively impact survival of MM patients under current standards of care.
Collapse
Affiliation(s)
- A Miller
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Y Asmann
- Division of Biomedical Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - L Cattaneo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - E Braggio
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - J Keats
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - D Auclair
- Multiple Myeloma Research Foundation, Norwalk CT, USA
| | - S Lonial
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | | | - S J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Hematology, Mayo Clinic Rochester, MN, USA
| | - A K Stewart
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
157
|
Johanns TM, Bowman-Kirigin JA, Liu C, Dunn GP. Targeting Neoantigens in Glioblastoma: An Overview of Cancer Immunogenomics and Translational Implications. Neurosurgery 2017; 64:165-176. [PMID: 28899059 PMCID: PMC6287409 DOI: 10.1093/neuros/nyx321] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/27/2017] [Indexed: 12/25/2022] Open
Affiliation(s)
- Tanner M. Johanns
- Division of Oncology, Department of Medicine, Washington University School of
Medicine, St. Louis, Missouri
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington
Univer-sity School of Medicine, St. Louis, Missouri
| | - Jay A. Bowman-Kirigin
- Center for Human Immunology and Immunotherapy Prog-rams, Washington University
School of Medicine, St. Louis, Missouri
- Depart-ment of Neurological Surgery, Washing-ton University School of Medicine,
St. Louis, Missouri
| | - Connor Liu
- Center for Human Immunology and Immunotherapy Prog-rams, Washington University
School of Medicine, St. Louis, Missouri
- Depart-ment of Neurological Surgery, Washing-ton University School of Medicine,
St. Louis, Missouri
| | - Gavin P. Dunn
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington
Univer-sity School of Medicine, St. Louis, Missouri
- Depart-ment of Neurological Surgery, Washing-ton University School of Medicine,
St. Louis, Missouri
- Department of Pathology and Immunology, Washington University School of
Medicine, St. Louis, Missouri
| |
Collapse
|
158
|
Bjerregaard AM, Nielsen M, Hadrup SR, Szallasi Z, Eklund AC. MuPeXI: prediction of neo-epitopes from tumor sequencing data. Cancer Immunol Immunother 2017; 66:1123-1130. [PMID: 28429069 PMCID: PMC11028452 DOI: 10.1007/s00262-017-2001-3] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/12/2017] [Indexed: 01/09/2023]
Abstract
Personalization of immunotherapies such as cancer vaccines and adoptive T cell therapy depends on identification of patient-specific neo-epitopes that can be specifically targeted. MuPeXI, the mutant peptide extractor and informer, is a program to identify tumor-specific peptides and assess their potential to be neo-epitopes. The program input is a file with somatic mutation calls, a list of HLA types, and optionally a gene expression profile. The output is a table with all tumor-specific peptides derived from nucleotide substitutions, insertions, and deletions, along with comprehensive annotation, including HLA binding and similarity to normal peptides. The peptides are sorted according to a priority score which is intended to roughly predict immunogenicity. We applied MuPeXI to three tumors for which predicted MHC-binding peptides had been screened for T cell reactivity, and found that MuPeXI was able to prioritize immunogenic peptides with an area under the curve of 0.63. Compared to other available tools, MuPeXI provides more information and is easier to use. MuPeXI is available as stand-alone software and as a web server at http://www.cbs.dtu.dk/services/MuPeXI .
Collapse
Affiliation(s)
- Anne-Mette Bjerregaard
- Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, 2800, Lyngby, Denmark.
| | - Morten Nielsen
- Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, 2800, Lyngby, Denmark
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Sine Reker Hadrup
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Zoltan Szallasi
- Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, 2800, Lyngby, Denmark
- Computational Health Informatics Program, Boston Children's Hospital, USA, Harvard Medical School, Boston, USA
| | - Aron Charles Eklund
- Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, 2800, Lyngby, Denmark.
| |
Collapse
|
159
|
Chang TC, Carter RA, Li Y, Li Y, Wang H, Edmonson MN, Chen X, Arnold P, Geiger TL, Wu G, Peng J, Dyer M, Downing JR, Green DR, Thomas PG, Zhang J. The neoepitope landscape in pediatric cancers. Genome Med 2017; 9:78. [PMID: 28854978 PMCID: PMC5577668 DOI: 10.1186/s13073-017-0468-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/10/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Neoepitopes derived from tumor-specific somatic mutations are promising targets for immunotherapy in childhood cancers. However, the potential for such therapies in targeting these epitopes remains uncertain due to a lack of knowledge of the neoepitope landscape in childhood cancer. Studies to date have focused primarily on missense mutations without exploring gene fusions, which are a major class of oncogenic drivers in pediatric cancer. METHODS We developed an analytical workflow for identification of putative neoepitopes based on somatic missense mutations and gene fusions using whole-genome sequencing data. Transcriptome sequencing data were incorporated to interrogate the expression status of the neoepitopes. RESULTS We present the neoepitope landscape of somatic alterations including missense mutations and oncogenic gene fusions identified in 540 childhood cancer genomes and transcriptomes representing 23 cancer subtypes. We found that 88% of leukemias, 78% of central nervous system tumors, and 90% of solid tumors had at least one predicted neoepitope. Mutation hotspots in KRAS and histone H3 genes encode potential epitopes in multiple patients. Additionally, the ETV6-RUNX1 fusion was found to encode putative neoepitopes in a high proportion (69.6%) of the pediatric leukemia harboring this fusion. CONCLUSIONS Our study presents a comprehensive repertoire of potential neoepitopes in childhood cancers, and will facilitate the development of immunotherapeutic approaches designed to exploit them. The source code of the workflow is available at GitHub ( https://github.com/zhanglabstjude/neoepitope ).
Collapse
Affiliation(s)
- Ti-Cheng Chang
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Robert A Carter
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Yongjin Li
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Yuxin Li
- Department of Structural Biology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA.,St Jude Proteomics Facility, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Hong Wang
- Department of Structural Biology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Michael N Edmonson
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Xiang Chen
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Paula Arnold
- Department of Pathology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Terrence L Geiger
- Department of Pathology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Gang Wu
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Junmin Peng
- Department of Structural Biology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA.,St Jude Proteomics Facility, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Michael Dyer
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - James R Downing
- Department of Pathology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Paul G Thomas
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA
| | - Jinghui Zhang
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA.
| |
Collapse
|
160
|
van Attekum MH, Eldering E, Kater AP. Chronic lymphocytic leukemia cells are active participants in microenvironmental cross-talk. Haematologica 2017; 102:1469-1476. [PMID: 28775118 PMCID: PMC5685246 DOI: 10.3324/haematol.2016.142679] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
The importance of the tumor microenvironment in chronic lymphocytic leukemia is
widely accepted. Nevertheless, the understanding of the complex interplay
between the various types of bystander cells and chronic lymphocytic leukemia
cells is incomplete. Numerous studies have indicated that bystander cells
provide chronic lymphocytic leukemia-supportive functions, but it has also
become clear that chronic lymphocytic leukemia cells actively engage in the
formation of a supportive tumor microenvironment through several cross-talk
mechanisms. In this review, we describe how chronic lymphocytic leukemia cells
participate in this interplay by inducing migration and tumor-supportive
differentiation of bystander cells. Furthermore, chronic lymphocytic
leukemia-mediated alterations in the interactions between bystander cells are
discussed. Upon bystander cell interaction, chronic lymphocytic leukemia cells
secrete cytokines and chemokines such as migratory factors [chemokine
(C-C motif) ligand 22 and chemokine (CC motif) ligand 2], which result
in further recruitment of T cells but also of monocyte-derived cells. Within the
tumor microenvironment, chronic lymphocytic leukemia cells induce
differentiation towards a tumor-supportive M2 phenotype of monocyte-derived
cells and suppress phagocytosis, but also induce increased numbers of supportive
regulatory T cells. Like other tumor types, the differentiation of stromal cells
towards supportive cancer-associated fibroblasts is critically dependent on
chronic lymphocytic leukemia-derived factors such as exosomes and
platelet-derived growth factor. Lastly, both chronic lymphocytic leukemia and
bystander cells induce a tolerogenic tumor microenvironment; chronic lymphocytic
leukemia-secreted cytokines, such as interleukin-10, suppress cytotoxic T-cell
functions, while chronic lymphocytic leukemia-associated monocyte-derived cells
contribute to suppression of T-cell function by producing the immune checkpoint
factor, programmed cell death-ligand 1. Deeper understanding of the active
involvement and cross-talk of chronic lymphocytic leukemia cells in shaping the
tumor microenvironment may offer novel clues for designing therapeutic
strategies.
Collapse
Affiliation(s)
- Martijn Ha van Attekum
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, the Netherlands.,Department of Hematology, Academic Medical Center, University of Amsterdam, the Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, the Netherlands.,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Academic Medical Center, University of Amsterdam, the Netherlands
| | - Arnon P Kater
- Department of Hematology, Academic Medical Center, University of Amsterdam, the Netherlands .,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Academic Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|
161
|
Zhou Z, Wu S, Lai J, Shi Y, Qiu C, Chen Z, Wang Y, Gu X, Zhou J, Chen S. Identification of trunk mutations in gastric carcinoma: a case study. BMC Med Genomics 2017; 10:49. [PMID: 28716121 PMCID: PMC5520061 DOI: 10.1186/s12920-017-0285-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/13/2017] [Indexed: 02/06/2023] Open
Abstract
Background Intratumor heterogeneity (ITH) poses an urgent challenge for cancer precision medicine because it can cause drug resistance against cancer target therapy and immunotherapy. The search for trunk mutations that are present in all cancer cells is therefore critical for each patient. Case presentation In this study, we aimed to evaluate the efficiency of multiregional sequencing for the identification of trunk mutations present in all regions of a tumor as a case study. We applied multiregional whole-exome sequencing (WES) to investigate the genetic heterogeneity and homogeneity of a case of gastric carcinoma. Approximately 83% of common missense mutations present in two samples and approximately 89% of common missense mutations present in three samples were trunk mutations. Notably, trunk mutations appeared to have higher variant allele frequencies (VAFs) than non-trunk mutations. Conclusions Our results indicate that small-scale multiregional sampling and subsequent screening of low VAF somatic mutations might be a cost-effective strategy for identifying the majority of trunk mutations in gastric carcinoma. Electronic supplementary material The online version of this article (doi:10.1186/s12920-017-0285-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhan Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China
| | - Shanshan Wu
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China
| | - Jun Lai
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China
| | - Yuan Shi
- Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang, Hangzhou, 310058, China
| | - Chixiao Qiu
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China
| | - Zhe Chen
- Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang, Hangzhou, 310058, China
| | - Yufeng Wang
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Xun Gu
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Jie Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China.
| | - Shuqing Chen
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China. .,International Center for Precision Medicine, Zhejiang California International NanoSystems Institute, Zhejiang, Hangzhou, 310058, China.
| |
Collapse
|
162
|
Mouw KW, Goldberg MS, Konstantinopoulos PA, D'Andrea AD. DNA Damage and Repair Biomarkers of Immunotherapy Response. Cancer Discov 2017; 7:675-693. [PMID: 28630051 PMCID: PMC5659200 DOI: 10.1158/2159-8290.cd-17-0226] [Citation(s) in RCA: 468] [Impact Index Per Article: 66.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/05/2017] [Accepted: 05/18/2017] [Indexed: 12/16/2022]
Abstract
DNA-damaging agents are widely used in clinical oncology and exploit deficiencies in tumor DNA repair. Given the expanding role of immune checkpoint blockade as a therapeutic strategy, the interaction of tumor DNA damage with the immune system has recently come into focus, and it is now clear that the tumor DNA repair landscape has an important role in driving response to immune checkpoint blockade. Here, we summarize the mechanisms by which DNA damage and genomic instability have been found to shape the antitumor immune response and describe clinical efforts to use DNA repair biomarkers to guide use of immune-directed therapies.Significance: Only a subset of patients respond to immune checkpoint blockade, and reliable predictive biomarkers of response are needed to guide therapy decisions. DNA repair deficiency is common among tumors, and emerging experimental and clinical evidence suggests that features of genomic instability are associated with response to immune-directed therapies. Cancer Discov; 7(7); 675-93. ©2017 AACR.
Collapse
Affiliation(s)
- Kent W Mouw
- Department of Radiation Oncology, Brigham & Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Michael S Goldberg
- Harvard Medical School, Boston, Massachusetts
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Panagiotis A Konstantinopoulos
- Harvard Medical School, Boston, Massachusetts
- Medical Gynecology Oncology Program, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alan D D'Andrea
- Department of Radiation Oncology, Brigham & Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
- Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
163
|
Abstract
Historically, immune-based therapies have played a leading role in the treatment of hematologic malignancies, with the efficacy of stem cell transplantation largely attributable to donor immunity against malignant cells. As new and more targeted immunotherapies have developed, their role in the treatment of hematologic malignancies is evolving and expanding. Herein, we discuss approaches for antigen discovery and review known and novel tumor antigens in hematologic malignancies. We further explore the role of established and investigational immunotherapies in hematologic malignancies, with a focus on personalization of treatment modalities such as cancer vaccines and adoptive cell therapy. Finally, we identify areas of active investigation and development. Immunotherapy is at an exciting crossroads for the treatment of hematologic malignancies, with further investigation aimed at producing effective, targeted immune therapies that maximize antitumor effects while minimizing toxicity.
Collapse
Affiliation(s)
- David A. Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
164
|
Capietto AH, Jhunjhunwala S, Delamarre L. Characterizing neoantigens for personalized cancer immunotherapy. Curr Opin Immunol 2017; 46:58-65. [PMID: 28478383 DOI: 10.1016/j.coi.2017.04.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/17/2017] [Indexed: 12/18/2022]
Abstract
Somatic mutations can generate neoantigens that are presented on MHC molecules and drive effective T cells responses against cancer. Mutation load in cancer patients predicts response to immune checkpoint blockade therapy. Additionally, vaccination targeting neoantigens controls established tumor growth in preclinical models. These recent findings led to a renewed interest in the field of cancer vaccines and the development of antigen-targeted cancer immunotherapies. However, targeting neoantigens is challenging, as most mutations are unique to each cancer patient. In addition, only a small fraction of the mutations are immunogenic and therefore their accurate prediction is critical. In this review, we discuss the properties of neoantigens that influence their immunogenicity, along with questions that remain to be addressed in order to improve prediction algorithms.
Collapse
|
165
|
Pfeifer CR, Alvey CM, Irianto J, Discher DE. Genome variation across cancers scales with tissue stiffness - an invasion-mutation mechanism and implications for immune cell infiltration. ACTA ACUST UNITED AC 2017; 2:103-114. [PMID: 29082336 DOI: 10.1016/j.coisb.2017.04.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Many different types of soft and solid tumors have now been sequenced, and meta-analyses suggest that genomic variation across tumors scales with the stiffness of the tumors' tissues of origin. The opinion expressed here is based on a review of current genomics data, and it considers multiple 'mechanogenomics' mechanisms to potentially explain this scaling of mutation rate with tissue stiffness. Since stiff solid tissues have higher density of fibrous collagen matrix, which should decrease tissue porosity, cancer cell proliferation could be affected and so could invasion into stiff tissues as the nucleus is squeezed sufficiently to enhance DNA damage. Diversification of a cancer genome after constricted migration is now clear. Understanding genome changes that give rise to neo-antigens is important to selection as well as to the development of immunotherapies, and we discuss engineered monocytes/macrophages as particularly relevant to understanding infiltration into solid tumors.
Collapse
Affiliation(s)
- Charlotte R Pfeifer
- Physical Sciences Oncology Center at Penn (PSOC@Penn), University of Pennsylvania, Philadelphia, PA 19104.,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104.,Graduate Group / Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA 19104
| | - Cory M Alvey
- Physical Sciences Oncology Center at Penn (PSOC@Penn), University of Pennsylvania, Philadelphia, PA 19104.,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104.,Graduate Group / Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104
| | - Jerome Irianto
- Physical Sciences Oncology Center at Penn (PSOC@Penn), University of Pennsylvania, Philadelphia, PA 19104.,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Dennis E Discher
- Physical Sciences Oncology Center at Penn (PSOC@Penn), University of Pennsylvania, Philadelphia, PA 19104.,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104.,Graduate Group / Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA 19104.,Graduate Group / Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
166
|
Purroy N, Wu CJ. Coevolution of Leukemia and Host Immune Cells in Chronic Lymphocytic Leukemia. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026740. [PMID: 28096240 DOI: 10.1101/cshperspect.a026740] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cumulative studies on the dissection of changes in driver genetic lesions in cancer across the course of the disease have provided powerful insights into the adaptive mechanisms of tumors in response to the selective pressures of therapy and environmental changes. In particular, the advent of next-generation-sequencing (NGS)-based technologies and its implementation for the large-scale comprehensive analyses of cancers have greatly advanced our understanding of cancer as a complex dynamic system wherein genetically distinct subclones interact and compete during tumor evolution. Aside from genetic evolution arising from interactions intrinsic to the cell subpopulations within tumors, it is increasingly appreciated that reciprocal interactions between the tumor cell and cellular constituents of the microenvironment further exert selective pressures on specific clones that can impact the balance between tumor immunity and immunologic evasion and escape. Herein, we review the evidence supporting these concepts, with a particular focus on chronic lymphocytic leukemia (CLL), a disease that has been highly amenable to genomic interrogation and studies of clonal heterogeneity and evolution. Better knowledge of the basis for immune escape has an important clinical impact on prognostic stratification and on the pursuit of new therapeutic opportunities.
Collapse
Affiliation(s)
- Noelia Purroy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142.,Harvard Medical School, Boston, Massachusetts 02115
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142.,Harvard Medical School, Boston, Massachusetts 02115.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115
| |
Collapse
|
167
|
Zhou Z, Lyu X, Wu J, Yang X, Wu S, Zhou J, Gu X, Su Z, Chen S. TSNAD: an integrated software for cancer somatic mutation and tumour-specific neoantigen detection. ROYAL SOCIETY OPEN SCIENCE 2017; 4:170050. [PMID: 28484631 PMCID: PMC5414268 DOI: 10.1098/rsos.170050] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/06/2017] [Indexed: 06/02/2023]
Abstract
Tumour antigens have attracted much attention because of their importance to cancer diagnosis, prognosis and targeted therapy. With the development of cancer genomics, the identification of tumour-specific neoantigens became possible, which is a crucial step for cancer immunotherapy. In this study, we developed software called the tumour-specific neoantigen detector for detecting cancer somatic mutations following the best practices of the genome analysis toolkit and predicting potential tumour-specific neoantigens, which could be either extracellular mutations of membrane proteins or mutated peptides presented by class I major histocompatibility complex molecules. This pipeline was beneficial to the biologist with little programmatic background. We also applied the software to the somatic mutations from the International Cancer Genome Consortium database to predict numerous potential tumour-specific neoantigens. This software is freely available from https://github.com/jiujiezz/tsnad.
Collapse
Affiliation(s)
- Zhan Zhou
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xingzheng Lyu
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310013, People's Republic of China
| | - Jingcheng Wu
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xiaoyue Yang
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Shanshan Wu
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Jie Zhou
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xun Gu
- Department of Genetics, Development and Cell Biology, Program of Bioinformatics and Computational Biology, Iowa State University, Ames, IA 50010, USA
| | - Zhixi Su
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, People's Republic of China
| | - Shuqing Chen
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| |
Collapse
|
168
|
Spira A, Yurgelun MB, Alexandrov L, Rao A, Bejar R, Polyak K, Giannakis M, Shilatifard A, Finn OJ, Dhodapkar M, Kay NE, Braggio E, Vilar E, Mazzilli SA, Rebbeck TR, Garber JE, Velculescu VE, Disis ML, Wallace DC, Lippman SM. Precancer Atlas to Drive Precision Prevention Trials. Cancer Res 2017; 77:1510-1541. [PMID: 28373404 PMCID: PMC6681830 DOI: 10.1158/0008-5472.can-16-2346] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 02/07/2023]
Abstract
Cancer development is a complex process driven by inherited and acquired molecular and cellular alterations. Prevention is the holy grail of cancer elimination, but making this a reality will take a fundamental rethinking and deep understanding of premalignant biology. In this Perspective, we propose a national concerted effort to create a Precancer Atlas (PCA), integrating multi-omics and immunity - basic tenets of the neoplastic process. The biology of neoplasia caused by germline mutations has led to paradigm-changing precision prevention efforts, including: tumor testing for mismatch repair (MMR) deficiency in Lynch syndrome establishing a new paradigm, combinatorial chemoprevention efficacy in familial adenomatous polyposis (FAP), signal of benefit from imaging-based early detection research in high-germline risk for pancreatic neoplasia, elucidating early ontogeny in BRCA1-mutation carriers leading to an international breast cancer prevention trial, and insights into the intricate germline-somatic-immunity interaction landscape. Emerging genetic and pharmacologic (metformin) disruption of mitochondrial (mt) respiration increased autophagy to prevent cancer in a Li-Fraumeni mouse model (biology reproduced in clinical pilot) and revealed profound influences of subtle changes in mt DNA background variation on obesity, aging, and cancer risk. The elaborate communication between the immune system and neoplasia includes an increasingly complex cellular microenvironment and dynamic interactions between host genetics, environmental factors, and microbes in shaping the immune response. Cancer vaccines are in early murine and clinical precancer studies, building on the recent successes of immunotherapy and HPV vaccine immune prevention. Molecular monitoring in Barrett's esophagus to avoid overdiagnosis/treatment highlights an important PCA theme. Next generation sequencing (NGS) discovered age-related clonal hematopoiesis of indeterminate potential (CHIP). Ultra-deep NGS reports over the past year have redefined the premalignant landscape remarkably identifying tiny clones in the blood of up to 95% of women in their 50s, suggesting that potentially premalignant clones are ubiquitous. Similar data from eyelid skin and peritoneal and uterine lavage fluid provide unprecedented opportunities to dissect the earliest phases of stem/progenitor clonal (and microenvironment) evolution/diversity with new single-cell and liquid biopsy technologies. Cancer mutational signatures reflect exogenous or endogenous processes imprinted over time in precursors. Accelerating the prevention of cancer will require a large-scale, longitudinal effort, leveraging diverse disciplines (from genetics, biochemistry, and immunology to mathematics, computational biology, and engineering), initiatives, technologies, and models in developing an integrated multi-omics and immunity PCA - an immense national resource to interrogate, target, and intercept events that drive oncogenesis. Cancer Res; 77(7); 1510-41. ©2017 AACR.
Collapse
Affiliation(s)
- Avrum Spira
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Bioinformatics, Boston University School of Medicine, Boston, Massachusetts
| | - Matthew B Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ludmil Alexandrov
- Theoretical Division, Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, New Mexico
| | - Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Rafael Bejar
- Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ali Shilatifard
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Madhav Dhodapkar
- Department of Hematology and Immunology, Yale Cancer Center, New Haven, Connecticut
| | - Neil E Kay
- Department of Hematology, Mayo Clinic Hospital, Rochester, Minnesota
| | - Esteban Braggio
- Department of Hematology, Mayo Clinic Hospital, Phoenix, Arizona
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarah A Mazzilli
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Bioinformatics, Boston University School of Medicine, Boston, Massachusetts
| | - Timothy R Rebbeck
- Division of Hematology and Oncology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Victor E Velculescu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
- Department of Pathology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Mary L Disis
- Department of Medicine, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott M Lippman
- Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, California.
| |
Collapse
|
169
|
Kuai R, Ochyl LJ, Bahjat KS, Schwendeman A, Moon JJ. Designer vaccine nanodiscs for personalized cancer immunotherapy. NATURE MATERIALS 2017; 16:489-496. [PMID: 28024156 PMCID: PMC5374005 DOI: 10.1038/nmat4822] [Citation(s) in RCA: 717] [Impact Index Per Article: 102.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/08/2016] [Indexed: 05/04/2023]
Abstract
Despite the tremendous potential of peptide-based cancer vaccines, their efficacy has been limited in humans. Recent innovations in tumour exome sequencing have signalled the new era of personalized immunotherapy with patient-specific neoantigens, but a general methodology for stimulating strong CD8α+ cytotoxic T-lymphocyte (CTL) responses remains lacking. Here we demonstrate that high-density lipoprotein-mimicking nanodiscs coupled with antigen (Ag) peptides and adjuvants can markedly improve Ag/adjuvant co-delivery to lymphoid organs and sustain Ag presentation on dendritic cells. Strikingly, nanodiscs elicited up to 47-fold greater frequencies of neoantigen-specific CTLs than soluble vaccines and even 31-fold greater than perhaps the strongest adjuvant in clinical trials (that is, CpG in Montanide). Moreover, multi-epitope vaccination generated broad-spectrum T-cell responses that potently inhibited tumour growth. Nanodiscs eliminated established MC-38 and B16F10 tumours when combined with anti-PD-1 and anti-CTLA-4 therapy. These findings represent a new powerful approach for cancer immunotherapy and suggest a general strategy for personalized nanomedicine.
Collapse
Affiliation(s)
- Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lukasz J. Ochyl
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Keith S. Bahjat
- Discovery Research, Bristol-Myers Squibb Biologics Discovery California, Redwood City, CA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Co-corresponding authors. J.J.M. () or A.S. ()
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Co-corresponding authors. J.J.M. () or A.S. ()
| |
Collapse
|
170
|
Khodadoust M, Olsson N, Wagar L, Haabeth O, Chen B, Swaminathan K, Rawson K, Liu C, Steiner D, Lund P, Rao S, Zhang L, Marceau C, Stehr H, Newman A, Czerwinski DK, Carlton V, Moorhead M, Faham M, Kohrt H, Carette J, Green M, Davis M, Levy R, Elias JE, Alizadeh A. Antigen presentation profiling reveals recognition of lymphoma immunoglobulin neoantigens. Nature 2017; 543:723-727. [PMID: 28329770 PMCID: PMC5808925 DOI: 10.1038/nature21433] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 01/30/2017] [Indexed: 02/07/2023]
Abstract
Cancer somatic mutations can generate neoantigens that distinguish malignant from normal cells. However, the personalized identification and validation of neoantigens remains a major challenge. Here we discover neoantigens in human mantle-cell lymphomas by using an integrated genomic and proteomic strategy that interrogates tumour antigen peptides presented by major histocompatibility complex (MHC) class I and class II molecules. We applied this approach to systematically characterize MHC ligands from 17 patients. Remarkably, all discovered neoantigenic peptides were exclusively derived from the lymphoma immunoglobulin heavy- or light-chain variable regions. Although we identified MHC presentation of private polymorphic germline alleles, no mutated peptides were recovered from non-immunoglobulin somatically mutated genes. Somatic mutations within the immunoglobulin variable region were almost exclusively presented by MHC class II. We isolated circulating CD4+ T cells specific for immunoglobulin-derived neoantigens and found these cells could mediate killing of autologous lymphoma cells. These results demonstrate that an integrative approach combining MHC isolation, peptide identification, and exome sequencing is an effective platform to uncover tumour neoantigens. Application of this strategy to human lymphoma implicates immunoglobulin neoantigens as targets for lymphoma immunotherapy.
Collapse
Affiliation(s)
- M.S. Khodadoust
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
| | - N. Olsson
- Department of Chemical & Systems Biology, Stanford University, Stanford, California, USA
| | - L.E. Wagar
- Department of Microbiology & Immunology, Stanford University, Stanford, California, USA
| | - O.A.W. Haabeth
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
| | - B. Chen
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
| | - K. Swaminathan
- Department of Chemical & Systems Biology, Stanford University, Stanford, California, USA
| | - K. Rawson
- Department of Chemical & Systems Biology, Stanford University, Stanford, California, USA
| | - C.L. Liu
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
| | - D. Steiner
- Department of Pathology, Stanford University, Stanford, California, USA
| | - P. Lund
- Department of Microbiology & Immunology, Stanford University, Stanford, California, USA
| | - S. Rao
- Department of Chemical & Systems Biology, Stanford University, Stanford, California, USA
| | - L. Zhang
- Department of Chemical & Systems Biology, Stanford University, Stanford, California, USA
| | - C. Marceau
- Department of Microbiology & Immunology, Stanford University, Stanford, California, USA
| | - H. Stehr
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
| | - A.M. Newman
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
- Center for Cancer Systems Biology, Stanford University, Stanford, California, USA
| | - D. K. Czerwinski
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
| | | | - M. Moorhead
- Adaptive Biotechnologies, Seattle, Washington, USA
| | - M. Faham
- Adaptive Biotechnologies, Seattle, Washington, USA
| | - H.E. Kohrt
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
| | - J. Carette
- Department of Microbiology & Immunology, Stanford University, Stanford, California, USA
| | - M.R. Green
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
| | - M.M. Davis
- Department of Microbiology & Immunology, Stanford University, Stanford, California, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, California, USA
| | - R. Levy
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
| | - J. E. Elias
- Department of Chemical & Systems Biology, Stanford University, Stanford, California, USA
| | - A.A. Alizadeh
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, California, USA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
171
|
Luen S, Virassamy B, Savas P, Salgado R, Loi S. The genomic landscape of breast cancer and its interaction with host immunity. Breast 2017; 29:241-50. [PMID: 27481651 DOI: 10.1016/j.breast.2016.07.015] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 07/08/2016] [Indexed: 12/15/2022] Open
Abstract
Molecular profiling of thousands of primary breast cancers has uncovered remarkable genomic diversity between breast cancer subtypes, and even within subtypes. Only a few driver genes are recurrently altered at high frequency highlighting great challenges for precision medicine. Considerable evidence also confirms the role of host immunosurveillance in influencing response to therapy and prognosis in HER2+ and triple negative breast cancer. The role of immunosurveillance in ER + disease remains unclear. Advances in both these fields have lead to intensified interest in the interaction between genomic landscapes and host anti-tumour immune responses in breast cancer. In this review, we discuss the potential genomic determinants of host anti-tumour immunity - mutational load, driver alterations, mutational processes and neoantigens - and their relationship with immunity in breast cancer. Significant differences exist in both the genomic and immune characteristics amongst breast cancer subtypes. While ER + disease appears to be less immunogenic than HER2+ and triple negative breast cancer, it displays the greatest degree of heterogeneity. Mutational and neoantigen load appears to incompletely explains immune responses in breast cancer. Driver alterations do not appear to increase immunogenicity. Instead, they could contribute to immune-evasion or an immunosuppressive microenvironment, and therefore represent potential therapeutic targets. Finally, we also discuss the tailoring of immunotherapeutic strategies by genomic alterations, with possible multimodal combination approaches to maximise clinical benefits.
Collapse
Affiliation(s)
- Stephen Luen
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Balaji Virassamy
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter Savas
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Sherene Loi
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
172
|
Feng S, Han M, Zhou L, Wang Q, Li Z, Li Y, Lu H, Liu T, Ma Y, Liu S, Cheng J. NS5ABP37 inhibits liver cancer by impeding lipogenesis and cholesterogenesis. Cancer Sci 2017; 108:12-22. [PMID: 27862769 PMCID: PMC5276832 DOI: 10.1111/cas.13117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/27/2016] [Accepted: 10/30/2016] [Indexed: 12/12/2022] Open
Abstract
The molecular mechanism underlying non‐alcoholic fatty liver disease progression to hepatocellular carcinoma (HCC) remains unknown. In this study, immunohistochemistry staining results showed that NS5ABP37 protein, which is in a state of lower expression in tumor tissues, decreased with increasing degree of HCC malignancy. Two cell models, HepG2 and L02, were used to analyze the mechanism between NS5ABP37 and HCC. In agreement, NS5ABP37 protein overexpression significantly suppressed cell proliferation, caused G1/S cell cycle arrest, and induced apoptosis by increasing caspase‐3/7 activity and cleaved caspase‐3 levels. In addition, NS5ABP37 overexpression resulted in decreased intracellular triglyceride and total cholesterol contents, with level reduction in sterol regulatory element‐binding proteins (SREBPs) and downstream effectors. Furthermore, NS5ABP37 overexpression decreased SREBP1c and SREBP2 levels by reducing their respective promoters. Finally, reactive oxygen species levels and endoplasmic reticulum stress were both induced by NS5ABP37 overexpression. These findings together indicate that NS5ABP37 inhibits cancer cell proliferation and promotes apoptosis, by altering SREBP‐dependent lipogenesis and cholesterogenesis in HepG2 and L02 cells and inducing oxidative stress and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Shenghu Feng
- Beijing Ditan Hospital, Peking University Teaching Hospital, Beijing, China
| | - Ming Han
- Beijing Ditan Hospital, Peking University Teaching Hospital, Beijing, China
| | - Li Zhou
- Beijing Ditan Hospital, Peking University Teaching Hospital, Beijing, China
| | - Qi Wang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhongshu Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yaru Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hongping Lu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ting Liu
- Beijing Ditan Hospital, Peking University Teaching Hospital, Beijing, China
| | - Yanhua Ma
- Beijing Ditan Hospital, Peking University Teaching Hospital, Beijing, China
| | - Shunai Liu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jun Cheng
- Beijing Ditan Hospital, Peking University Teaching Hospital, Beijing, China.,Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
173
|
Heong V, Ngoi N, Tan DSP. Update on immune checkpoint inhibitors in gynecological cancers. J Gynecol Oncol 2017; 28:e20. [PMID: 28028993 PMCID: PMC5323287 DOI: 10.3802/jgo.2017.28.e20] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/04/2016] [Accepted: 11/28/2016] [Indexed: 12/21/2022] Open
Abstract
In recent years, progress in our understanding of immune-modulatory signaling pathways in immune cells and the tumor microenvironment (TME) has led to rejuvenated interest in cancer immunotherapy. In particular, immunotherapy targeting the immune checkpoint receptors such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell-death 1 (PD-1), and programmed cell-death ligand 1 (PD-L1) have demonstrated clinical activity in a wide variety of tumors, including gynecological cancers. This review will focus on the emerging clinical data on the therapeutic role of immune checkpoint inhibitors, and potential strategies to enhance the efficacy of this class of compounds, in the context of gynecological cancers. It is anticipated that future biomarker-directed clinical trials will provide further insights into the mechanisms underlying response and resistance to immunotherapy, and help guide our approach to designing therapeutic combinations that have the potential to enhance the benefit of immunotherapy in patients with gynecologic cancers.
Collapse
Affiliation(s)
- Valerie Heong
- Department of Hematology-Oncology, National University Hospital, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Natalie Ngoi
- Department of Hematology-Oncology, National University Hospital, Singapore, Singapore
| | - David Shao Peng Tan
- Department of Hematology-Oncology, National University Hospital, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
174
|
Mennonna D, Maccalli C, Romano MC, Garavaglia C, Capocefalo F, Bordoni R, Severgnini M, De Bellis G, Sidney J, Sette A, Gori A, Longhi R, Braga M, Ghirardelli L, Baldari L, Orsenigo E, Albarello L, Zino E, Fleischhauer K, Mazzola G, Ferrero N, Amoroso A, Casorati G, Parmiani G, Dellabona P. T cell neoepitope discovery in colorectal cancer by high throughput profiling of somatic mutations in expressed genes. Gut 2017; 66:454-463. [PMID: 26681737 PMCID: PMC5534766 DOI: 10.1136/gutjnl-2015-309453] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 11/04/2015] [Accepted: 11/06/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Patient-specific (unique) tumour antigens, encoded by somatically mutated cancer genes, generate neoepitopes that are implicated in the induction of tumour-controlling T cell responses. Recent advancements in massive DNA sequencing combined with robust T cell epitope predictions have allowed their systematic identification in several malignancies. DESIGN We undertook the identification of unique neoepitopes in colorectal cancers (CRCs) by using high-throughput sequencing of cDNAs expressed by standard cancer cell cultures, and by related cancer stem/initiating cells (CSCs) cultures, coupled with a reverse immunology approach not requiring human leukocyte antigen (HLA) allele-specific epitope predictions. RESULTS Several unique mutated antigens of CRC, shared by standard cancer and related CSC cultures, were identified by this strategy. CD8+ and CD4+ T cells, either autologous to the patient or derived from HLA-matched healthy donors, were readily expanded in vitro by peptides spanning different cancer mutations and specifically recognised differentiated cancer cells and CSC cultures, expressing the mutations. Neoepitope-specific CD8+ T cell frequency was also increased in a patient, compared with healthy donors, supporting the occurrence of clonal expansion in vivo. CONCLUSIONS These results provide a proof-of-concept approach for the identification of unique neoepitopes that are immunogenic in patients with CRC and can also target T cells against the most aggressive CSC component.
Collapse
Affiliation(s)
- Daniele Mennonna
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Maccalli
- Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Michele C Romano
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Claudio Garavaglia
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Filippo Capocefalo
- Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Bordoni
- Institute for Biomedical Technologies, National Research Council, Segrate, Italy
| | - Marco Severgnini
- Institute for Biomedical Technologies, National Research Council, Segrate, Italy
| | - Gianluca De Bellis
- Institute for Biomedical Technologies, National Research Council, Segrate, Italy
| | - John Sidney
- La Jolla Institute for Allergy & Immunology, La Jolla, California, USA
| | - Alessandro Sette
- La Jolla Institute for Allergy & Immunology, La Jolla, California, USA
| | - Alessandro Gori
- Institute of Molecular Recognition Chemistry, National Research Council, Milan, Italy
| | - Renato Longhi
- Institute of Molecular Recognition Chemistry, National Research Council, Milan, Italy
| | - Marco Braga
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Luca Ghirardelli
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Ludovica Baldari
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Elena Orsenigo
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Luca Albarello
- Department of Pathology, San Raffaele Scientific Institute, Milano, Italy
| | - Elisabetta Zino
- Unit of Molecular and Functional Immunogenetics, San Raffaele Scientific Institute, Milan, Italy
| | - Katharina Fleischhauer
- Unit of Molecular and Functional Immunogenetics, San Raffaele Scientific Institute, Milan, Italy,Institute for Experimental Cellular Therapy, University Hospital Essen, Essen, Germany
| | - Gina Mazzola
- Department of Medical Sciences, Center for Transplantation Biology and Immunogenetics, University of Turin, Turin, Italy
| | - Norma Ferrero
- Department of Medical Sciences, Center for Transplantation Biology and Immunogenetics, University of Turin, Turin, Italy
| | - Antonio Amoroso
- Department of Medical Sciences, Center for Transplantation Biology and Immunogenetics, University of Turin, Turin, Italy
| | - Giulia Casorati
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Giorgio Parmiani
- Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Dellabona
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
175
|
Haebe S, Weigert O. It's a long way to the top (if you want to personalize immunotherapy). J Immunother Cancer 2017; 5:6. [PMID: 28116089 PMCID: PMC5240409 DOI: 10.1186/s40425-016-0207-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/28/2016] [Indexed: 01/21/2023] Open
Abstract
Harnessing the immune system to attack tumor cells by targeting tumor-associated or –preferably– tumor-specific antigens has emerged as a promising but challenging treatment option for malignant lymphomas. Follicular lymphoma is among the most common lymphomas worldwide and remains incurable for most patients. Considered to be an immunogenic disease it represents an interesting disease entity for various immunotherapeutic approaches. In an article published in the May issue of Clinical Cancer Research, Nielsen and colleagues provided important proof-of-principle data on the immunogenicity of follicular lymphoma that might represent a first step towards personalized adoptive immunotherapies in this disease. The authors combined targeted next-generation sequencing and in silico analyses to explore the concept of somatic neoepitope prediction. Neoantigen-specific CD8+ T-cells could be identified in a small subset of patients selected for in vitro immunogenicity experiments, however at remarkably low frequencies and in only a few patients at single time-points. Of note, the immunogenic neoepitopes were derived from mutant CREBBP and MEF2B, two genes that have previously been shown to be functionally and prognostically relevant in this disease. In this commentary we discuss the promises but also the challenges of how to translate these findings into clinical practice.
Collapse
Affiliation(s)
- Sarah Haebe
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Max-Lebsche Platz 30, 81377 Munich, Germany
| | - Oliver Weigert
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Max-Lebsche Platz 30, 81377 Munich, Germany ; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
176
|
Yin Q, Sivina M, Robins H, Yusko E, Vignali M, O'Brien S, Keating MJ, Ferrajoli A, Estrov Z, Jain N, Wierda WG, Burger JA. Ibrutinib Therapy Increases T Cell Repertoire Diversity in Patients with Chronic Lymphocytic Leukemia. THE JOURNAL OF IMMUNOLOGY 2017; 198:1740-1747. [PMID: 28077600 DOI: 10.4049/jimmunol.1601190] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 12/13/2016] [Indexed: 11/19/2022]
Abstract
The Bruton's tyrosine kinase inhibitor ibrutinib is a highly effective, new targeted therapy for chronic lymphocytic leukemia (CLL) that thwarts leukemia cell survival, growth, and tissue homing. The effects of ibrutinib treatment on the T cell compartment, which is clonally expanded and thought to support the growth of malignant B cells in CLL, are not fully characterized. Using next-generation sequencing technology, we characterized the diversity of TCRβ-chains in peripheral blood T cells from 15 CLL patients before and after 1 y of ibrutinib therapy. We noted elevated CD4+ and CD8+ T cell numbers and a restricted TCRβ repertoire in all pretreatment samples. After 1 y of ibrutinib therapy, elevated peripheral blood T cell numbers and T cell-related cytokine levels had normalized, and T cell repertoire diversity increased significantly. Dominant TCRβ clones in pretreatment samples declined or became undetectable, and the number of productive unique clones increased significantly during ibrutinib therapy, with the emergence of large numbers of low-frequency TCRβ clones. Importantly, broader TCR repertoire diversity was associated with clinical efficacy and lower rates of infections during ibrutinib therapy. These data demonstrate that ibrutinib therapy increases diversification of the T cell compartment in CLL patients, which contributes to cellular immune reconstitution.
Collapse
Affiliation(s)
- Qingsong Yin
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77230.,Department of Leukemia, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Henan Institute of Hematology, Zhengzhou, Henan 450009, China
| | - Mariela Sivina
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77230
| | - Harlan Robins
- Adaptive Biotechnologies, Seattle, WA 98102; and.,Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Erik Yusko
- Adaptive Biotechnologies, Seattle, WA 98102; and
| | | | - Susan O'Brien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77230
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77230
| | - Alessandra Ferrajoli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77230
| | - Zeev Estrov
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77230
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77230
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77230
| | - Jan A Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77230;
| |
Collapse
|
177
|
Genetically Modified T-Cell-Based Adoptive Immunotherapy in Hematological Malignancies. J Immunol Res 2017; 2017:5210459. [PMID: 28116322 PMCID: PMC5237740 DOI: 10.1155/2017/5210459] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/05/2016] [Indexed: 12/24/2022] Open
Abstract
A significant proportion of hematological malignancies remain limited in treatment options. Immune system modulation serves as a promising therapeutic approach to eliminate malignant cells. Cytotoxic T lymphocytes (CTLs) play a central role in antitumor immunity; unfortunately, nonspecific approaches for targeted recognition of tumor cells by CTLs to mediate tumor immune evasion in hematological malignancies imply multiple mechanisms, which may or may not be clinically relevant. Recently, genetically modified T-cell-based adoptive immunotherapy approaches, including chimeric antigen receptor (CAR) T-cell therapy and engineered T-cell receptor (TCR) T-cell therapy, promise to overcome immune evasion by redirecting the specificity of CTLs to tumor cells. In clinic trials, CAR-T-cell- and TCR-T-cell-based adoptive immunotherapy have produced encouraging clinical outcomes, thereby demonstrating their therapeutic potential in mitigating tumor development. The purpose of the present review is to (1) provide a detailed overview of the multiple mechanisms for immune evasion related with T-cell-based therapies; (2) provide a current summary of the applications of CAR-T-cell- as well as neoantigen-specific TCR-T-cell-based adoptive immunotherapy and routes taken to overcome immune evasion; and (3) evaluate alternative approaches targeting immune evasion via optimization of CAR-T and TCR-T-cell immunotherapies.
Collapse
|
178
|
Paik PK, Hellmann MD. 'Genotype/immunotype' correlations in resected NSCLC. Ann Oncol 2017; 28:7-8. [PMID: 28039156 DOI: 10.1093/annonc/mdw624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- P K Paik
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
- Weill Cornell Medical College, New York, USA
| | - M D Hellmann
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
- Weill Cornell Medical College, New York, USA
| |
Collapse
|
179
|
Predicted neoantigen load in non-hypermutated endometrial cancers: Correlation with outcome and tumor-specific genomic alterations. Gynecol Oncol Rep 2016; 19:42-45. [PMID: 28070553 PMCID: PMC5219603 DOI: 10.1016/j.gore.2016.12.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/12/2016] [Accepted: 12/27/2016] [Indexed: 11/24/2022] Open
Abstract
Elevated neoantigen load has been previously correlated with improved outcome and response to immune checkpoint blockade in various tumor types. In endometrial cancer, previous studies of neoantigen load prediction have shown that the hypermutated MSI and POLE-mutated tumors harbor significantly higher predicted neoantigen load compared to the hypomutated CN-low/endometrioid and CN-high/serous-like tumors. Here, we report that predicted neoantigen load may be a prognostic factor in hypomutated endometrial cancers, both in CN-low/endometrioid and CN-high/serous-like tumors. Specifically, in the TCGA dataset, CN-low/endometrioid tumors with neoantigen load in the highest tertile were associated with significantly improved progression free survival (PFS) (p = 0.031), while CN-high/serous-like tumors with neoantigen load in the lowest tertile were associated with worse PFS (p = 0.041). Importantly, certain tumor-specific genomic alterations were enriched in tumors with lower neoantigen load, including CTNNB1 mutations in CN-low/endometrioid tumors and MYC amplification and PIK3CA mutations in CN-high/serous-like tumors. These findings suggest that predicted neoantigen load and specific genomic alterations (such as CTNNB1 mutations, MYC amplification and PIK3CA mutations) may be biomarkers of response to immunotherapy in hypomutated endometrial cancers, and argues that these exploratory biomarkers should be incorporated in clinical trials of immune checkpoint blockade in this disease. Neoantigen load is associated with outcome in hypomutated endometrial cancers. CTNNB1 mutations are enriched in CN-low/endometrioid tumors with low neoantigen load. MYC amplification and PIK3CA mutations are enriched in CN-high/serous-like tumors with low neoantigen load.
Collapse
|
180
|
Boyiadzis M, Bishop MR, Abonour R, Anderson KC, Ansell SM, Avigan D, Barbarotta L, Barrett AJ, Van Besien K, Bergsagel PL, Borrello I, Brody J, Brufsky J, Cairo M, Chari A, Cohen A, Cortes J, Forman SJ, Friedberg JW, Fuchs EJ, Gore SD, Jagannath S, Kahl BS, Kline J, Kochenderfer JN, Kwak LW, Levy R, de Lima M, Litzow MR, Mahindra A, Miller J, Munshi NC, Orlowski RZ, Pagel JM, Porter DL, Russell SJ, Schwartz K, Shipp MA, Siegel D, Stone RM, Tallman MS, Timmerman JM, Van Rhee F, Waller EK, Welsh A, Werner M, Wiernik PH, Dhodapkar MV. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of hematologic malignancies: multiple myeloma, lymphoma, and acute leukemia. J Immunother Cancer 2016; 4:90. [PMID: 28018601 PMCID: PMC5168808 DOI: 10.1186/s40425-016-0188-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/07/2016] [Indexed: 12/24/2022] Open
Abstract
Increasing knowledge concerning the biology of hematologic malignancies as well as the role of the immune system in the control of these diseases has led to the development and approval of immunotherapies that are resulting in impressive clinical responses. Therefore, the Society for Immunotherapy of Cancer (SITC) convened a hematologic malignancy Cancer Immunotherapy Guidelines panel consisting of physicians, nurses, patient advocates, and patients to develop consensus recommendations for the clinical application of immunotherapy for patients with multiple myeloma, lymphoma, and acute leukemia. These recommendations were developed following the previously established process based on the Institute of Medicine's clinical practice guidelines. In doing so, a systematic literature search was performed for high-impact studies from 2004 to 2014 and was supplemented with further literature as identified by the panel. The consensus panel met in December of 2014 with the goal to generate consensus recommendations for the clinical use of immunotherapy in patients with hematologic malignancies. During this meeting, consensus panel voting along with discussion were used to rate and review the strength of the supporting evidence from the literature search. These consensus recommendations focus on issues related to patient selection, toxicity management, clinical endpoints, and the sequencing or combination of therapies. Overall, immunotherapy is rapidly emerging as an effective therapeutic strategy for the management of hematologic malignances. Evidence-based consensus recommendations for its clinical application are provided and will be updated as the field evolves.
Collapse
Affiliation(s)
- Michael Boyiadzis
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical Center, 5150 Centre Avenue, Suite 564, Pittsburg, PA 15232 USA
| | - Michael R. Bishop
- Hematopoietic Cellular Therapy Program, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Rafat Abonour
- Indiana University School of Medicine, 980 W. Walnut St., Walther Hall-R3, C400, Indianapolis, IN 46202 USA
| | | | | | - David Avigan
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215 USA
| | - Lisa Barbarotta
- Smilow Cancer Hospital at Yale New Haven, 35 Park Street, New Haven, CT 06519 USA
| | - Austin John Barrett
- National Institutes of Health, Building 10-CRC Room 3-5330, Bethesda, MD 20814 USA
| | - Koen Van Besien
- Weill Cornell Medical College, 407 E 71st St, New York, NY 10065 USA
| | | | - Ivan Borrello
- Johns Hopkins School of Medicine, 1650 Orleans St, Baltimore, MD 21231 USA
| | - Joshua Brody
- Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY 10029 USA
| | - Jill Brufsky
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA 15232 USA
| | - Mitchell Cairo
- New York Medical College at Maria Fareri Children’s Hospital, 100 Woods Road, Valhalla, New York 10595 USA
| | - Ajai Chari
- Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY 10029 USA
| | - Adam Cohen
- Abramson Cancer Center at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Jorge Cortes
- MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Stephen J. Forman
- City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010 USA
| | - Jonathan W. Friedberg
- Wilmot Cancer Institute, University of Rochester, 601 Elmwood Avenue, Box 704, Rochester, NY 14642 USA
| | - Ephraim J. Fuchs
- Johns Hopkins University School of Medicine, 401 N. Broadway, Baltimore, MD 21231 USA
| | - Steven D. Gore
- Yale Cancer Center, 333 Cedar Street, New Haven, CT 06511 USA
| | - Sundar Jagannath
- Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY 10029 USA
| | - Brad S. Kahl
- Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Justin Kline
- The University of Chicago, 5841 S. Maryland Ave, Chicago, IL 60637 USA
| | - James N. Kochenderfer
- National Institutes of Health, National Cancer Institute, 8500 Roseweood Drive, Bethesda, MD 20814 USA
| | - Larry W. Kwak
- City of Hope National Medical Center, 1500 E. Duarte Road, Beckman Bldg., Room 4117, Duarte, CA 91010 USA
| | - Ronald Levy
- Division of Medical Oncology, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305 USA
| | - Marcos de Lima
- Department of Medicine-Hematology and Oncology, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106 USA
| | - Mark R. Litzow
- Department of Hematology, Mayo Clinic Cancer Center, 200 First Street SW, Rochester, MN 55905 USA
| | - Anuj Mahindra
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, Box 0324, San Francisco, CA 94143 USA
| | - Jeffrey Miller
- Division of Hematology/Oncology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN 55455 USA
| | - Nikhil C. Munshi
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana B106, Boston, MA 02215 USA
| | - Robert Z. Orlowski
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 429, Houston, TX 77030 USA
| | - John M. Pagel
- Swedish Cancer Institute, 1221 Madison Street, Suite 1020, Seattle, WA 98104 USA
| | - David L. Porter
- University of Pennsylvania, 3400 Civic Center Blvd, PCAM 12 South Pavilion, Philadelphia, PA 19104 USA
| | | | - Karl Schwartz
- Patients Against Lymphoma, 3774 Buckwampum Road, Riegelsville, PA 18077 USA
| | - Margaret A. Shipp
- Dana-Farber Cancer Institute, 450 Brookline Ave, Mayer 513, Boston, MA 02215 USA
| | - David Siegel
- Hackensack University Medical Center, 92 2nd St., Hackensack, NJ 07601 USA
| | - Richard M. Stone
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215 USA
| | - Martin S. Tallman
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065 USA
| | - John M. Timmerman
- University of California, Los Angeles, 10833 LeConte Ave., Los Angeles, CA 90095 USA
| | - Frits Van Rhee
- University of Arkansas for Medical Sciences, Myeloma Institute, 4301 W Markham #816, Little Rock, AR 72205 USA
| | - Edmund K. Waller
- Winship Cancer Institute, Emory University, 1365B Clifton Road NE, Atlanta, GA 30322 USA
| | - Ann Welsh
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA 15213 USA
| | - Michael Werner
- Patient Advocate, 33 East Bellevue Place, Chicago, IL 60611 USA
| | - Peter H. Wiernik
- Cancer Research Foundation of New York, 43 Longview Lane, Chappaqua, NY 10514 USA
| | - Madhav V. Dhodapkar
- Department of Hematology & Immunobiology, Yale University, 333 Cedar Street, Box 208021, New Haven, CT 06510 USA
| |
Collapse
|
181
|
Braun DA, Burke KP, Van Allen EM. Genomic Approaches to Understanding Response and Resistance to Immunotherapy. Clin Cancer Res 2016. [PMID: 27698000 DOI: 10.1158/1078-0432.ccr-16-0066.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunotherapy has led to a paradigm shift in the treatment of some malignancies, providing long-term, durable responses for patients with advanced cancers. However, such therapy has benefited only a subset of patients, with some patients failing to respond to treatment at all and others achieving a limited response followed by tumor progression. Understanding factors contributing to an effective response and further elucidating mechanisms of resistance will be crucial as these therapies are applied more broadly. Genomics-based approaches have significantly advanced the study of response and resistance to immunotherapy in general, and to immune checkpoint blockade more specifically. Here, we review how genomic and transcriptomic approaches have identified both somatic and germline positive correlates of response, including high mutational/neoantigen load and low intratumoral heterogeneity, among others. The genomic analysis of resistant tumors has additionally identified crucial factors involved in resistance to immune checkpoint blockade, including loss of PTEN and upregulation of other immune checkpoints. Overall, the continued use of genomic techniques at the point of care, combined with appropriate functional studies, would ideally lead to a better understanding of why certain patients respond to immune-based therapies, allowing clinicians to identify the subset of patients likely to benefit from such therapy, and potentially providing insight into how other therapies may be added in combination to increase the number of patients who may benefit from immunotherapy. Clin Cancer Res; 22(23); 5642-50. ©2016 AACR.
Collapse
Affiliation(s)
- David A Braun
- Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Kelly P Burke
- Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Eliezer M Van Allen
- Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts. .,Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
182
|
Rapid and Continued T-Cell Differentiation into Long-term Effector and Memory Stem Cells in Vaccinated Melanoma Patients. Clin Cancer Res 2016; 23:3285-3296. [DOI: 10.1158/1078-0432.ccr-16-1708] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/30/2016] [Accepted: 11/05/2016] [Indexed: 11/16/2022]
|
183
|
Gupta S, Chaudhary K, Dhanda SK, Kumar R, Kumar S, Sehgal M, Nagpal G, Raghava GPS. A Platform for Designing Genome-Based Personalized Immunotherapy or Vaccine against Cancer. PLoS One 2016; 11:e0166372. [PMID: 27832200 PMCID: PMC5104390 DOI: 10.1371/journal.pone.0166372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 10/27/2016] [Indexed: 02/01/2023] Open
Abstract
Due to advancement in sequencing technology, genomes of thousands of cancer tissues or cell-lines have been sequenced. Identification of cancer-specific epitopes or neoepitopes from cancer genomes is one of the major challenges in the field of immunotherapy or vaccine development. This paper describes a platform Cancertope, developed for designing genome-based immunotherapy or vaccine against a cancer cell. Broadly, the integrated resources on this platform are apportioned into three precise sections. First section explains a cancer-specific database of neoepitopes generated from genome of 905 cancer cell lines. This database harbors wide range of epitopes (e.g., B-cell, CD8+ T-cell, HLA class I, HLA class II) against 60 cancer-specific vaccine antigens. Second section describes a partially personalized module developed for predicting potential neoepitopes against a user-specific cancer genome. Finally, we describe a fully personalized module developed for identification of neoepitopes from genomes of cancerous and healthy cells of a cancer-patient. In order to assist the scientific community, wide range of tools are incorporated in this platform that includes screening of epitopes against human reference proteome (http://www.imtech.res.in/raghava/cancertope/).
Collapse
Affiliation(s)
- Sudheer Gupta
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Kumardeep Chaudhary
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Sandeep Kumar Dhanda
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Rahul Kumar
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Shailesh Kumar
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Manika Sehgal
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Gandharva Nagpal
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | | |
Collapse
|
184
|
Zolkind P, Dunn GP, Lin T, Griffith M, Griffith OL, Uppaluri R. Neoantigens in immunotherapy and personalized vaccines: Implications for head and neck squamous cell carcinoma. Oral Oncol 2016; 71:169-176. [PMID: 27751760 DOI: 10.1016/j.oraloncology.2016.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 12/15/2022]
Abstract
The recent success of immunotherapies has demonstrated the potency of tumor-specific immune cells in mediating tumor rejection and generating durable tumor immunity. Our understanding of the scientific basis of these responses results from the confluence of a better comprehension of the cancer immunoediting process and the revolution in next generation sequencing of cancer genomes. Recent evidence suggests that T cell specificity for cancer cell expressed mutant proteins - termed neoantigens - is an important component of immune mediated tumor rejection. Improved neoantigen prediction algorithms have made it possible to predict and monitor immune responses to checkpoint inhibitors and adoptively transferred autologous lymphocytes and have enabled the development of tumor-specific therapeutic vaccines. Herein, we review the current research on cancer neoantigens in immunotherapies and its implications for the future of head and neck cancer management.
Collapse
Affiliation(s)
- Paul Zolkind
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, United States
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, United States; Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, United States
| | - Tianxiang Lin
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, United States
| | - Malachi Griffith
- McDonnell Genome Institute and Department of Genetics, Washington University School of Medicine, St. Louis, United States
| | - Obi L Griffith
- Department of Medicine, Washington University School of Medicine, St. Louis, United States
| | - Ravindra Uppaluri
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, United States; Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA, United States.
| |
Collapse
|
185
|
Braun DA, Burke KP, Van Allen EM. Genomic Approaches to Understanding Response and Resistance to Immunotherapy. Clin Cancer Res 2016; 22:5642-5650. [PMID: 27698000 DOI: 10.1158/1078-0432.ccr-16-0066] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/19/2016] [Accepted: 08/25/2016] [Indexed: 12/12/2022]
Abstract
Immunotherapy has led to a paradigm shift in the treatment of some malignancies, providing long-term, durable responses for patients with advanced cancers. However, such therapy has benefited only a subset of patients, with some patients failing to respond to treatment at all and others achieving a limited response followed by tumor progression. Understanding factors contributing to an effective response and further elucidating mechanisms of resistance will be crucial as these therapies are applied more broadly. Genomics-based approaches have significantly advanced the study of response and resistance to immunotherapy in general, and to immune checkpoint blockade more specifically. Here, we review how genomic and transcriptomic approaches have identified both somatic and germline positive correlates of response, including high mutational/neoantigen load and low intratumoral heterogeneity, among others. The genomic analysis of resistant tumors has additionally identified crucial factors involved in resistance to immune checkpoint blockade, including loss of PTEN and upregulation of other immune checkpoints. Overall, the continued use of genomic techniques at the point of care, combined with appropriate functional studies, would ideally lead to a better understanding of why certain patients respond to immune-based therapies, allowing clinicians to identify the subset of patients likely to benefit from such therapy, and potentially providing insight into how other therapies may be added in combination to increase the number of patients who may benefit from immunotherapy. Clin Cancer Res; 22(23); 5642-50. ©2016 AACR.
Collapse
Affiliation(s)
- David A Braun
- Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Kelly P Burke
- Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Eliezer M Van Allen
- Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts. .,Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
186
|
Platten M, Bunse L, Wick W, Bunse T. Concepts in glioma immunotherapy. Cancer Immunol Immunother 2016; 65:1269-75. [PMID: 27460064 PMCID: PMC11029493 DOI: 10.1007/s00262-016-1874-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 07/20/2016] [Indexed: 01/12/2023]
Abstract
Immunotherapeutic concepts in neurooncology have been developed for many decades but have mainly been hampered by poor definition of relevant antigens and selective measures to target the central nervous system. Independent of the recent remarkable successes in clinical immunooncology with checkpoint inhibitors and vaccines, immunotherapy of brain tumors in general and gliomas in particular has evolved with novel neurooncology-specific concepts over the past years providing new phase 1 approaches of individualized immunotherapy to first phase three clinical trials. These concepts are driven by a high medical need in the absence of approved targeted therapies and refute the classic dogma that the central nervous system is immune-privileged and hence inaccessible to potent antitumor immunity. Instead, measures have been taken to improve the odds for successful immunotherapies, including rational targeting of relevant antigens and integration of immunotherapies into standard of care primary radiochemotherapy to increase the efficacy of antitumor immunity in a meaningful time window. This review highlights concepts and challenges associated with epitope discovery and selection and trial design.
Collapse
Affiliation(s)
- Michael Platten
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
- Department of Neurology, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
- National Center of Tumor Diseases (NCT), University Hospital, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany.
| | - Lukas Bunse
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- National Center of Tumor Diseases (NCT), University Hospital, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
- DKTK Clinical Cooperation Unit Neurooncology, German Cancer Research Center [DKFZ], Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Theresa Bunse
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| |
Collapse
|
187
|
Frazzi R, Auffray C, Ferrari A, Filippini P, Rutella S, Cesario A. Integrative systems medicine approaches to identify molecular targets in lymphoid malignancies. J Transl Med 2016; 14:252. [PMID: 27580852 PMCID: PMC5007715 DOI: 10.1186/s12967-016-1018-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/18/2016] [Indexed: 12/22/2022] Open
Abstract
Although survival rates for lymphoproliferative disorders are steadily increasing both in the US and in Europe, there is need for optimizing front-line therapies and developing more effective salvage strategies. Recent advances in molecular genetics have highlighted the biological diversity of lymphoproliferative disorders. In particular, integrative approaches including whole genome sequencing, whole exome sequencing, and transcriptome or RNA sequencing have been instrumental to the identification of molecular targets for treatment. Herein, we will discuss how genomic, epigenomic and proteomic approaches in lymphoproliferative disorders have supported the discovery of molecular lesions and their therapeutic targeting in the clinic.
Collapse
Affiliation(s)
- Raffaele Frazzi
- Laboratory of Translational Research, IRCCS "Arcispedale S. Maria Nuova", Reggio Emilia, Italy
| | - Charles Auffray
- European Institute for Systems Biology and Medicine (EISBM), Paris, France
| | - Angela Ferrari
- Division of Hematology, IRCCS "Arcispedale S. Maria Nuova", Reggio Emilia, Italy
| | - Perla Filippini
- Division of Translational Medicine, Sidra Medical and Research Centre, Doha, Qatar
| | - Sergio Rutella
- John van Geest Cancer Research Centre, College of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, UK.
| | - Alfredo Cesario
- Clinical Governance and International Research Activities, Fondazione Policlinico Gemelli, Rome, Italy.,Division of Thoracic Surgery, Università Cattolica del Sacro Cuore, Rome, Italy.,European Association of Systems Medicine, Aachen, Germany
| |
Collapse
|
188
|
Bobisse S, Foukas PG, Coukos G, Harari A. Neoantigen-based cancer immunotherapy. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:262. [PMID: 27563649 DOI: 10.21037/atm.2016.06.17] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Emerging clinical evidence on the role of the antitumor activity of the immune system has generated great interest in immunotherapy in all cancer types. Recent clinical data clearly demonstrated that human tumor cells express antigenic peptides (epitopes) that can be recognized by autologous tumor-specific T cells and that enhancement of such immune reactivity can potentially lead to cancer control and cancer regression in patients with advanced disease. However, in most cases, it is unclear which tumor antigens (Ags) mediated cancer regression. Mounting evidence indicates that numerous endogenous mutated cancer proteins, a hallmark of tumor cells, can be processed into peptides and presented on the surface of tumor cells, leading to their immune recognition in vivo as "non-self" or foreign. Massively parallel sequencing has now overcome the challenge of rapidly identifying the comprehensive mutational spectrum of individual tumors (i.e., the "mutanome") and current technologies, as well as computational tools, have emerged that allow the identification of private epitopes derived from their mutanome and called neoantigens (neoAgs). On this basis, both CD4(+) and CD8(+) neoantigen-specific T cells have been identified in multiple human cancers and shown to be associated with a favorable clinical outcome. Notably, emerging data also indicate that neoantigen recognition represents a major factor in the activity of clinical immunotherapies. In the post-genome era, the mutanome holds promise as a long-awaited 'gold mine' for the discovery of unique cancer cell targets, which are exclusively tumor-specific and unlikely to drive immune tolerance, hence offering the chance for highly promising clinical programs of cancer immunotherapy.
Collapse
Affiliation(s)
- Sara Bobisse
- Ludwig Cancer Center and Department of Oncology, University of Lausanne, Lausanne, Switzerland;; Center of Experimental Therapeutics, University of Lausanne, Lausanne, Switzerland
| | - Periklis G Foukas
- Ludwig Cancer Center and Department of Oncology, University of Lausanne, Lausanne, Switzerland;; Center of Experimental Therapeutics, University of Lausanne, Lausanne, Switzerland;; 2nd Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, Attikon University Hospital, Athens, Greece
| | - George Coukos
- Ludwig Cancer Center and Department of Oncology, University of Lausanne, Lausanne, Switzerland;; Center of Experimental Therapeutics, University of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Ludwig Cancer Center and Department of Oncology, University of Lausanne, Lausanne, Switzerland;; Center of Experimental Therapeutics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
189
|
Li B, Severson E, Pignon JC, Zhao H, Li T, Novak J, Jiang P, Shen H, Aster JC, Rodig S, Signoretti S, Liu JS, Liu XS. Comprehensive analyses of tumor immunity: implications for cancer immunotherapy. Genome Biol 2016; 17:174. [PMID: 27549193 PMCID: PMC4993001 DOI: 10.1186/s13059-016-1028-7] [Citation(s) in RCA: 1517] [Impact Index Per Article: 189.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 07/15/2016] [Indexed: 02/07/2023] Open
Abstract
Background Understanding the interactions between tumor and the host immune system is critical to finding prognostic biomarkers, reducing drug resistance, and developing new therapies. Novel computational methods are needed to estimate tumor-infiltrating immune cells and understand tumor–immune interactions in cancers. Results We analyze tumor-infiltrating immune cells in over 10,000 RNA-seq samples across 23 cancer types from The Cancer Genome Atlas (TCGA). Our computationally inferred immune infiltrates associate much more strongly with patient clinical features, viral infection status, and cancer genetic alterations than other computational approaches. Analysis of cancer/testis antigen expression and CD8 T-cell abundance suggests that MAGEA3 is a potential immune target in melanoma, but not in non-small cell lung cancer, and implicates SPAG5 as an alternative cancer vaccine target in multiple cancers. We find that melanomas expressing high levels of CTLA4 separate into two distinct groups with respect to CD8 T-cell infiltration, which might influence clinical responses to anti-CTLA4 agents. We observe similar dichotomy of TIM3 expression with respect to CD8 T cells in kidney cancer and validate it experimentally. The abundance of immune infiltration, together with our downstream analyses and findings, are accessible through TIMER, a public resource at http://cistrome.org/TIMER. Conclusions We develop a computational approach to study tumor-infiltrating immune cells and their interactions with cancer cells. Our resource of immune-infiltrate levels, clinical associations, as well as predicted therapeutic markers may inform effective cancer vaccine and checkpoint blockade therapies. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-1028-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bo Li
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, 450 Brookline Ave., Boston, MA, 02215, USA.,Department of Statistics, Harvard University, 1 Oxford St., Cambridge, MA, 02138, USA
| | - Eric Severson
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, 450 Brookline Ave., Boston, MA, 02215, USA.,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St., Boston, MA, 02215, USA
| | - Jean-Christophe Pignon
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St., Boston, MA, 02215, USA
| | - Haoquan Zhao
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, 450 Brookline Ave., Boston, MA, 02215, USA
| | - Taiwen Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 14 Renmin South Rd 3rd Section, Wuhou, Chengdu, Sichuan, 610041, China
| | - Jesse Novak
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St., Boston, MA, 02215, USA
| | - Peng Jiang
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, 450 Brookline Ave., Boston, MA, 02215, USA
| | - Hui Shen
- Center for Epigenetics, Van Andel Research Institute, 333 Bostwick Ave N.E., Grand Rapids, MI, 49503, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St., Boston, MA, 02215, USA
| | - Scott Rodig
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St., Boston, MA, 02215, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St., Boston, MA, 02215, USA
| | - Jun S Liu
- Department of Statistics, Harvard University, 1 Oxford St., Cambridge, MA, 02138, USA.
| | - X Shirley Liu
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, 450 Brookline Ave., Boston, MA, 02215, USA.
| |
Collapse
|
190
|
Siska PJ, van der Windt GJW, Kishton RJ, Cohen S, Eisner W, MacIver NJ, Kater AP, Weinberg JB, Rathmell JC. Suppression of Glut1 and Glucose Metabolism by Decreased Akt/mTORC1 Signaling Drives T Cell Impairment in B Cell Leukemia. THE JOURNAL OF IMMUNOLOGY 2016; 197:2532-40. [PMID: 27511728 DOI: 10.4049/jimmunol.1502464] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 07/15/2016] [Indexed: 12/14/2022]
Abstract
Leukemia can promote T cell dysfunction and exhaustion that contributes to increased susceptibility to infection and mortality. The treatment-independent mechanisms that mediate leukemia-associated T cell impairments are poorly understood, but metabolism tightly regulates T cell function and may contribute. In this study, we show that B cell leukemia causes T cells to become activated and hyporesponsive with increased PD-1 and TIM3 expression similar to exhausted T cells and that T cells from leukemic hosts become metabolically impaired. Metabolic defects included reduced Akt/mammalian target of rapamycin complex 1 (mTORC1) signaling, decreased expression of the glucose transporter Glut1 and hexokinase 2, and reduced glucose uptake. These metabolic changes correlated with increased regulatory T cell frequency and expression of PD-L1 and Gal-9 on both leukemic and stromal cells in the leukemic microenvironment. PD-1, however, was not sufficient to drive T cell impairment, as in vivo and in vitro anti-PD-1 blockade on its own only modestly improved T cell function. Importantly, impaired T cell metabolism directly contributed to dysfunction, as a rescue of T cell metabolism by genetically increasing Akt/mTORC1 signaling or expression of Glut1 partially restored T cell function. Enforced Akt/mTORC1 signaling also decreased expression of inhibitory receptors TIM3 and PD-1, as well as partially improved antileukemia immunity. Similar findings were obtained in T cells from patients with acute or chronic B cell leukemia, which were also metabolically exhausted and had defective Akt/mTORC1 signaling, reduced expression of Glut1 and hexokinase 2, and decreased glucose metabolism. Thus, B cell leukemia-induced inhibition of T cell Akt/mTORC1 signaling and glucose metabolism drives T cell dysfunction.
Collapse
Affiliation(s)
- Peter J Siska
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710; Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232; Department of Cancer Biology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | | | - Rigel J Kishton
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710
| | - Sivan Cohen
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710
| | - William Eisner
- Department of Pediatrics, Duke University, Durham, NC 27710
| | | | - Arnon P Kater
- Department of Hematology, Academic Medical Center, 1100 DD Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - J Brice Weinberg
- Department of Medicine, Duke University, Durham, NC 27708; and Department of Medicine, Durham Veterans Affairs Medical Center, Durham, NC 27705
| | - Jeffrey C Rathmell
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710; Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232; Department of Cancer Biology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232;
| |
Collapse
|
191
|
Neagu MR, Reardon DA. An Update on the Role of Immunotherapy and Vaccine Strategies for Primary Brain Tumors. Curr Treat Options Oncol 2016; 16:54. [PMID: 26454859 DOI: 10.1007/s11864-015-0371-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Existing therapies for glioblastoma (GBM), the most common malignant primary brain tumor in adults, have fallen short of improving the dismal patient outcomes, with an average 14-16-month median overall survival. The biological complexity and adaptability of GBM, redundancy of dysregulated signaling pathways, and poor penetration of therapies through the blood-brain barrier contribute to poor therapeutic progress. The current standard of care for newly diagnosed GBM consists of maximal safe resection, followed by fractionated radiotherapy combined with concurrent temozolomide (TMZ) and 6-12 cycles of adjuvant TMZ. At progression, bevacizumab with or without additional chemotherapy is an option for salvage therapy. The recent FDA approval of sipuleucel-T for prostate cancer and ipilumimab, nivolumab, and pembrolizumab for select solid tumors and the ongoing trials showing clinical efficacy and response durability herald a new era of cancer treatment with the potential to change standard-of-care treatment across multiple cancers. The evaluation of various immunotherapeutics is advancing for GBM, putting into question the dogma of the CNS as an immuno-privileged site. While the field is yet young, both active immunotherapy involving vaccine strategies and cellular therapy as well as reversal of GBM-induced global immune-suppression through immune checkpoint blockade are showing promising results and revealing essential immunological insights regarding kinetics of the immune response, immune evasion, and correlative biomarkers. The future holds exciting promise in establishing new treatment options for GBM that harness the patients' own immune system by activating it with immune checkpoint inhibitors, providing specificity using vaccine therapy, and allowing for modulation and enhancement by combinatorial approaches.
Collapse
Affiliation(s)
- Martha R Neagu
- Dana-Farber Cancer Institute, G4200, 44 Binney St, Boston, MA, 02115, USA
- Pappas Center for Neuro-Oncology, Massachusetts General Hospital, WACC 8-835m 55 Fruit St, Boston, MA, 02114, USA
| | - David A Reardon
- Dana-Farber Cancer Institute, G4200, 44 Binney St, Boston, MA, 02115, USA.
| |
Collapse
|
192
|
Garofalo A, Sholl L, Reardon B, Taylor-Weiner A, Amin-Mansour A, Miao D, Liu D, Oliver N, MacConaill L, Ducar M, Rojas-Rudilla V, Giannakis M, Ghazani A, Gray S, Janne P, Garber J, Joffe S, Lindeman N, Wagle N, Garraway LA, Van Allen EM. The impact of tumor profiling approaches and genomic data strategies for cancer precision medicine. Genome Med 2016; 8:79. [PMID: 27460824 PMCID: PMC4962446 DOI: 10.1186/s13073-016-0333-9] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 07/08/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The diversity of clinical tumor profiling approaches (small panels to whole exomes with matched or unmatched germline analysis) may engender uncertainty about their benefits and liabilities, particularly in light of reported germline false positives in tumor-only profiling and use of global mutational and/or neoantigen data. The goal of this study was to determine the impact of genomic analysis strategies on error rates and data interpretation across contexts and ancestries. METHODS We modeled common tumor profiling modalities-large (n = 300 genes), medium (n = 48 genes), and small (n = 15 genes) panels-using clinical whole exomes (WES) from 157 patients with lung or colon adenocarcinoma. We created a tumor-only analysis algorithm to assess germline false positive rates, the impact of patient ancestry on tumor-only results, and neoantigen detection. RESULTS After optimizing a germline filtering strategy, the germline false positive rate with tumor-only large panel sequencing was 14 % (144/1012 variants). For patients whose tumor-only results underwent molecular pathologist review (n = 91), 50/54 (93 %) false positives were correctly interpreted as uncertain variants. Increased germline false positives were observed in tumor-only sequencing of non-European compared with European ancestry patients (p < 0.001; Fisher's exact) when basic germline filtering approaches were used; however, the ExAC database (60,706 germline exomes) mitigated this disparity (p = 0.53). Matched and unmatched large panel mutational load correlated with WES mutational load (r(2) = 0.99 and 0.93, respectively; p < 0.001). Neoantigen load also correlated (r(2) = 0.80; p < 0.001), though WES identified a broader spectrum of neoantigens. Small panels did not predict mutational or neoantigen load. CONCLUSIONS Large tumor-only targeted panels are sufficient for most somatic variant identification and mutational load prediction if paired with expanded germline analysis strategies and molecular pathologist review. Paired germline sequencing reduced overall false positive mutation calls and WES provided the most neoantigens. Without patient-matched germline data, large germline databases are needed to minimize false positive mutation calling and mitigate ethnic disparities.
Collapse
Affiliation(s)
- Andrea Garofalo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Brendan Reardon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Amaro Taylor-Weiner
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Ali Amin-Mansour
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Diana Miao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Nelly Oliver
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA
| | - Laura MacConaill
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA.,Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Matthew Ducar
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Arezou Ghazani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA
| | - Stacy Gray
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA
| | - Pasi Janne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA
| | - Judy Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA
| | - Steve Joffe
- Department of Medical Ethics and Health Policy, University of Pennsylvania, Philadelphia, PA, USA
| | - Neal Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Nikhil Wagle
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA.,Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02115, USA
| | - Levi A Garraway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA. .,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA. .,Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02115, USA.
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02115, USA. .,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA. .,Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
193
|
Is there really a specific graft-versus-leukaemia effect? Bone Marrow Transplant 2016; 51:1413-1415. [DOI: 10.1038/bmt.2016.183] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 05/21/2016] [Indexed: 11/09/2022]
|
194
|
Porta-Pardo E, Godzik A. Mutation Drivers of Immunological Responses to Cancer. Cancer Immunol Res 2016; 4:789-98. [PMID: 27401919 DOI: 10.1158/2326-6066.cir-15-0233] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 06/06/2016] [Indexed: 01/10/2023]
Abstract
In cancer immunology, somatic missense mutations have been mostly studied with regard to their role in the generation of neoantigens. However, growing evidence suggests that mutations in certain genes, such as CASP8 or TP53, influence the immune response against a tumor by other mechanisms. Identifying these genes and mechanisms is important because, just as the identification of cancer driver genes led to the development of personalized cancer therapies, a comprehensive catalog of such cancer immunity drivers will aid in the development of therapies aimed at restoring antitumor immunity. Here, we present an algorithm, domainXplorer, that can be used to identify potential cancer immunity drivers. To demonstrate its potential, we used it to analyze a dataset of 5,164 tumor samples from The Cancer Genome Atlas (TCGA) and to identify protein domains in which mutation status correlates with the presence of immune cells in cancer tissue (immune infiltrate). We identified 122 such protein regions, including several that belong to proteins with known roles in immune response, such as C2, CD163L1, or FCγR2A. In several cases, we show that mutations within the same protein can be associated with more or less immune cell infiltration, depending on the specific domain mutated. These results expand the catalog of potential cancer immunity drivers and highlight the importance of taking into account the structural context of somatic mutations when analyzing their potential association with immune phenotypes. Cancer Immunol Res; 4(9); 789-98. ©2016 AACR.
Collapse
Affiliation(s)
- Eduard Porta-Pardo
- Program on Bioinformatics and Systems Biology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Adam Godzik
- Program on Bioinformatics and Systems Biology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California.
| |
Collapse
|
195
|
Austin R, Smyth MJ, Lane SW. Harnessing the immune system in acute myeloid leukaemia. Crit Rev Oncol Hematol 2016; 103:62-77. [DOI: 10.1016/j.critrevonc.2016.04.020] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/13/2016] [Accepted: 04/28/2016] [Indexed: 12/13/2022] Open
|
196
|
Nahas MR, Avigan D. Challenges in vaccine therapy in hematological malignancies and strategies to overcome them. Expert Opin Biol Ther 2016; 16:1093-104. [DOI: 10.1080/14712598.2016.1190828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
197
|
Kim JM, Chen DS. Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann Oncol 2016; 27:1492-504. [PMID: 27207108 DOI: 10.1093/annonc/mdw217] [Citation(s) in RCA: 426] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/13/2016] [Indexed: 12/13/2022] Open
Abstract
The emergence of programmed death-ligand 1 (PD-L1)/programmed death-1 (PD-1)-targeted therapy has demonstrated the importance of the PD-L1 : PD-1 interaction in inhibiting anticancer T-cell immunity in multiple human cancers, generating durable responses and extended overall survival. However, not all patients treated with PD-L1/PD-1-targeted therapy experience tumor shrinkage, durable responses, or prolonged survival. To extend such benefits to more cancer patients, it is necessary to understand why some patients experience primary or secondary immune escape, in which the immune response is incapable of eradicating all cancer cells. Understanding immune escape from PD-L1/PD-1-targeted therapy will be important to the development of rational immune-combination therapy and predictive diagnostics and to the identification of novel immune targets. Factors that likely relate to immune escape include the lack of strong cancer antigens or epitopes recognized by T cells, minimal activation of cancer-specific T cells, poor infiltration of T cells into tumors, downregulation of the major histocompatibility complex on cancer cells, and immunosuppressive factors and cells in the tumor microenvironment. Precisely identifying and understanding these mechanisms of immune escape in individual cancer patients will allow for personalized cancer immunotherapy, in which monotherapy and combination immunotherapy are chosen based on the presence of specific immune biology. This approach may enable treatment with immunotherapy without inducing immune escape, resulting in a larger proportion of patients obtaining clinical benefit.
Collapse
Affiliation(s)
- J M Kim
- Genentech, South San Francisco
| | - D S Chen
- Genentech, South San Francisco Stanford Medical Oncology, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
198
|
Martin SD, Brown SD, Wick DA, Nielsen JS, Kroeger DR, Twumasi-Boateng K, Holt RA, Nelson BH. Low Mutation Burden in Ovarian Cancer May Limit the Utility of Neoantigen-Targeted Vaccines. PLoS One 2016; 11:e0155189. [PMID: 27192170 PMCID: PMC4871527 DOI: 10.1371/journal.pone.0155189] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/25/2016] [Indexed: 01/07/2023] Open
Abstract
Due to advances in sequencing technology, somatically mutated cancer antigens, or neoantigens, are now readily identifiable and have become compelling targets for immunotherapy. In particular, neoantigen-targeted vaccines have shown promise in several pre-clinical and clinical studies. However, to date, neoantigen-targeted vaccine studies have involved tumors with exceptionally high mutation burdens. It remains unclear whether neoantigen-targeted vaccines will be broadly applicable to cancers with intermediate to low mutation burdens, such as ovarian cancer. To address this, we assessed whether a derivative of the murine ovarian tumor model ID8 could be targeted with neoantigen vaccines. We performed whole exome and transcriptome sequencing on ID8-G7 cells. We identified 92 somatic mutations, 39 of which were transcribed, missense mutations. For the 17 top predicted MHC class I binding mutations, we immunized mice subcutaneously with synthetic long peptide vaccines encoding the relevant mutation. Seven of 17 vaccines induced robust mutation-specific CD4 and/or CD8 T cell responses. However, none of the vaccines prolonged survival of tumor-bearing mice in either the prophylactic or therapeutic setting. Moreover, none of the neoantigen-specific T cell lines recognized ID8-G7 tumor cells in vitro, indicating that the corresponding mutations did not give rise to bonafide MHC-presented epitopes. Additionally, bioinformatic analysis of The Cancer Genome Atlas data revealed that only 12% (26/220) of HGSC cases had a ≥90% likelihood of harboring at least one authentic, naturally processed and presented neoantigen versus 51% (80/158) of lung cancers. Our findings highlight the limitations of applying neoantigen-targeted vaccines to tumor types with intermediate/low mutation burdens.
Collapse
Affiliation(s)
- Spencer D. Martin
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, Canada
- Michael Smith’s Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, Canada
| | - Scott D. Brown
- Michael Smith’s Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, Canada
| | - Darin A. Wick
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Julie S. Nielsen
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - David R. Kroeger
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Kwame Twumasi-Boateng
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Robert A. Holt
- Michael Smith’s Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, Canada
- Molecular Biology and Biochemistry, Simon Fraser University, Vancouver, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Brad H. Nelson
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- Department of Microbiology and Biochemistry, University of Victoria, Victoria, Canada
| |
Collapse
|
199
|
Lai J, Zhou Z, Tang XJ, Gao ZB, Zhou J, Chen SQ. A Tumor-Specific Neo-Antigen Caused by a Frameshift Mutation in BAP1 Is a Potential Personalized Biomarker in Malignant Peritoneal Mesothelioma. Int J Mol Sci 2016; 17:ijms17050739. [PMID: 27187383 PMCID: PMC4881561 DOI: 10.3390/ijms17050739] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/28/2016] [Accepted: 05/04/2016] [Indexed: 01/28/2023] Open
Abstract
Malignant peritoneal mesothelioma (MPM) is an aggressive rare malignancy associated with asbestos exposure. A better understanding of the molecular pathogenesis of MPM will help develop a targeted therapy strategy. Oncogene targeted depth sequencing was performed on a tumor sample and paired peripheral blood DNA from a patient with malignant mesothelioma of the peritoneum. Four somatic base-substitutions in NOTCH2, NSD1, PDE4DIP, and ATP10B and 1 insert frameshift mutation in BAP1 were validated by the Sanger method at the transcriptional level. A 13-amino acids neo-peptide of the truncated Bap1 protein, which was produced as a result of this novel frameshift mutation, was predicted to be presented by this patient's HLA-B protein. The polyclonal antibody of the synthesized 13-mer neo-peptide was produced in rabbits. Western blotting results showed a good antibody-neoantigen specificity, and Immunohistochemistry (IHC) staining with the antibody of the neo-peptide clearly differentiated neoplastic cells from normal cells. A search of the Catalogue of Somatic Mutations in Cancer (COSMIC) database also revealed that 53.2% of mutations in BAP1 were frameshift indels with neo-peptide formation. An identified tumor-specific neo-antigen could be the potential molecular biomarker for personalized diagnosis to precisely subtype rare malignancies such as MPM.
Collapse
Affiliation(s)
- Jun Lai
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Zhan Zhou
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Xiao-Jing Tang
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Zhi-Bin Gao
- Department of Obstetrics and Gynecology, Yuyao People's Hospital, 800 Chengdong Road, Yuyao 315400, China.
| | - Jie Zhou
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Shu-Qing Chen
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| |
Collapse
|
200
|
Bassani-Sternberg M, Coukos G. Mass spectrometry-based antigen discovery for cancer immunotherapy. Curr Opin Immunol 2016; 41:9-17. [PMID: 27155075 DOI: 10.1016/j.coi.2016.04.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 04/19/2016] [Indexed: 12/14/2022]
Abstract
The antigenic landscape of tumors is distinct from healthy cells and has been the rationale behind a variety of vaccination trials. Typically the target tumor-associated antigens have been of self origin and have rarely induced effective anti-tumor responses. Recent data show that activation of the immune system by immune checkpoint blocking therapies leads to tumor rejection and that recognition of mutated antigens, known as 'neo-antigens' plays a key role. Discovery of neo-antigens relies mainly on prediction-based interrogation of the 'mutanome' using genomic information as input, followed by T-cell screening. Recent breakthroughs in mass spectrometry (MS) based immunopeptidomics have allowed the discovery of very large pools of naturally presented peptides, among them neo-epitopes. This review highlights the current progress related to neo-antigens discovery with emphasis on prediction algorithms and MS as well as the synergy of the two methodologies and how they can be exploited to develop effective personalized immunotherapy.
Collapse
Affiliation(s)
- Michal Bassani-Sternberg
- Ludwig Institute for Cancer Research, University of Lausanne Branch & Department of Oncology, CHUV, Lausanne, Switzerland.
| | - George Coukos
- Ludwig Institute for Cancer Research, University of Lausanne Branch & Department of Oncology, CHUV, Lausanne, Switzerland
| |
Collapse
|