301
|
Affiliation(s)
- Hiroshi Fukui
- Department of Gastroenterology, Endocrinology and Metabolism, Nara Medical University, Nara, Japan
| |
Collapse
|
302
|
Balachandran VP, Łuksza M, Zhao JN, Makarov V, Moral JA, Remark R, Herbst B, Askan G, Bhanot U, Senbabaoglu Y, Wells DK, Cary CIO, Grbovic-Huezo O, Attiyeh M, Medina B, Zhang J, Loo J, Saglimbeni J, Abu-Akeel M, Zappasodi R, Riaz N, Smoragiewicz M, Kelley ZL, Basturk O, Gönen M, Levine AJ, Allen PJ, Fearon DT, Merad M, Gnjatic S, Iacobuzio-Donahue CA, Wolchok JD, DeMatteo RP, Chan TA, Greenbaum BD, Merghoub T, Leach SD. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 2017; 551:512-516. [PMID: 29132146 PMCID: PMC6145146 DOI: 10.1038/nature24462] [Citation(s) in RCA: 835] [Impact Index Per Article: 104.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 10/02/2017] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma is a lethal cancer with fewer than 7% of patients surviving past 5 years. T-cell immunity has been linked to the exceptional outcome of the few long-term survivors, yet the relevant antigens remain unknown. Here we use genetic, immunohistochemical and transcriptional immunoprofiling, computational biophysics, and functional assays to identify T-cell antigens in long-term survivors of pancreatic cancer. Using whole-exome sequencing and in silico neoantigen prediction, we found that tumours with both the highest neoantigen number and the most abundant CD8+ T-cell infiltrates, but neither alone, stratified patients with the longest survival. Investigating the specific neoantigen qualities promoting T-cell activation in long-term survivors, we discovered that these individuals were enriched in neoantigen qualities defined by a fitness model, and neoantigens in the tumour antigen MUC16 (also known as CA125). A neoantigen quality fitness model conferring greater immunogenicity to neoantigens with differential presentation and homology to infectious disease-derived peptides identified long-term survivors in two independent datasets, whereas a neoantigen quantity model ascribing greater immunogenicity to increasing neoantigen number alone did not. We detected intratumoural and lasting circulating T-cell reactivity to both high-quality and MUC16 neoantigens in long-term survivors of pancreatic cancer, including clones with specificity to both high-quality neoantigens and predicted cross-reactive microbial epitopes, consistent with neoantigen molecular mimicry. Notably, we observed selective loss of high-quality and MUC16 neoantigenic clones on metastatic progression, suggesting neoantigen immunoediting. Our results identify neoantigens with unique qualities as T-cell targets in pancreatic ductal adenocarcinoma. More broadly, we identify neoantigen quality as a biomarker for immunogenic tumours that may guide the application of immunotherapies.
Collapse
Affiliation(s)
- Vinod P. Balachandran
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marta Łuksza
- The Simons Center for Systems Biology, Institute for Advanced Study, Princeton, NJ, USA
| | - Julia N. Zhao
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Makarov
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John Alec Moral
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Romain Remark
- Tisch Cancer Institute, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Herbst
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gokce Askan
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pathology Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Umesh Bhanot
- Pathology Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yasin Senbabaoglu
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel K. Wells
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Olivera Grbovic-Huezo
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc Attiyeh
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin Medina
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jennifer Zhang
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jennifer Loo
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph Saglimbeni
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohsen Abu-Akeel
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roberta Zappasodi
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem Riaz
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Radiation Oncology Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin Smoragiewicz
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Z. Larkin Kelley
- Cold Spring Harbor Laboratory, New York, NY, USA. Department of Microbiology and Immunology, Weill Cornell Medical School, New York, NY, USA
| | - Olca Basturk
- Pathology Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Mithat Gönen
- Biostatistics Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arnold J. Levine
- The Simons Center for Systems Biology, Institute for Advanced Study, Princeton, NJ, USA
| | - Peter J. Allen
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Douglas T. Fearon
- Cold Spring Harbor Laboratory, New York, NY, USA. Department of Microbiology and Immunology, Weill Cornell Medical School, New York, NY, USA
| | - Miriam Merad
- Tisch Cancer Institute, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Tisch Cancer Institute, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christine A. Iacobuzio-Donahue
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pathology Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jedd D. Wolchok
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Melanoma and Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, Cornell University, New York, NY, USA
- Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald P. DeMatteo
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy A. Chan
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Radiation Oncology Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin D. Greenbaum
- Tisch Cancer Institute, Departments of Medicine, Hematology and Medical Oncology, Oncological Sciences, and Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Taha Merghoub
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven D. Leach
- Departments of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
303
|
Bai J, Behera M, Bruner DW. The gut microbiome, symptoms, and targeted interventions in children with cancer: a systematic review. Support Care Cancer 2017; 26:427-439. [PMID: 29168036 DOI: 10.1007/s00520-017-3982-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 11/15/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE The gut microbiome plays a critical role in maintaining children's health and in preventing and treating children's disease. Current application of the gut microbiome in childhood cancer is still lacking. This study aimed to systematically review the following: (1) alternations in the gut microbiome throughout cancer treatment trajectories in children, (2) the associations between the gut microbiome and gastrointestinal (GI) symptoms and psychoneurological symptoms (PNS), and (3) the efficacy of therapeutic interventions in the gut microbiome in children with cancer. METHODS PubMed, EMBASE, the Cochrane Library, and the American Society of Clinical Oncology abstract were searched. Eligible studies included all study types in which the gut microbiome was primarily reported in children with cancer. The Mixed Methods Assessment Tool was used to evaluate the methodology quality of included studies. Seven studies met our eligibility criteria, including two cohort studies, two case-control studies, and three randomized controlled trails. RESULTS The findings showed that the diversity estimates of the gut microbiome in children with cancer were lower than those of healthy controls both pre- and post-treatment. Children with cancer showed a significantly lower relative abundance of healthy gut microbiome (e.g., Clostridium XIVa and Bifidobacterium) during and after cancer treatment. No adequate literature was identified to support the associations between dysbiosis of the gut microbiome and GI symptoms/PNS. The use of prebiotics (fructooligosaccharides) and probiotics (Bifidobacterium or Lactobacilli) appears to improve the microenvironment of the gut around 1 month (4-5 weeks) during chemotherapy rather than at the beginning of treatment. Data also suggest that both prebiotic and probiotic interventions decrease clinical side effects (e.g., infection and morbidity risk) in children with cancer. CONCLUSIONS This study adds to the evidence that dysbiosis of the gut microbiome can be improved using prebiotic and probiotic supplementations in children with cancer. More well-designed experimental studies are needed to confirm this conclusion. Further studies are needed to examine the associations between the gut microbiome and GI symptoms/PNS in childhood cancer.
Collapse
Affiliation(s)
- Jinbing Bai
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA.
| | - Madhusmita Behera
- Department of Hematology and Oncology, School of Medicine, Emory University, Atlanta, GA, USA.,Winship Research Informatics, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Deborah Watkins Bruner
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA.,Education and Training, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
304
|
Lactobacillus acidophilus Metabolizes Dietary Plant Glucosides and Externalizes Their Bioactive Phytochemicals. mBio 2017; 8:mBio.01421-17. [PMID: 29162708 PMCID: PMC5698550 DOI: 10.1128/mbio.01421-17] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Therapeutically active glycosylated phytochemicals are ubiquitous in the human diet. The human gut microbiota (HGM) modulates the bioactivities of these compounds, which consequently affect host physiology and microbiota composition. Despite a significant impact on human health, the key players and the underpinning mechanisms of this interplay remain uncharacterized. Here, we demonstrate the growth of Lactobacillus acidophilus on mono- and diglucosyl dietary plant glycosides (PGs) possessing small aromatic aglycones. Transcriptional analysis revealed the upregulation of host interaction genes and identified two loci that encode phosphotransferase system (PTS) transporters and phospho-β-glucosidases, which mediate the uptake and deglucosylation of these compounds, respectively. Inactivating these transport and hydrolysis genes abolished or severely reduced growth on PG, establishing the specificity of the loci to distinct groups of PGs. Following intracellular deglucosylation, the aglycones of PGs are externalized, rendering them available for absorption by the host or for further modification by other microbiota taxa. The PG utilization loci are conserved in L. acidophilus and closely related lactobacilli, in correlation with versatile growth on these compounds. Growth on the tested PG appeared more common among human gut lactobacilli than among counterparts from other ecologic niches. The PGs that supported the growth of L. acidophilus were utilized poorly or not at all by other common HGM strains, underscoring the metabolic specialization of L. acidophilus. These findings highlight the role of human gut L. acidophilus and select lactobacilli in the bioconversion of glycoconjugated phytochemicals, which is likely to have an important impact on the HGM and human host. Thousands of therapeutically active plant-derived compounds are widely present in berries, fruits, nuts, and beverages like tea and wine. The bioactivity and bioavailability of these compounds, which are typically glycosylated, are altered by microbial bioconversions in the human gut. Remarkably, little is known about the bioconversion of PGs by the gut microbial community, despite the significance of this metabolic facet to human health. Our work provides the first molecular insights into the metabolic routes of diet relevant and therapeutically active PGs by Lactobacillus acidophilus and related human gut lactobacilli. This taxonomic group is adept at metabolizing the glucoside moieties of select PG and externalizes their aglycones. The study highlights an important role of lactobacilli in the bioconversion of dietary PG and presents a framework from which to derive molecular insights into their metabolism by members of the human gut microbiota.
Collapse
|
305
|
Scheel AH, Baenfer G, Baretton G, Dietel M, Diezko R, Henkel T, Heukamp LC, Jasani B, Jöhrens K, Kirchner T, Lasitschka F, Petersen I, Reu S, Schildhaus HU, Schirmacher P, Schwamborn K, Sommer U, Stoss O, Tiemann M, Warth A, Weichert W, Wolf J, Büttner R, Rüschoff J. Interlaboratory concordance of PD-L1 immunohistochemistry for non-small-cell lung cancer. Histopathology 2017; 72:449-459. [PMID: 28851100 DOI: 10.1111/his.13375] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/25/2017] [Indexed: 12/18/2022]
Abstract
AIMS Programmed death ligand 1 (PD-L1) immunohistochemistry has become a mandatory diagnostic test in the treatment of lung cancer. Several research initiatives have started to harmonise the five PD-L1 immunohistochemistry assays that have been used in clinical trials. Here, we report data on interlaboratory and interassay concordance for commercial assays ('assays') and laboratory-developed tests (LDTs) at 10 German testing sites. METHODS AND RESULTS To assess interlaboratory concordance, a tissue microarray containing 21 pulmonary carcinoma specimens was centrally prepared. Pre-cut sections were stained at 10 sites by the use of assays 28-8, 22C3, SP263, and SP142, as well as 11 LDTs. Assay performance was evaluated with a second tissue microarray containing 11 cell lines with defined PD-L1 expression. Quality control was centrally performed by manual and digital analyses. The assays yielded reproducible IHC staining patterns at all sites. In agreement with previous studies, 22C3, 28-8 and SP263 showed similar staining patterns, whereas SP142 was distinct. Among the LDTs, six of 11 protocols showed staining patterns similar to those of assays 22C3 and 28-8. Interlaboratory concordance of tumour cell scoring by use of a six-step system was moderate (Light's κ = 0.43-0.69), whereas the clinically approved cut-offs of ≥1% and ≥50% showed substantial concordance (κ = 0.73-0.89). Immune cell scoring by the use of SP142 yielded moderate concordance (κ = 0.42). CONCLUSIONS The data confirm the previously described staining patterns of the assays, and show that they can be reproducibly employed at different sites. LDTs with staining results similar to those of the assays are implementable, but have to be carefully validated.
Collapse
Affiliation(s)
- Andreas H Scheel
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | - Gustavo Baretton
- Institute of Pathology, University Hospital Dresden, Dresden, Germany
| | - Manfred Dietel
- Institute of Pathology, Charité-University Hospital Berlin, Berlin, Germany
| | - Rolf Diezko
- Targos Molecular Pathology GmbH, Kassel, Germany
| | | | | | | | - Korinna Jöhrens
- Institute of Pathology, Charité-University Hospital Berlin, Berlin, Germany
| | - Thomas Kirchner
- Institute of Pathology, LMU University Hospital Munich, Munich, Germany
| | - Felix Lasitschka
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Iver Petersen
- Institute of Pathology, University Hospital Jena, Jena, Germany
| | - Simone Reu
- Institute of Pathology, LMU University Hospital Munich, Munich, Germany
| | | | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Ulrich Sommer
- Institute of Pathology, University Hospital Dresden, Dresden, Germany
| | - Oliver Stoss
- Targos Molecular Pathology GmbH, Kassel, Germany
| | - Markus Tiemann
- Institute for Haematopathology Hamburg, Hamburg, Germany
| | - Arne Warth
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, TUM University Hospital Munich, Munich, Germany
| | - Jürgen Wolf
- Medical Clinic I, University Hospital Cologne, Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany.,Targos Molecular Pathology GmbH, Kassel, Germany
| | - Josef Rüschoff
- Targos Molecular Pathology GmbH, Kassel, Germany.,Institute of Pathology Nordhessen, Kassel, Germany
| |
Collapse
|
306
|
Liu HX, Tao LL, Zhang J, Zhu YG, Zheng Y, Liu D, Zhou M, Ke H, Shi MM, Qu JM. Difference of lower airway microbiome in bilateral protected specimen brush between lung cancer patients with unilateral lobar masses and control subjects. Int J Cancer 2017; 142:769-778. [PMID: 29023689 DOI: 10.1002/ijc.31098] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 09/17/2017] [Accepted: 09/25/2017] [Indexed: 12/20/2022]
Abstract
The functional role of respiratory microbiota has attracted an accumulating attention recently. However, the role of respiratory microbiome in lung carcinogenesis is mostly unknown. Our study aimed to characterize and compare bilateral lower airway microbiome of lung cancer patients with unilateral lobar masses and control subjects. Protected bronchial specimen brushing samples were collected from 24 lung cancer patients with unilateral lobar masses (paired samples from cancerous site and the contralateral noncancerous site) and 18 healthy controls undergoing bronchoscopies and further analyzed by 16S rRNA amplicon sequencing. As results, significant decreases in microbial diversity were observed in patients with lung cancer in comparison to the controls, alpha diversity steadily declined from healthy site to noncancerous to cancerous site. Genus Streptococcus was significantly more abundant in cancer cases than the controls, while Staphylococcus was more abundant in the controls. The area under the curve of genus Streptococcus used to predict lung cancer was 0.693 (sensitivity = 87.5%, specificity = 55.6%). The abundance of genus Streptococcus and Neisseria displayed an increasing trend whereas Staphylococcus and Dialister gradually declined from healthy to noncancerous to cancerous site. Collectively, lung cancer-associated microbiota profile is distinct from that found in healthy controls, and the altered cancer-associated microbiota is not restricted to tumor tissue. The genus Streptococcus was abundant in lung cancer patients and exhibited moderate classification potential. The gradual microbiota profile shift from healthy site to noncancerous to paired cancerous site suggested a change of the microenvironment associated with the development of lung cancer.
Collapse
Affiliation(s)
- Hai-Xia Liu
- Huadong Hospital Affiliated to Fudan University, No. 221, West Yan An Road, Shanghai, 200040, China
| | - Li-Li Tao
- UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX
| | - Jing Zhang
- Zhongshan Hospital Affiliated to Fudan University, No. 180, Feng Lin Road, Shanghai, 200032, China
| | - Ying-Gang Zhu
- Huadong Hospital Affiliated to Fudan University, No. 221, West Yan An Road, Shanghai, 200040, China
| | - Yu Zheng
- Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, No. 2000, Jiangyue Road, Shanghai, 200112, China
| | - Dong Liu
- Huadong Hospital Affiliated to Fudan University, No. 221, West Yan An Road, Shanghai, 200040, China
| | - Min Zhou
- Rui Jin Hospital, School of Medicine, Shanghai Jiaotong University, No. 197, Rui Jin Er Road, Shanghai, 200025, China
| | - Hui Ke
- Shanghai Pulmonary Hospital Affiliated to Tongji University, No. 507, Yangpu District, Zheng Min Road, Shanghai, 200433, China
| | - Meng-Meng Shi
- Rui Jin Hospital, School of Medicine, Shanghai Jiaotong University, No. 197, Rui Jin Er Road, Shanghai, 200025, China
| | - Jie-Ming Qu
- Huadong Hospital Affiliated to Fudan University, No. 221, West Yan An Road, Shanghai, 200040, China.,Rui Jin Hospital, School of Medicine, Shanghai Jiaotong University, No. 197, Rui Jin Er Road, Shanghai, 200025, China
| |
Collapse
|
307
|
Özdemir BC, Siefker-Radtke AO, Campbell MT, Subudhi SK. Current and Future Applications of Novel Immunotherapies in Urological Oncology: A Critical Review of the Literature. Eur Urol Focus 2017; 4:442-454. [PMID: 29056275 DOI: 10.1016/j.euf.2017.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/03/2017] [Accepted: 10/10/2017] [Indexed: 01/05/2023]
Abstract
CONTEXT Immunotherapies promote anticancer responses with varying levels of success based on the tumor type. OBJECTIVE In this narrative review article, we searched the literature regarding immunotherapies in genitourinary malignancies to define the state of the field, explore future applications of immune checkpoint inhibitors, cytokines, vaccines, and cellular therapies in urological oncology and evaluate possible strategies to improve the selection of patients who might benefit from such approaches. EVIDENCE ACQUISITION We reviewed related literature, with a focus on recent studies about immunotherapies, predictors of response, and ongoing clinical trials. EVIDENCE SYNTHESIS Immunotherapies based on immune checkpoint blockade are approved as first- and second-line therapies for urothelial carcinoma (UC) and second-line therapies for renal cell carcinoma with limited success in prostate cancer. Programmed death-ligand 1 is the most commonly used predictive biomarker outside of UC; however, a substantial proportion of patients with tumors negative for programmed death-ligand 1 expression benefit from checkpoint inhibition, limiting its sensitivity. A high mutational load and molecular subtypes of UC are emerging as additional potential predictors. Genomic sequencing and gene expression analysis associate alterations of genes implicated in DNA repair pathways, such as BRCA1 and BRCA2, with immune checkpoint therapies. In prostate cancer, the vaccine, sipuleucel-T, is the only Food and Drug Administration-approved immunotherapy. CONCLUSIONS Immunotherapies are emerging as exciting new treatment options with a tolerable toxicity profile in urological cancers. Checkpoint inhibitors are effective only in a subset of patients, demanding personalized approaches that consider various clinical and molecular parameters to predict patient response. Clinical trials investigating the optimal timing, sequence, and combination of immunotherapies with standard of care and novel agents will guide therapy choices and improve patient outcome. PATIENT SUMMARY Clinical data supports the safety and efficacy of immune checkpoint inhibitors alone or in combination with other therapies in urological cancers.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| | | | - Matthew T Campbell
- Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Sumit K Subudhi
- Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
308
|
Jacqueline C, Brazier L, Faugère D, Renaud F, Thomas F, Roche B. Can intestinal microbiota be associated with non-intestinal cancers? Sci Rep 2017; 7:12722. [PMID: 28983086 PMCID: PMC5629204 DOI: 10.1038/s41598-017-11644-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/25/2017] [Indexed: 02/07/2023] Open
Abstract
While the role of intestinal microbiota is increasingly recognized in the etiology of digestive cancers, its effects on the development of cancer in other parts of the body have been little studied. Through new-generation sequencing, we aimed to identify an association between the structure of intestinal microbiota and the presence of eye disc tumor in Drosophila larvae. First, we observed a parental effect on the diversity and structure of bacterial communities. Second, we identified a bacterial signature (at the family level) of cancer: cancerous larvae host a significantly lower relative abundance of Bacillaceae than individuals that did not develop the tumor. Thus, for the first time, we showed that a non-digestive cancer, i.e., in the brain, could be associated with an altered composition of the gut microbial community. Finally, we discuss the potential implications of the immune system in the gut-brain axis concept to explain the long-distant effect of intestinal microbiota on brain tumors. We also highlight the potential of our results in a therapeutic perspective for brain cancer that could be generalized for other cancers.
Collapse
Affiliation(s)
- Camille Jacqueline
- CREEC, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France.
- MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France.
| | - Lionel Brazier
- MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France
| | - Dominique Faugère
- CREEC, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France
- MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France
| | - François Renaud
- CREEC, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France
- MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France
| | - Frédéric Thomas
- CREEC, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France
- MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France
| | - Benjamin Roche
- MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394, Montpellier Cedex 5, France
- International Center for Mathematical and Computational Modeling of Complex Systems (UMI IRD/UPMC UMMISCO), 32 Avenue Henri Varagnat, 93143, Bondy Cedex, France
| |
Collapse
|
309
|
Hurez V, Padrón Á, Svatek RS, Curiel TJ. Considerations for successful cancer immunotherapy in aged hosts. Exp Gerontol 2017; 107:27-36. [PMID: 28987644 DOI: 10.1016/j.exger.2017.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/30/2017] [Accepted: 10/03/2017] [Indexed: 12/22/2022]
Abstract
Improvements in understanding cancer immunopathogenesis have now led to unprecedented successes in immunotherapy to treat numerous cancers. Although aging is the most important risk factor for cancer, most pre-clinical cancer immunotherapy studies are undertaken in young hosts. This review covers age-related immune changes as they affect cancer immune surveillance, immunopathogenesis and immune therapy responses. Declining T cell function with age can impede efficacy of age-related cancer immunotherapies, but examples of successful approaches to breach this barrier have been reported. It is further recognized now that immune functions with age do not simply decline, but that they change in potentially detrimental ways. For example, detrimental immune cell populations can become predominant during aging (notably pro-inflammatory cells), the prevalence or function of suppressive cells can increase (notably myeloid derived suppressor cells), drugs can have age-specific effects on immune cells, and attributes of the aged microenvironment can impede or subvert immunity. Key advances in these and related areas will be reviewed as they pertain to cancer immunotherapy in the aged, and areas requiring additional study and some speculations on future research directions will be addressed. We prefer the term Age Related Immune Dysfunction (ARID) as most encompassing the totality of age-associated immune changes.
Collapse
Affiliation(s)
- Vincent Hurez
- Department of Medicine, University of Texas Health San Antonio, TX 78229, USA
| | - Álvaro Padrón
- Department of Medicine, University of Texas Health San Antonio, TX 78229, USA
| | - Robert S Svatek
- Department of Urology, University of Texas Health San Antonio, TX 78229, USA; The UT Health Cancer Center, University of Texas Health San Antonio, TX 78229, USA
| | - Tyler J Curiel
- Department of Medicine, University of Texas Health San Antonio, TX 78229, USA; The UT Health Cancer Center, University of Texas Health San Antonio, TX 78229, USA; Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, TX 78229, USA; The Barshop Institute for Aging and Longevity Studies, University of Texas Health San Antonio, TX 78229, USA.
| |
Collapse
|
310
|
Desired Turbulence? Gut-Lung Axis, Immunity, and Lung Cancer. JOURNAL OF ONCOLOGY 2017; 2017:5035371. [PMID: 29075294 PMCID: PMC5623803 DOI: 10.1155/2017/5035371] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/30/2017] [Accepted: 08/03/2017] [Indexed: 02/08/2023]
Abstract
The microbiota includes different microorganisms consisting of bacteria, fungi, viruses, and protozoa distributed over many human body surfaces including the skin, vagina, gut, and airways, with the highest density found in the intestine. The gut microbiota strongly influences our metabolic, endocrine, and immune systems, as well as both the peripheral and central nervous systems. Recently, a dialogue between the gut and lung microbiota has been discovered, suggesting that changes in one compartment could impact the other compartment, whether in relation to microbial composition or function. Further, this bidirectional axis is evidenced in an, either beneficial or malignant, altered immune response in one compartment following changes in the other compartment. Stimulation of the immune system arises from the microbial cells themselves, but also from their metabolites. It can be either direct or mediated by stimulated immune cells in one site impacting the other site. Additionally, this interaction may lead to immunological boost, assisting the innate immune system in its antitumour response. Thus, this review offers an insight into the composition of these sites, the gut and the lung, their role in shaping the immune system, and, finally, their role in the response to lung cancer.
Collapse
|
311
|
Pitt JM, Vétizou M, Daillère R, Roberti MP, Yamazaki T, Routy B, Lepage P, Boneca IG, Chamaillard M, Kroemer G, Zitvogel L. Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. Immunity 2017; 44:1255-69. [PMID: 27332730 DOI: 10.1016/j.immuni.2016.06.001] [Citation(s) in RCA: 786] [Impact Index Per Article: 98.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Indexed: 12/11/2022]
Abstract
Inhibition of immune regulatory checkpoints, such as CTLA-4 and the PD-1-PD-L1 axis, is at the forefront of immunotherapy for cancers of various histological types. However, such immunotherapies fail to control neoplasia in a significant proportion of patients. Here, we review how a range of cancer-cell-autonomous cues, tumor-microenvironmental factors, and host-related influences might account for the heterogeneous responses and failures often encountered during therapies using immune-checkpoint blockade. Furthermore, we describe the emerging evidence of how the strong interrelationship between the immune system and the host microbiota can determine responses to cancer therapies, and we introduce a concept by which prior or concomitant modulation of the gut microbiome could optimize therapeutic outcomes upon immune-checkpoint blockade.
Collapse
Affiliation(s)
- Jonathan M Pitt
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France
| | - Marie Vétizou
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France
| | - Romain Daillère
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France
| | - María Paula Roberti
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France
| | - Takahiro Yamazaki
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France
| | - Bertrand Routy
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France
| | - Patricia Lepage
- Micalis UMR 1319, Institut National de la Recherche Agronomique, 78360 Jouy-en-Josas, France
| | - Ivo Gomperts Boneca
- Unit of Biology and Genetics of the Bacterial Cell Wall, Institut Pasteur, 75015 Paris, France; Equipe Avenir, INSERM, 75015 Paris, France
| | - Mathias Chamaillard
- Université de Lille, Centre National de la Recherche Scientifique, INSERM, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1019, UMR 8204, Centre d'Infection et d'Immunité de Lille, 59000 Lille, France
| | - Guido Kroemer
- INSERM U848, 94800 Villejuif, France; Metabolomics Platform, Gustave Roussy Cancer Campus, 94800 Villejuif, France; Equipe 11 Labellisée Ligue contre le Cancer, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie, 75005 Paris, France; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Laurence Zitvogel
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France; Center of Clinical Investigations CICBT1428, Gustave Roussy Cancer Campus, 94805 Villejuif Cedex 05, France.
| |
Collapse
|
312
|
Ceppi F, Beck-Popovic M, Bourquin JP, Renella R. Opportunities and challenges in the immunological therapy of pediatric malignancy: a concise snapshot. Eur J Pediatr 2017; 176:1163-1172. [PMID: 28803259 DOI: 10.1007/s00431-017-2982-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/24/2017] [Accepted: 08/02/2017] [Indexed: 12/18/2022]
Abstract
Over the last 50 years, collaborative clinical trials have reduced the number of children dying from pediatric cancer significantly. Unfortunately, certain tumor types have remained resistant to conventional surgical, radiotherapy and chemotherapy combinations, and relapsing and/or refractory disease remains associated with dismal outcomes. Recently, renewed attention has been given to the role for immunotherapies in pediatric oncology. In fact, these combine several attractive features, including (but possibly not limited to) the specificity for cancer cells, potentially in vivo persistence and longevity, and potency against refractory disease. In this narrative review designed for the academic pediatrician, we will concisely review the biological underpinnings behind the immunological therapy of pediatric neoplasms and illustrate the current humoral, cellular approaches, and novel drugs targeting the immune checkpoint, oncolytic viruses, and tumor vaccines. We will also comment on the future directions, challenges, and open questions faced by the field. What is Known: • Cancer immunotherapy drives immune cells and its humoral weaponry to eliminate tumor cells. • This occurs by recognizing antigens ideally expressed only on tumoral, but not normal/healthy, cells. What is New: • Clinical immunotherapy trials have shown responses in children with relapsing/refractory neoplasms. • Novel humoral/cellular immunotherapies, immune checkpoint inhibitors, oncolytic viruses, and tumor vaccines are currently being investigated in pediatric oncology.
Collapse
Affiliation(s)
- Francesco Ceppi
- Pediatric Hematology-Oncology Research Laboratory & Pediatric Hematology-Oncology Unit, Division of Pediatrics, Department Woman-Mother-Child, University Hospital of Lausanne, Lausanne, Switzerland
| | - Maja Beck-Popovic
- Pediatric Hematology-Oncology Research Laboratory & Pediatric Hematology-Oncology Unit, Division of Pediatrics, Department Woman-Mother-Child, University Hospital of Lausanne, Lausanne, Switzerland
| | - Jean-Pierre Bourquin
- Leukemia Research Program and Division of Pediatric Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Raffaele Renella
- Pediatric Hematology-Oncology Research Laboratory & Pediatric Hematology-Oncology Unit, Division of Pediatrics, Department Woman-Mother-Child, University Hospital of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
313
|
Vanpouille-Box C, Lhuillier C, Bezu L, Aranda F, Yamazaki T, Kepp O, Fucikova J, Spisek R, Demaria S, Formenti SC, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immune checkpoint blockers for cancer therapy. Oncoimmunology 2017; 6:e1373237. [PMID: 29147629 DOI: 10.1080/2162402x.2017.1373237] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint blockers (ICBs) are literally revolutionizing the clinical management of an ever more diversified panel of oncological indications. Although considerable attention persists around the inhibition of cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1) signaling, several other co-inhibitory T-cell receptors are being evaluated as potential targets for the development of novel ICBs. Moreover, substantial efforts are being devoted to the identification of biomarkers that reliably predict the likelihood of each patient to obtain clinical benefits from ICBs in the absence of severe toxicity. Tailoring the delivery of specific ICBs or combinations thereof to selected patient populations in the context of precision medicine programs constitutes indeed a major objective of the future of ICB-based immunotherapy. Here, we discuss recent preclinical and clinical advances on the development of ICBs for oncological indications.
Collapse
Affiliation(s)
| | - Claire Lhuillier
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lucillia Bezu
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Fernando Aranda
- Immunoreceptors of the Innate and Adaptive System Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Oliver Kepp
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Université Paris Descartes/Paris V, Paris, France.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
314
|
Merlano MC, Granetto C, Fea E, Ricci V, Garrone O. Heterogeneity of colon cancer: from bench to bedside. ESMO Open 2017; 2:e000218. [PMID: 29209524 PMCID: PMC5703395 DOI: 10.1136/esmoopen-2017-000218] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 05/18/2017] [Indexed: 12/17/2022] Open
Abstract
The large bowel shows biomolecular, anatomical and bacterial changes that proceed from the proximal to the distal tract. These changes account for the different behaviour of colon cancers arising from the diverse sides of the colon-rectum as well as for the sensitivity to the therapy, including immunotherapy. The gut microbiota plays an important role in the modulation of the immune response and differs between the right colon cancer and the left colorectal cancer. The qualitative and quantitative difference of the commensal bacteria between the right side and the left side induces epigenetic changes in the intestinal epithelial cells as well as in the resident immune population. The second player in the pathological homeostasis of colorectal cancer is the differences of the genetic features of cancer cells and the different effects that microsatellite instability, chromosomal instability and the CpG island methylator phenotype induce on the immunological organisation of the tumour microenvironment. The third player is the immunological composition of the tumour microenvironment, which changes under the influence of both genetic structures and gut microbiota. All these three players influence each other. This review describes these three aspects, highlights their interactions and discusses data from reported clinical trials.
Collapse
Affiliation(s)
- Marco C Merlano
- Medical Oncology, A.O. S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Cristina Granetto
- Medical Oncology, A.O. S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Elena Fea
- Medical Oncology, A.O. S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Vincenzo Ricci
- Medical Oncology, A.O. S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Ornella Garrone
- Medical Oncology, A.O. S. Croce and Carle Teaching Hospital, Cuneo, Italy
| |
Collapse
|
315
|
Berraondo P, Minute L, Ajona D, Corrales L, Melero I, Pio R. Innate immune mediators in cancer: between defense and resistance. Immunol Rev 2017; 274:290-306. [PMID: 27782320 DOI: 10.1111/imr.12464] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic inflammation in the tumor microenvironment and evasion of the antitumor effector immune response are two of the emerging hallmarks required for oncogenesis and cancer progression. The innate immune system not only plays a critical role in perpetuating these tumor-promoting hallmarks but also in developing antitumor adaptive immune responses. Thus, understanding the dual role of the innate system in cancer immunology is required for the design of combined immunotherapy strategies able to tackle established tumors. Here, we review recent advances in the understanding of the role of cell populations and soluble components of the innate immune system in cancer, with a focus on complement, the adapter molecule Stimulator of Interferon Genes, natural killer cells, myeloid cells, and B cells.
Collapse
Affiliation(s)
- Pedro Berraondo
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Luna Minute
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Daniel Ajona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Solid Tumors and Biomarkers, CIMA, Pamplona, Spain.,Deparment of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | | | - Ignacio Melero
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Ruben Pio
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain. .,Program of Solid Tumors and Biomarkers, CIMA, Pamplona, Spain. .,Deparment of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| |
Collapse
|
316
|
Integrating Next-Generation Dendritic Cell Vaccines into the Current Cancer Immunotherapy Landscape. Trends Immunol 2017; 38:577-593. [DOI: 10.1016/j.it.2017.05.006] [Citation(s) in RCA: 222] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/03/2017] [Accepted: 05/10/2017] [Indexed: 12/22/2022]
|
317
|
Baba Y, Iwatsuki M, Yoshida N, Watanabe M, Baba H. Review of the gut microbiome and esophageal cancer: Pathogenesis and potential clinical implications. Ann Gastroenterol Surg 2017; 1:99-104. [PMID: 29863142 PMCID: PMC5881342 DOI: 10.1002/ags3.12014] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/22/2017] [Indexed: 12/17/2022] Open
Abstract
Esophageal cancer ranks among the most aggressive malignant diseases. The limited improvements in treatment outcomes provided by conventional therapies have prompted us to seek innovative strategies for treating this cancer. More than 100 trillion microorganisms inhabit the human intestinal tract and play a crucial role in health and disease conditions, including cancer. The human intestinal microbiome is thought to influence tumor development and progression in the gastrointestinal tract by various mechanisms. For example, Fusobacterium nucleatum, which primarily inhabits the oral cavity and causes periodontal disease, might contribute to aggressive tumor behavior through activation of chemokines such as CCL20 in esophageal cancer tissue. Composition of the intestinal microbiota is influenced by diet, lifestyle, antibiotics, and pro- and prebiotics. Therefore, by better understanding how the bacterial microbiota contributes to esophageal carcinogenesis, we might develop novel cancer prevention and treatment strategies through targeting the gastrointestinal microflora. This review discusses the current knowledge, available data and information on the relationship of microbiota with esophagitis, Barrett's esophagus, esophageal adenocarcinoma and squamous cell carcinoma.
Collapse
Affiliation(s)
- Yoshifumi Baba
- Department of Gastroenterological Surgery Graduate School of Medical Science Kumamoto University Kumamoto Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery Graduate School of Medical Science Kumamoto University Kumamoto Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery Graduate School of Medical Science Kumamoto University Kumamoto Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery Cancer Institute Hospital Japanese Foundation for Cancer Research Tokyo Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery Graduate School of Medical Science Kumamoto University Kumamoto Japan
| |
Collapse
|
318
|
Abstract
The human gut microbiome modulates many host processes, including metabolism, inflammation, and immune and cellular responses. It is becoming increasingly apparent that the microbiome can also influence the development of cancer. In preclinical models, the host response to cancer treatment has been improved by modulating the gut microbiome; this is known to have an altered composition in many diseases, including cancer. In addition, cancer treatment with microbial agents or their products has the potential to shrink tumours. However, the microbiome could also negatively influence cancer prognosis through the production of potentially oncogenic toxins and metabolites by bacteria. Thus, future antineoplastic treatments could combine the modulation of the microbiome and its products with immunotherapeutics and more conventional approaches that directly target malignant cells.
Collapse
|
319
|
Xu C, Ruan B, Jiang Y, Xue T, Wang Z, Lu H, Wei M, Wang S, Ye Z, Zhai D, Wang L, Lu Z. Antibiotics-induced gut microbiota dysbiosis promotes tumor initiation via affecting APC-Th1 development in mice. Biochem Biophys Res Commun 2017; 488:418-424. [PMID: 28506830 DOI: 10.1016/j.bbrc.2017.05.071] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 05/12/2017] [Indexed: 12/16/2022]
Abstract
Gut microbiota is critical for maintaining body immune homeostasis and thus affects tumor growth and therapeutic efficiency. Here, we investigated the link between microbiota and tumorgenesis in a mice model of subcutaneous melanoma cell transplantation, and explored the underlying mechanism. We found disruption of gut microbiota by pretreating mice with antibiotics promote tumor growth and remodeling the immune compartment within the primary tumor. Indeed, gut microbial dysbiosis reduced the infiltrated mature antigen-presenting cells of tumor, together with lower levels of co-stimulators, such as CD80, CD86 and MHCII, as well as defective Th1 cytokines, including IFNγ, TNFα, IL12p40, and IL12p35. Meantime, splenic APCs displayed blunted ability in triggering T cell proliferation and IFNγ secretion. However, oral administration of LPS restored the immune surveillance effects and thus inhibited tumor growth in the antibiotics induced gut microbiota dysbiosis group. Taken together, these data highly supported that antibiotics induced gut microbiota dysbiosis promotes tumor initiation, while LPS supplementation would restore the effective immune surveillance and repress tumor initiation.
Collapse
Affiliation(s)
- Chengming Xu
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an 710032, PR China
| | - Banjun Ruan
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an 710032, PR China
| | - Yinghao Jiang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an 710032, PR China
| | - Ting Xue
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an 710032, PR China
| | - Zhenyu Wang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an 710032, PR China
| | - Huanyu Lu
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Fourth Military Medical University, Xi'an 710032, PR China
| | - Ming Wei
- Department of Pharmacology, Xi'an Medical University, Xi'an 710021, PR China
| | - Shan Wang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Zicheng Ye
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an 710032, PR China
| | - Dongsheng Zhai
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an 710032, PR China
| | - Li Wang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an 710032, PR China.
| | - Zifan Lu
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
320
|
Garg AD, Vara Perez M, Schaaf M, Agostinis P, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based anticancer immunotherapy. Oncoimmunology 2017; 6:e1328341. [PMID: 28811970 DOI: 10.1080/2162402x.2017.1328341] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/05/2017] [Indexed: 12/11/2022] Open
Abstract
Dendritic cell (DC)-based vaccines against cancer have been extensively developed over the past two decades. Typically DC-based cancer immunotherapy entails loading patient-derived DCs with an appropriate source of tumor-associated antigens (TAAs) and efficient DC stimulation through a so-called "maturation cocktail" (typically a combination of pro-inflammatory cytokines and Toll-like receptor agonists), followed by DC reintroduction into patients. DC vaccines have been documented to (re)activate tumor-specific T cells in both preclinical and clinical settings. There is considerable clinical interest in combining DC-based anticancer vaccines with T cell-targeting immunotherapies. This reflects the established capacity of DC-based vaccines to generate a pool of TAA-specific effector T cells and facilitate their infiltration into the tumor bed. In this Trial Watch, we survey the latest trends in the preclinical and clinical development of DC-based anticancer therapeutics. We also highlight how the emergence of immune checkpoint blockers and adoptive T-cell transfer-based approaches has modified the clinical niche for DC-based vaccines within the wide cancer immunotherapy landscape.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Monica Vara Perez
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Marco Schaaf
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
321
|
Abstract
The microbiota is composed of commensal bacteria and other microorganisms that live on the epithelial barriers of the host. The commensal microbiota is important for the health and survival of the organism. Microbiota influences physiological functions from the maintenance of barrier homeostasis locally to the regulation of metabolism, haematopoiesis, inflammation, immunity and other functions systemically. The microbiota is also involved in the initiation, progression and dissemination of cancer both at epithelial barriers and in sterile tissues. Recently, it has become evident that microbiota, and particularly the gut microbiota, modulates the response to cancer therapy and susceptibility to toxic side effects. In this Review, we discuss the evidence for the ability of the microbiota to modulate chemotherapy, radiotherapy and immunotherapy with a focus on the microbial species involved, their mechanism of action and the possibility of targeting the microbiota to improve anticancer efficacy while preventing toxicity.
Collapse
Affiliation(s)
- Soumen Roy
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
322
|
Balar AV, Weber JS. PD-1 and PD-L1 antibodies in cancer: current status and future directions. Cancer Immunol Immunother 2017; 66:551-564. [PMID: 28213726 PMCID: PMC11028560 DOI: 10.1007/s00262-017-1954-6] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/06/2017] [Indexed: 12/20/2022]
Abstract
Immunotherapy has moved to the center stage of cancer treatment with the recent success of trials in solid tumors with PD-1/PD-L1 axis blockade. Programmed death-1 or PD-1 is a checkpoint molecule on T cells that plays a vital role in limiting adaptive immune responses and preventing autoimmune and auto-inflammatory reactivity in the normal host. In cancer patients, PD-1 expression is very high on T cells in the tumor microenvironment, and PD-L1, its primary ligand, is variably expressed on tumor cells and antigen-presenting cells within tumors, providing a potent inhibitory influence within the tumor microenvironment. While PD-L1 expression on tumors is often regarded as a negative prognostic factor, it is clearly associated with a positive outcome for treatment with PD-1/PD-L1 blocking antibodies, and has been used to select patients for this therapy. Responses of long duration, a minority of patients with atypical responses in which progression may precede tumor shrinkage, and a pattern of autoimmune side effects often seen with this class of drugs characterize therapy with PD-1/PD-L1 blocking drugs. While excellent efficacy has been seen with a limited number of tumor types, most epithelial cancers do not show responses of long duration with these agents. In the current review, we will briefly summarize the scientific background data supporting the development of PD-1/PD-L1 blockade, and then describe the track record of these antibodies in multiple different histologies ranging from melanoma and lung cancer to less common tumor types as well as discuss biomarkers that may assist in patient selection.
Collapse
Affiliation(s)
- Arjun Vasant Balar
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, 522 First Avenue, 1310 Smilow Research Building, New York, NY, 10016, USA
| | - Jeffrey S Weber
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, 522 First Avenue, 1310 Smilow Research Building, New York, NY, 10016, USA.
| |
Collapse
|
323
|
Abstract
How can we treat cancer more effectively? Traditionally, tumours from the same anatomical site are treated as one tumour entity. This concept has been challenged by recent breakthroughs in cancer genomics and translational research that have enabled molecular tumour profiling. The identification and validation of cancer drivers that are shared between different tumour types, spurred the new paradigm to target driver pathways across anatomical sites by off-label drug use, or within so-called basket or umbrella trials which are designed to test whether molecular alterations in one tumour entity can be extrapolated to all others. However, recent clinical and preclinical studies suggest that there are tissue- and cell type-specific differences in tumorigenesis and the organization of oncogenic signalling pathways. In this Opinion article, we focus on the molecular, cellular, systemic and environmental determinants of organ-specific tumorigenesis and the mechanisms of context-specific oncogenic signalling outputs. Investigation, recognition and in-depth biological understanding of these differences will be vital for the design of next-generation clinical trials and the implementation of molecularly guided cancer therapies in the future.
Collapse
Affiliation(s)
- Günter Schneider
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, 81675 München, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Marc Schmidt-Supprian
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Department of Medicine III, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, 81675 München, Germany
| | - Roland Rad
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, 81675 München, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Dieter Saur
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, 81675 München, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
324
|
Combined toll-like receptor 3/7/9 deficiency on host cells results in T-cell-dependent control of tumour growth. Nat Commun 2017; 8:14600. [PMID: 28300057 PMCID: PMC5356072 DOI: 10.1038/ncomms14600] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 01/16/2017] [Indexed: 02/07/2023] Open
Abstract
Toll-like receptors (TLRs) are located either on the cell surface or intracellularly in endosomes and their activation normally contributes to the induction of protective immune responses. However, in cancer their activation by endogenous ligands can modulate tumour progression. It is currently unknown how endosomal TLRs regulate endogenous anti-tumour immunity. Here we show that TLR3, 7 and 9 deficiencies on host cells, after initial tumour growth, result in complete tumour regression and induction of anti-tumour immunity. Tumour regression requires the combined absence of all three receptors, is dependent on both CD4 and CD8 T cells and protects the mice from subsequent tumour challenge. While tumours in control mice are infiltrated by higher numbers of regulatory T cells, tumour regression in TLR-deficient mice is paralleled by altered vascular structure and strongly induced influx of cytotoxic and cytokine-producing effector T cells. Thus, endosomal TLRs may represent a molecular link between the inflamed tumour cell phenotype, anti-tumour immunity and the regulation of T-cell activation. Activation of Toll-like receptor (TLR) is generally associated with increased immune activity. Here, the authors show, using syngeneic mouse models, that combined deficiency of TLR 3/7/9 in the host induces an inflamed tumour phenotype and results in T cell dependent tumour regression after an initial growth.
Collapse
|
325
|
Anderson KG, Stromnes IM, Greenberg PD. Obstacles Posed by the Tumor Microenvironment to T cell Activity: A Case for Synergistic Therapies. Cancer Cell 2017; 31:311-325. [PMID: 28292435 PMCID: PMC5423788 DOI: 10.1016/j.ccell.2017.02.008] [Citation(s) in RCA: 502] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 12/13/2022]
Abstract
T cell dysfunction in solid tumors results from multiple mechanisms. Altered signaling pathways in tumor cells help produce a suppressive tumor microenvironment enriched for inhibitory cells, posing a major obstacle for cancer immunity. Metabolic constraints to cell function and survival shape tumor progression and immune cell function. In the face of persistent antigen, chronic T cell receptor signaling drives T lymphocytes to a functionally exhausted state. Here we discuss how the tumor and its microenvironment influences T cell trafficking and function with a focus on melanoma, and pancreatic and ovarian cancer, and discuss how scientific advances may help overcome these hurdles.
Collapse
Affiliation(s)
- Kristin G Anderson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Mail Stop D3-100, P.O. Box 19024, Seattle, WA 98109, USA; Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Ingunn M Stromnes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Mail Stop D3-100, P.O. Box 19024, Seattle, WA 98109, USA; Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Philip D Greenberg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Mail Stop D3-100, P.O. Box 19024, Seattle, WA 98109, USA; Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
326
|
Özdemir BC, Dotto GP. Racial Differences in Cancer Susceptibility and Survival: More Than the Color of the Skin? Trends Cancer 2017; 3:181-197. [PMID: 28718431 DOI: 10.1016/j.trecan.2017.02.002] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022]
Abstract
Epidemiological studies point to race as a determining factor in cancer susceptibility. In US registries recording cancer incidence and survival by race (distinguishing 'black versus white'), individuals of African ancestry have a globally increased risk of malignancies compared with Caucasians and Asian Americans. Differences in socioeconomic status and health-care access play a key role. However, the lesser disease susceptibility of Hispanic populations with comparable lifestyles and socioeconomic status as African Americans (Hispanic paradox) points to the concomitant importance of genetic determinants. Here, we overview the molecular basis of racial disparity in cancer susceptibility ranging from genetic polymorphisms and cancer-driver gene mutations to obesity, chronic inflammation, and immune responses. We discuss implications for race-adapted cancer screening programs and clinical trials to reduce disparities in cancer burden.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | - Gian-Paolo Dotto
- Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, 1066 Épalinges, Switzerland; Harvard Dermatology Department and Cutaneous Biology Research Center, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02129, USA.
| |
Collapse
|
327
|
Tsilimigras MCB, Fodor A, Jobin C. Carcinogenesis and therapeutics: the microbiota perspective. Nat Microbiol 2017; 2:17008. [PMID: 28225000 PMCID: PMC6423540 DOI: 10.1038/nmicrobiol.2017.8] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/10/2017] [Indexed: 12/18/2022]
Abstract
Cancer arises from the acquisition of multiple genetic and epigenetic changes in host cells over the span of many years, promoting oncogenic traits and carcinogenesis. Most cancers develop following random somatic alterations of key oncogenic genes, which are favoured by a number of risk factors, including lifestyle, diet and inflammation. Importantly, the environment where tumours evolve provides a unique source of signalling cues that affects cancer cell growth, survival, movement and metastasis. Recently, there has been increased interest in how the microbiota, the collection of microorganisms inhabiting the host body surface and cavities, shapes a micro-environment for host cells that can either promote or prevent cancer formation. The microbiota, particularly the intestinal biota, plays a central role in host physiology, and the composition and activity of this consortium of microorganisms is directly influenced by known cancer risk factors such as lifestyle, diet and inflammation. In this REVIEW, we discuss the pro- and anticarcinogenic role of the microbiota, as well as highlighting the therapeutic potential of microorganisms in tumourigenesis. The broad impacts, and, at times, opposing roles of the microbiota in carcinogenesis serve to illustrate the complex and sometimes conflicted relationship between microorganisms and the host-a relationship that could potentially be harnessed for therapeutic benefits.
Collapse
Affiliation(s)
- Matthew C. B. Tsilimigras
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, USA
| | - Anthony Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, USA
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, Florida 32611, USA
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, Florida 32611, USA
| |
Collapse
|
328
|
Chen KL, Jung P, Kulkoyluoglu-Cotul E, Liguori C, Lumibao J, Mazewski C, Ranard K, Rowles JL, Wang Y, Xue L, Madak-Erdogan Z. Impact of Diet and Nutrition on Cancer Hallmarks. ACTA ACUST UNITED AC 2017; 7. [PMID: 30581989 DOI: 10.15406/jcpcr.2017.07.00240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diet and nutrition are undeniably two factors that have a major impact on the prevention, progression, and treatment of various cancers. In this review, we will discuss how bioactives from diet and nutritional status affect each of the hallmarks of cancer. We will present recent research and discuss using diet and nutrition as a means to prevent and treat cancer.
Collapse
Affiliation(s)
- Karen L Chen
- Division of Nutritional Sciences, University of Illinois, USA
| | - Paul Jung
- Department of Food Science and Human Nutrition, University of Illinois, USA
| | | | - Carli Liguori
- Department of Food Science and Human Nutrition, University of Illinois, USA
| | - Jan Lumibao
- Division of Nutritional Sciences, University of Illinois, USA
| | - Candice Mazewski
- Department of Food Science and Human Nutrition, University of Illinois, USA
| | | | - Joe L Rowles
- Division of Nutritional Sciences, University of Illinois, USA
| | - Yanling Wang
- Department of Food Science and Human Nutrition, University of Illinois, USA
| | - Louisa Xue
- Division of Nutritional Sciences, University of Illinois, USA
| | - Zeynep Madak-Erdogan
- Division of Nutritional Sciences, University of Illinois, USA.,Department of Food Science and Human Nutrition, University of Illinois, USA
| |
Collapse
|
329
|
Tosolini M, Pont F, Poupot M, Vergez F, Nicolau-Travers ML, Vermijlen D, Sarry JE, Dieli F, Fournié JJ. Assessment of tumor-infiltrating TCRV γ9V δ2 γδ lymphocyte abundance by deconvolution of human cancers microarrays. Oncoimmunology 2017; 6:e1284723. [PMID: 28405516 DOI: 10.1080/2162402x.2017.1284723] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 10/20/2022] Open
Abstract
Most human blood γδ cells are cytolytic TCRVγ9Vδ2+ lymphocytes with antitumor activity. They are currently investigated in several clinical trials of cancer immunotherapy but so far, their tumor infiltration has not been systematically explored across human cancers. Novel algorithms allowing the deconvolution of bulk tumor transcriptomes to find the relative proportions of infiltrating leucocytes, such as CIBERSORT, should be appropriate for this aim but in practice they fail to accurately recognize γδ T lymphocytes. Here, by implementing machine learning from microarray data, we first improved the computational identification of blood-derived TCRVγ9Vδ2+ γδ lymphocytes and then applied this strategy to assess their abundance as tumor infiltrating lymphocytes (γδ TIL) in ∼10,000 cancer biopsies from 50 types of hematological and solid malignancies. We observed considerable inter-individual variation of TCRVγ9Vδ2+γδ TIL abundance both within each type and across the spectrum of cancers tested. We report their prominence in B cell-acute lymphoblastic leukemia (B-ALL), acute promyelocytic leukemia (M3-AML) and chronic myeloid leukemia (CML) as well as in inflammatory breast, prostate, esophagus, pancreas and lung carcinoma. Across all cancers, the abundance of αβ TILs and TCRVγ9Vδ2+ γδ TILs did not correlate. αβ TIL abundance paralleled the mutational load of tumors and positively correlated with inflammation, infiltration of monocytes, macrophages and dendritic cells (DC), antigen processing and presentation, and cytolytic activity, in line with an association with a favorable outcome. In contrast, the abundance of TCRVγ9Vδ2+ γδ TILs did not correlate with these hallmarks and was variably associated with outcome, suggesting that distinct contexts underlie TCRVγ9Vδ2+ γδ TIL and αβ TIL mobilizations in cancer.
Collapse
Affiliation(s)
- Marie Tosolini
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Laboratoire d'Excellence TOUCAN, Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France; Pôle Technologique du Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; Institut Universitaire du Cancer de Toulouse (IUCT), Toulouse, France
| | - Frédéric Pont
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Pôle Technologique du Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
| | - Mary Poupot
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Laboratoire d'Excellence TOUCAN, Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| | - François Vergez
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Institut Universitaire du Cancer de Toulouse (IUCT), Toulouse, France
| | | | - David Vermijlen
- Central Laboratory for Advanced Diagnostics and Biomedical Research (CLADIBIOR), University of Palermo , Palermo, Italy
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| | - Francesco Dieli
- Department of Biopharmacy - Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Bruxelles, Belgium
| | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Laboratoire d'Excellence TOUCAN, Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| |
Collapse
|
330
|
Governa V, Trella E, Mele V, Tornillo L, Amicarella F, Cremonesi E, Muraro MG, Xu H, Droeser R, Däster SR, Bolli M, Rosso R, Oertli D, Eppenberger-Castori S, Terracciano LM, Iezzi G, Spagnoli GC. The Interplay Between Neutrophils and CD8 + T Cells Improves Survival in Human Colorectal Cancer. Clin Cancer Res 2017; 23:3847-3858. [PMID: 28108544 DOI: 10.1158/1078-0432.ccr-16-2047] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 12/14/2016] [Accepted: 12/21/2016] [Indexed: 01/11/2023]
Abstract
Purpose: Tumor infiltration by different T lymphocyte subsets is known to be associated with favorable prognosis in colorectal cancer. Still debated is the role of innate immune system. We investigated clinical relevance, phenotypes, and functional features of colorectal cancer-infiltrating CD66b+ neutrophils and their crosstalk with CD8+ T cells.Experimental Design: CD66b+ and CD8+ cell infiltration was analyzed by IHC on a tissue microarray including >650 evaluable colorectal cancer samples. Phenotypic profiles of tissue-infiltrating and peripheral blood CD66b+ cells were evaluated by flow cytometry. CD66b+/CD8+ cells crosstalk was investigated by in vitro experiments.Results: CD66b+ cell infiltration in colorectal cancer is significantly associated with increased survival. Interestingly, neutrophils frequently colocalize with CD8+ T cells in colorectal cancer. Functional studies indicate that although neutrophils are devoid of direct antitumor potential, coculture with peripheral blood or tumor-associated neutrophils (TAN) enhances CD8+ T-cell activation, proliferation, and cytokine release induced by suboptimal concentrations of anti-CD3 mAb. Moreover, under optimal activation conditions, CD8+ cell stimulation in the presence of CD66b+ cells results in increasing numbers of cells expressing CD45RO/CD62L "central memory" phenotype. Importantly, combined tumor infiltration by CD66b+ and CD8+ T lymphocytes is associated with significantly better prognosis, as compared with CD8+ T-cell infiltration alone.Conclusions: Neutrophils enhance the responsiveness of CD8+ T cells to T-cell receptor triggering. Accordingly, infiltration by neutrophils enhances the prognostic significance of colorectal cancer infiltration by CD8+ T cells, suggesting that they might effectively promote antitumor immunity. Clin Cancer Res; 23(14); 3847-58. ©2017 AACR.
Collapse
Affiliation(s)
- Valeria Governa
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Molecular Pathology Division, Institute of Pathology, Basel University Hospital and University of Basel, Switzerland
| | - Emanuele Trella
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Valentina Mele
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Luigi Tornillo
- Molecular Pathology Division, Institute of Pathology, Basel University Hospital and University of Basel, Switzerland
| | - Francesca Amicarella
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Eleonora Cremonesi
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Manuele Giuseppe Muraro
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Hui Xu
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Raoul Droeser
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of General Surgery, Basel University Hospital and University of Basel, Switzerland
| | - Silvio R Däster
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of General Surgery, Basel University Hospital and University of Basel, Switzerland
| | - Martin Bolli
- Department of Visceral Surgery, St. Claraspital, Basel, Switzerland
| | - Raffaele Rosso
- Department of Visceral Surgery, Ospedale Civico, Lugano, Switzerland
| | - Daniel Oertli
- Department of General Surgery, Basel University Hospital and University of Basel, Switzerland
| | - Serenella Eppenberger-Castori
- Molecular Pathology Division, Institute of Pathology, Basel University Hospital and University of Basel, Switzerland
| | - Luigi M Terracciano
- Molecular Pathology Division, Institute of Pathology, Basel University Hospital and University of Basel, Switzerland
| | - Giandomenica Iezzi
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Giulio C Spagnoli
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.
| |
Collapse
|
331
|
How to Escape the Immune Response: What Tumors Teach to Transplant Physicians/Immunologists. Transplantation 2017; 101:2825-2829. [PMID: 28072758 DOI: 10.1097/tp.0000000000001639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Recent progress in deciphering the mechanisms underlying the concepts of tumor immunosurveillance and immunoevasion has opened new opportunities for the development of effective antitumor therapies. Transplant physicians and immunologists have much to learn from those direct clinical translations of basic science. The 2016 Beaune Seminar in Transplant research brought together researchers from both fields to explore and discuss significant advances in cancer biology, immunotherapies and their potential impacts for the management of cancer in transplant recipients.
Collapse
|
332
|
Pagès F, Bay JO, Tartour E. L’immunothérapie des cancers : de l’espoir à la réalité. Bull Cancer 2017; 103 Suppl 1:S119-S121. [PMID: 28057173 DOI: 10.1016/s0007-4551(16)30367-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Franck Pagès
- Service d'immunologie biologique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris; INSERM UMRS1138 équipe 15, université Paris-Descartes et université Pierre-et-Marie-Curie, Centre de recherche des cordeliers 15, rue de l'école de médecine 75006 Paris.
| | - Jacques-Olivier Bay
- Service de thérapie cellulaire et d'hématologie clinique adulte, CHU Clermont-Ferrand, 1, place Lucie-et-Raymond-Aubrac, 63003 Clermont-Ferrand Cedex 01, France
| | - Eric Tartour
- Service d'immunologie biologique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris; Inserm U970, Paris Cardiovascular Research Center - PARCC, 56, rue Leblanc, 75015 Paris, France ; UMR-S970, université Paris-Descartes, Sorbonne-Paris-Cité, Paris, France ; équipe labellisée ligue contre le cancer, 56, rue Leblanc, 75015 Paris, France
| |
Collapse
|
333
|
Routy B, Letendre C, Enot D, Chénard-Poirier M, Mehraj V, Séguin NC, Guenda K, Gagnon K, Woerther PL, Ghez D, Lachance S. The influence of gut-decontamination prophylactic antibiotics on acute graft-versus-host disease and survival following allogeneic hematopoietic stem cell transplantation. Oncoimmunology 2016; 6:e1258506. [PMID: 28197380 DOI: 10.1080/2162402x.2016.1258506] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 02/08/2023] Open
Abstract
The intestinal microbiota plays a key role in the pathogenesis of acute graft-versus-host disease (aGVHD). High-dose conditioning regimens given prior to allogeneic hematopoietic stem cell transplantation (aHSCT) modulate the composition of gut microbiota and damage the gut epithelial barrier, resulting in increased systemic inflammation. We assessed whether gut decontamination with antibiotics (ATB) prior to aHSCT influenced the frequency of aGVHD and mortality in 500 patients from two Canadian centers between 2005 and 2012. The rate of grade II-IV aGVHD was higher in the ATB arm compared with the arm without ATB (42% vs 28%; p < 0.001). This difference was mainly driven by a 2-fold higher rate of grade II-IV gastrointestinal aGVHD (GI-GVHD) in the ATB arm compared with the arm without ATB (20.7% vs 10.8%; p = 0.003). Multivariate analyses adjusted for known aGVHD risk factors revealed that more patients in the ATB group developed clinically significant GI-GVHD and liver aGVHD; adjusted odds ratio (aOR) = 1.83; p = 0.023 and aOR = 3.56; p = 0.047, respectively. Importantly, median overall survival (OS) was significantly lower in the group receiving ATB and the OS at 10 y remained decreased in the ATB group; adjusted hazard ratio (aHR) = 1.61 (p < 0.001). Without undermining the role of ATB prophylaxis to prevent infection in aHSCT, we have shown that the use of ATB that targets intestinal bacteria is associated with a more severe aGVHD that involves the GI organs and impacts OS. Prospective studies that evaluate the contribution of bacterial decontamination to aGVHD are warranted.
Collapse
Affiliation(s)
- Bertrand Routy
- Gustave Roussy Comprehensive Cancer Center, Villejuif, France; Department of Hematology and Stem Cell Transplant Program, Hôpital Maisonneuve-Rosemont, University of Montreal, Montreal, QC, Canada
| | - Caroline Letendre
- Department of Hematology and Stem Cell Transplant Program, Hôpital Maisonneuve-Rosemont, University of Montreal , Montreal, QC, Canada
| | - David Enot
- Gustave Roussy Comprehensive Cancer Center , Villejuif, France
| | - Maxime Chénard-Poirier
- Department of Medicine, Division of Hematology and Oncology, Centre Hospitalier Universitaire de Québec , QC, Canada
| | - Vikram Mehraj
- Research Institute, McGill University Health Centre , Montreal, QC, Canada
| | - Noémie Charbonneau Séguin
- Department of Hematology and Stem Cell Transplant Program, Hôpital Maisonneuve-Rosemont, University of Montreal , Montreal, QC, Canada
| | - Khaled Guenda
- Department of Hematology and Stem Cell Transplant Program, Hôpital Maisonneuve-Rosemont, University of Montreal , Montreal, QC, Canada
| | - Kathia Gagnon
- Department of Hematology and Stem Cell Transplant Program, Hôpital Maisonneuve-Rosemont, University of Montreal , Montreal, QC, Canada
| | - Paul-Louis Woerther
- Gustave Roussy Comprehensive Cancer Center, Villejuif, France; Laboratory of Medical Microbiology, GRCC, Villejuif, France
| | - David Ghez
- Gustave Roussy Comprehensive Cancer Center , Villejuif, France
| | - Silvy Lachance
- Department of Hematology and Stem Cell Transplant Program, Hôpital Maisonneuve-Rosemont, University of Montreal , Montreal, QC, Canada
| |
Collapse
|
334
|
Murphy WJ. Being "penny-wise but pound foolish" in cancer immunotherapy research: the urgent need for mouse cancer models to reflect human modifying factors. J Immunother Cancer 2016; 4:88. [PMID: 28031818 PMCID: PMC5170894 DOI: 10.1186/s40425-016-0195-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/17/2016] [Indexed: 11/30/2022] Open
Abstract
Inbred mice are the mainstay for preclinical cancer assessment of potential therapeutics, especially immune-based approaches. However, the use of young, lean, inbred mice housed under specific-pathogen-free conditions does not mirror the human cancer scenario. This commentary discusses some of the issues in evaluating immunotherapeutics in mice given recent advances.
Collapse
Affiliation(s)
- William J Murphy
- Departments of Dermatology and Internal Medicine, UC Davis School of Medicine, Sacramento, CA USA
| |
Collapse
|
335
|
Beckmann JS, Lew D. Reconciling evidence-based medicine and precision medicine in the era of big data: challenges and opportunities. Genome Med 2016; 8:134. [PMID: 27993174 PMCID: PMC5165712 DOI: 10.1186/s13073-016-0388-7] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This era of groundbreaking scientific developments in high-resolution, high-throughput technologies is allowing the cost-effective collection and analysis of huge, disparate datasets on individual health. Proper data mining and translation of the vast datasets into clinically actionable knowledge will require the application of clinical bioinformatics. These developments have triggered multiple national initiatives in precision medicine—a data-driven approach centering on the individual. However, clinical implementation of precision medicine poses numerous challenges. Foremost, precision medicine needs to be contrasted with the powerful and widely used practice of evidence-based medicine, which is informed by meta-analyses or group-centered studies from which mean recommendations are derived. This “one size fits all” approach can provide inadequate solutions for outliers. Such outliers, which are far from an oddity as all of us fall into this category for some traits, can be better managed using precision medicine. Here, we argue that it is necessary and possible to bridge between precision medicine and evidence-based medicine. This will require worldwide and responsible data sharing, as well as regularly updated training programs. We also discuss the challenges and opportunities for achieving clinical utility in precision medicine. We project that, through collection, analyses and sharing of standardized medically relevant data globally, evidence-based precision medicine will shift progressively from therapy to prevention, thus leading eventually to improved, clinician-to-patient communication, citizen-centered healthcare and sustained well-being.
Collapse
Affiliation(s)
- Jacques S Beckmann
- Clinical Bioinformatics, SIB Swiss Institute of Bioinformatics, CH-1015, Lausanne, Switzerland.
| | - Daniel Lew
- Clinical Bioinformatics, SIB Swiss Institute of Bioinformatics, CH-1015, Lausanne, Switzerland
| |
Collapse
|
336
|
Abstract
Fundamental cancer research and the development of efficacious antineoplastic treatments both rely on experimental systems in which the relationship between malignant cells and immune cells can be studied. Mouse models of transplantable, carcinogen-induced or genetically engineered malignancies - each with their specific advantages and difficulties - have laid the foundations of oncoimmunology. These models have guided the immunosurveillance theory that postulates that evasion from immune control is an essential feature of cancer, the concept that the long-term effects of conventional cancer treatments mostly rely on the reinstatement of anticancer immune responses and the preclinical development of immunotherapies, including currently approved immune checkpoint blockers. Specific aspects of pharmacological development, as well as attempts to personalize cancer treatments using patient-derived xenografts, require the development of mouse models in which murine genes and cells are replaced with their human equivalents. Such 'humanized' mouse models are being progressively refined to characterize the leukocyte subpopulations that belong to the innate and acquired arms of the immune system as they infiltrate human cancers that are subjected to experimental therapies. We surmise that the ever-advancing refinement of murine preclinical models will accelerate the pace of therapeutic optimization in patients.
Collapse
Affiliation(s)
- Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), INSERM U1015, 114 rue Edouard Vaillant, 94805 Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer, CICBT1428, GRCC, 94805 Villejuif, France
| | - Jonathan M Pitt
- Gustave Roussy Cancer Campus (GRCC), INSERM U1015, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | - Romain Daillère
- Gustave Roussy Cancer Campus (GRCC), INSERM U1015, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; University of Queensland, Herston, QLD, Australia
| | - Guido Kroemer
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France
- University of Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
- University of Pierre et Marie Curie, 75006 Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, 75015 Paris, France
- Metabolomics and Cell Biology Platforms, GRCC, 94805 Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| |
Collapse
|
337
|
Abstract
Combinations of therapies are being actively pursued to expand therapeutic options and deal with cancer’s pervasive resistance to treatment. Research efforts to discover effective combination treatments have focused on drugs targeting intracellular processes of the cancer cells and in particular on small molecules that target aberrant kinases. Accordingly, most of the computational methods used to study, predict, and develop drug combinations concentrate on these modes of action and signaling processes within the cancer cell. This focus on the cancer cell overlooks significant opportunities to tackle other components of tumor biology that may offer greater potential for improving patient survival. Many alternative strategies have been developed to combat cancer; for example, targeting different cancer cellular processes such as epigenetic control; modulating stromal cells that interact with the tumor; strengthening physical barriers that confine tumor growth; boosting the immune system to attack tumor cells; and even regulating the microbiome to support antitumor responses. We suggest that to fully exploit these treatment modalities using effective drug combinations it is necessary to develop multiscale computational approaches that take into account the full complexity underlying the biology of a tumor, its microenvironment, and a patient’s response to the drugs. In this Opinion article, we discuss preliminary work in this area and the needs—in terms of both computational and data requirements—that will truly empower such combinations.
Collapse
Affiliation(s)
- Jonathan R Dry
- Oncology Innovative Medicines and Early Development, AstraZeneca, R&D Boston, Waltham, MA, 02451, USA.
| | - Mi Yang
- Rheinisch-Westfälische Technische Hochschule Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine, Aachen, 52057, Germany
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, CB10 1SD, UK. .,Rheinisch-Westfälische Technische Hochschule Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine, Aachen, 52057, Germany.
| |
Collapse
|
338
|
Stevens WBC, Netea MG, Kater AP, van der Velden WJFM. 'Trained immunity': consequences for lymphoid malignancies. Haematologica 2016; 101:1460-1468. [PMID: 27903713 DOI: 10.3324/haematol.2016.149252] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 06/29/2016] [Indexed: 12/15/2022] Open
Abstract
In hematological malignancies complex interactions exist between the immune system, microorganisms and malignant cells. On one hand, microorganisms can induce cancer, as illustrated by specific infection-induced lymphoproliferative diseases such as Helicobacter pylori-associated gastric mucosa-associated lymphoid tissue lymphoma. On the other hand, malignant cells create an immunosuppressive environment for their own benefit, but this also results in an increased risk of infections. Disrupted innate immunity contributes to the neoplastic transformation of blood cells by several mechanisms, including the uncontrolled clearance of microbial and autoantigens resulting in chronic immune stimulation and proliferation, chronic inflammation, and defective immune surveillance and anti-cancer immunity. Restoring dysfunction or enhancing responsiveness of the innate immune system might therefore represent a new angle for the prevention and treatment of hematological malignancies, in particular lymphoid malignancies and associated infections. Recently, it has been shown that cells of the innate immune system, such as monocytes/macrophages and natural killer cells, harbor features of immunological memory and display enhanced functionality long-term after stimulation with certain microorganisms and vaccines. These functional changes rely on epigenetic reprogramming and have been termed 'trained immunity'. In this review the concept of 'trained immunity' is discussed in the setting of lymphoid malignancies. Amelioration of infectious complications and hematological disease progression can be envisioned to result from the induction of trained immunity, but future studies are required to prove this exciting new hypothesis.
Collapse
Affiliation(s)
- Wendy B C Stevens
- Department of Hematology, Radboud University Medical Centre, Nijmegen
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Centre, and Radboud Center for Infectious Diseases, Nijmegen.,Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen
| | - Arnon P Kater
- Department of Hematology, Lymphoma and Myeloma Center Amsterdam (LYMMCARE) Academic Medical Center, University of Amsterdam, The Netherlands
| | - Walter J F M van der Velden
- Department of Hematology, Radboud University Medical Centre, Nijmegen .,Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen
| |
Collapse
|
339
|
Hurez V, Padrón ÁS, Svatek RS, Curiel TJ. Considerations for successful cancer immunotherapy in aged hosts. Clin Exp Immunol 2016; 187:53-63. [PMID: 27690272 DOI: 10.1111/cei.12875] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2016] [Indexed: 12/22/2022] Open
Abstract
Immunotherapy is now experiencing unprecedented successes in treating various cancers based on new understandings of cancer immunopathogenesis. Nonetheless, although ageing is the biggest risk factor for cancer, the majority of cancer immunotherapy preclinical studies are conducted in young hosts. This review will explore age-related changes in immunity as they relate to cancer immune surveillance, immunopathogenesis and responses to immunotherapy. Although it is recognized that declining T cell function with age poses a great challenge to developing effective age-related cancer immunotherapies, examples of successful approaches to overcome this hurdle have been developed. Further, it is now recognized that immune functions do not simply decline with age, but rather change in ways than can be detrimental. For example, with age, specific immune cell populations with detrimental functions can become predominant (such as cells producing proinflammatory cytokines), suppressive cells can become more numerous or more suppressive (such as myeloid-derived suppressor cells), drugs can affect aged immune cells distinctly and the aged microenvironment is becoming recognized as a significant barrier to address. Key developments in these and other areas will be surveyed as they relate to cancer immunotherapy in aged hosts, and areas in need of more study will be assessed with some speculations for the future. We propose the term 'age-related immune dysfunction' (ARID) as best representative of age-associated changes in immunity.
Collapse
Affiliation(s)
- V Hurez
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Á S Padrón
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - R S Svatek
- Department of Urology, University of Texas Health Science Center, San Antonio, TX, USA.,Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, TX, USA
| | - T J Curiel
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.,Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, TX, USA.,Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX, USA.,The Barshop Institute for Ageing and Longevity Studies, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
340
|
Routy JP, Routy B, Graziani GM, Mehraj V. The Kynurenine Pathway Is a Double-Edged Sword in Immune-Privileged Sites and in Cancer: Implications for Immunotherapy. Int J Tryptophan Res 2016; 9:67-77. [PMID: 27773992 PMCID: PMC5063567 DOI: 10.4137/ijtr.s38355] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 09/18/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022] Open
Abstract
The term “immune privilege” was originally coined to describe the suppression of inflammatory responses within organs protected by anatomic barriers, ie, the eyes, brain, placenta, and testes. However, cellular and metabolic processes, which orchestrate immune responses, also control inflammation within these sites. Our current understanding of tolerogenic mechanisms has extended the definition of immune privilege to include hair follicles, the colon, and cancer. By catabolizing tryptophan, cells expressing the enzyme indoleamine-2,3-dioxygenase produce kynurenine metabolites, which orchestrate local and systemic responses to control inflammation, thus maintaining immune privilege. This review highlights the double-edged role played by the kynurenine pathway (KP), which establishes and maintains immune-privileged sites while contributing to cancer immune escape. The identification of the underlying molecular drivers of the KP in immune-privileged sites and in cancer is essential for the development of novel therapies to treat autoimmunity and cancer and to improve transplantation outcomes.
Collapse
Affiliation(s)
- Jean-Pierre Routy
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada.; The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.; Professor of Medicine, Division of Hematology, McGill University Health Centre, Montreal, QC, Canada.; Louis Lowenstein Chair in Hematology and Oncology, McGill University, Montreal, QC, Canada
| | - Bertrand Routy
- Postdoctoral Fellow, Gustave Roussy Cancer Campus, Villejuif, France.; INSERM U1015, Villejuif, France
| | - Gina M Graziani
- Research Associate, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Vikram Mehraj
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada.; Postdoctoral Fellow, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
341
|
Semeraro M, Adam J, Stoll G, Louvet E, Chaba K, Poirier-Colame V, Sauvat A, Senovilla L, Vacchelli E, Bloy N, Humeau J, Buque A, Kepp O, Zitvogel L, André F, Mathieu MC, Delaloge S, Kroemer G. The ratio of CD8 +/FOXP3 T lymphocytes infiltrating breast tissues predicts the relapse of ductal carcinoma in situ. Oncoimmunology 2016; 5:e1218106. [PMID: 27853639 DOI: 10.1080/2162402x.2016.1218106] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 07/20/2016] [Accepted: 07/22/2016] [Indexed: 01/21/2023] Open
Abstract
In a series of 248 tumor samples obtained from image-guided biopsies from patients diagnosed with ductal carcinoma in situ of the breast, we attempted to identify biomarkers that predict microinfiltration at definitive surgery or relapse during follow-up. For this, we used immunohistochemical methods, followed by automated image analyses, to measure the mean diameter of nuclei (which correlates with ploidy), the phosphorylation of eukaryotic initiation factor 2α (eIF2α, which reflects endoplasmic reticulum stress) as well as the density and ratio of CD8+ cytotoxic T lymphocytes and FOXP3+ regulatory T cells. The median nuclear diameter of malignant cells correlated with eIF2α phosphorylation (in cancerous tissue), which in turn correlated with the density of the CD8+ infiltrate and the CD8+/FOXP3 ratio (both in cancerous and the adjacent non-cancerous parenchyma). Neither microinfiltration nor lymph node involvement was associated with the probability of relapse. Both correlated positively with the CD8+/FOXP3 ratio in the malignant area. In contrast, relapse was associated with a paucity of the CD8+ infiltrate as well as an unfavorable CD8+/FOXP3 ratio, both in malignant and non-malignant parenchyma. The combined analysis of the CD8+/FOXP3 ratio in cancerous and non-cancerous tissues revealed a significant impact of their interaction on the probability of relapse, but not on the presence of microinfiltration or lymph node metastasis. Altogether, these results support the idea of an immunosurveillance system that determines the risk of relapse in ductal carcinoma in situ of the breast.
Collapse
Affiliation(s)
- Michaela Semeraro
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Julien Adam
- Department of Medical Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif, France; Module de Développement en Pathologie Expérimentale-INSERM U981, Gustave-Roussy Cancer Campus, Villejuif, France
| | - Gautier Stoll
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Emilie Louvet
- Module de Développement en Pathologie Expérimentale-INSERM U981, Gustave-Roussy Cancer Campus , Villejuif, France
| | - Kariman Chaba
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Vichnou Poirier-Colame
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France; Department of Immuno-Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Allan Sauvat
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Laura Senovilla
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Erika Vacchelli
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Norma Bloy
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France; Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Juliette Humeau
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France; Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Aitziber Buque
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Oliver Kepp
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Laurence Zitvogel
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France; Department of Immuno-Oncology, Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1015, Villejuif, France
| | - Fabrice André
- INSERM, U981 "Identification of molecular predictors and new targets for cancer treatment", Villejuif, France; Department of Medical Oncology and Breast Cancer Group, Gustave Roussy Cancer Campus, Villejuif, France
| | - Marie-Christine Mathieu
- Department of Medical Biology and Pathology, Gustave Roussy Cancer Campus , Villejuif, France
| | - Suzette Delaloge
- Department of Medical Oncology and Breast Cancer Group, Gustave Roussy Cancer Campus , Villejuif, France
| | - Guido Kroemer
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
342
|
Abstract
Precision medicine relies on validated biomarkers with which to better classify patients by their probable disease risk, prognosis and/or response to treatment. Although affordable 'omics'-based technology has enabled faster identification of putative biomarkers, the validation of biomarkers is still stymied by low statistical power and poor reproducibility of results. This Review summarizes the successes and challenges of using different types of molecule as biomarkers, using lung cancer as a key illustrative example. Efforts at the national level of several countries to tie molecular measurement of samples to patient data via electronic medical records are the future of precision medicine research.
Collapse
Affiliation(s)
- Ashley J Vargas
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Room 3068A, MSC 425, 837 Convent Drive, Bethesda, Maryland 20892-4258, USA
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland 20850, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Room 3068A, MSC 425, 837 Convent Drive, Bethesda, Maryland 20892-4258, USA
| |
Collapse
|