6251
|
Abstract
Immune checkpoint-blocking therapies have yielded positive clinical data in a series of human malignancies. Recent work from Le and colleagues strongly supports the use of these therapies for mismatch repair-deficient tumors, independent of underlying tumor type. These data suggest the importance of sensing the consequences of DNA damage in cancer immunotherapy.
Collapse
Affiliation(s)
- Sander Kelderman
- Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Ton N Schumacher
- Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Pia Kvistborg
- Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
6252
|
Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WEE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, Waterhouse D, Ready N, Gainor J, Arén Frontera O, Havel L, Steins M, Garassino MC, Aerts JG, Domine M, Paz-Ares L, Reck M, Baudelet C, Harbison CT, Lestini B, Spigel DR. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med 2015; 373:123-35. [PMID: 26028407 PMCID: PMC4681400 DOI: 10.1056/nejmoa1504627] [Citation(s) in RCA: 6709] [Impact Index Per Article: 670.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Patients with advanced squamous-cell non-small-cell lung cancer (NSCLC) who have disease progression during or after first-line chemotherapy have limited treatment options. This randomized, open-label, international, phase 3 study evaluated the efficacy and safety of nivolumab, a fully human IgG4 programmed death 1 (PD-1) immune-checkpoint-inhibitor antibody, as compared with docetaxel in this patient population. METHODS We randomly assigned 272 patients to receive nivolumab, at a dose of 3 mg per kilogram of body weight every 2 weeks, or docetaxel, at a dose of 75 mg per square meter of body-surface area every 3 weeks. The primary end point was overall survival. RESULTS The median overall survival was 9.2 months (95% confidence interval [CI], 7.3 to 13.3) with nivolumab versus 6.0 months (95% CI, 5.1 to 7.3) with docetaxel. The risk of death was 41% lower with nivolumab than with docetaxel (hazard ratio, 0.59; 95% CI, 0.44 to 0.79; P<0.001). At 1 year, the overall survival rate was 42% (95% CI, 34 to 50) with nivolumab versus 24% (95% CI, 17 to 31) with docetaxel. The response rate was 20% with nivolumab versus 9% with docetaxel (P=0.008). The median progression-free survival was 3.5 months with nivolumab versus 2.8 months with docetaxel (hazard ratio for death or disease progression, 0.62; 95% CI, 0.47 to 0.81; P<0.001). The expression of the PD-1 ligand (PD-L1) was neither prognostic nor predictive of benefit. Treatment-related adverse events of grade 3 or 4 were reported in 7% of the patients in the nivolumab group as compared with 55% of those in the docetaxel group. CONCLUSIONS Among patients with advanced, previously treated squamous-cell NSCLC, overall survival, response rate, and progression-free survival were significantly better with nivolumab than with docetaxel, regardless of PD-L1 expression level. (Funded by Bristol-Myers Squibb; CheckMate 017 ClinicalTrials.gov number, NCT01642004.).
Collapse
Affiliation(s)
- Julie Brahmer
- From the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (J.B.); the City of Hope Comprehensive Cancer Center, Duarte, CA (K.L.R.); the Netherlands Cancer Institute and Antoni van Leeuwenhoek Hospital, Amsterdam (P.B.), Erasmus MC Cancer Institute, Rotterdam (J.G.A.), and Ziekenhuis Amphia, Breda (J.G.A.) - all in the Netherlands; the University Hospital of Perugia, Perugia (L.C.), and the Fondazione IRCCS Istituto Nazionale dei Tumori, Milan (M.C.G.) - both in Italy; the Department of Medical Oncology, West German Cancer Center, Universitätsklinikum Essen, and the Ruhrlandklinik, Universität Duisburg-Essen, Essen (W.E.E.E.), the Thoraxklinik, Heidelberg University Hospital, Heidelberg (M.S.), and the LungenClinic Grosshansdorf, Grosshansdorf (M.R.) - all in Germany; the N.N. Blokhin Russian Cancer Research Center, Moscow (E.P.); the H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL (S.A.); the Centrum Onkologii-Instytut im. Marii Skłodowskiej-Curie, Warsaw, Poland (A.P.); the University of Chicago Medicine and Biological Sciences, Chicago (E.E.V.); the Hospital Madrid Norte Sanchinarro (E.H.), the Hospital Universitario Fundación Jiménez Díaz, Madrid (M.D.), and the Hospital Universitario Virgen Del Rocío, Seville (L.P.-A.) - all in Spain; Oncology Hematology Care, Cincinnati (D.W.); the Duke University Medical Center, Durham, NC (N.R.); Massachusetts General Hospital, Boston (J.G.); Centro Internacional de Estudios Clinicos, Santiago, Chile (O.A.F.); Nemocnice Na Bulovce, Prague, Czech Republic (L.H.); Bristol-Myers Squibb, Princeton, NJ (C.B., C.T.H., B.L.); and the Sarah Cannon Research Institute and Tennessee Oncology, Nashville (D.R.S.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6253
|
Preusser M, Berghoff AS, Koller R, Zielinski CC, Hainfellner JA, Liebmann-Reindl S, Popitsch N, Geier CB, Streubel B, Birner P. Spectrum of gene mutations detected by next generation exome sequencing in brain metastases of lung adenocarcinoma. Eur J Cancer 2015; 51:1803-11. [PMID: 26164066 DOI: 10.1016/j.ejca.2015.06.107] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 06/15/2015] [Accepted: 06/19/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Brain metastases (BM) are a life-threatening complication. We aimed to analyse gene mutations in lung adenocarcinoma BM. METHODS We performed next generation sequencing (NGS) of a pre-defined set of 48 cancer-related genes in a cohort of 76 neurosurgical lung adenocarcinoma BM specimens using a cancer specific gene panel on the MiSeq platform (Illumina, San Diego, CA). NGS results were statistically correlated to patient characteristics. Data on ALK, ROS1, MET and FGFR1 gene status assessed by FISH were available from previous studies in the majority of patients. RESULTS Twenty-nine (60.4%) of the 48 investigated cancer-related genes were mutated in at least one BM sample and 64 (84.2%) of the 76 BM samples carried at least one mutated gene. The number of mutated genes per sample ranged from 0 to 9 (median 2). The most commonly mutated genes were TP53, KRAS and CDKN2A, which were affected in 35/76 (46.1%), 29/76 (38.2%) and 17/76 (22.4%) samples, respectively. Other potentially druggable alterations included EGFR mutations (3/76, 3.9% of samples), PIK3CA mutation (2/76, 2.6%), BRAF mutation (1/76, 1.3%) and SMO mutation (1/76, 1.3%). Presence of KRAS mutations was associated with positive smoking history (p=0.015, Chi square test) and presence of EGFR mutation correlated with unfavourable overall survival time from BM diagnosis (p=0.019, log rank test). CONCLUSIONS Deleterious gene mutations, some of them with potential therapeutic implications, are found in a high fraction of lung adenocarcinoma BM.
Collapse
Affiliation(s)
- Matthias Preusser
- Department of Medicine I, Medical University of Vienna, Austria; Comprehensive Cancer Center, Central Nervous System Unit (CCC-CNS), Medical University of Vienna, Austria
| | - Anna S Berghoff
- Department of Medicine I, Medical University of Vienna, Austria; Comprehensive Cancer Center, Central Nervous System Unit (CCC-CNS), Medical University of Vienna, Austria
| | - Romina Koller
- Department of Medicine I, Medical University of Vienna, Austria; Comprehensive Cancer Center, Central Nervous System Unit (CCC-CNS), Medical University of Vienna, Austria
| | - Christoph C Zielinski
- Department of Medicine I, Medical University of Vienna, Austria; Comprehensive Cancer Center, Central Nervous System Unit (CCC-CNS), Medical University of Vienna, Austria
| | - Johannes A Hainfellner
- Institute of Neurology, Medical University of Vienna, Austria; Comprehensive Cancer Center, Central Nervous System Unit (CCC-CNS), Medical University of Vienna, Austria
| | | | - Niko Popitsch
- Center for Integrative Bioinformatics Vienna (CIBIV), University of Vienna and Medical University of Vienna and Oxford NIHR Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Christoph B Geier
- Immunology Outpatient Clinic, Schwarzspanierstraße 15/1/9, A-1090 Vienna, Austria
| | - Berthold Streubel
- Core facilities, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Central Nervous System Unit (CCC-CNS), Medical University of Vienna, Austria.
| | - Peter Birner
- Department of Pathology, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Central Nervous System Unit (CCC-CNS), Medical University of Vienna, Austria
| |
Collapse
|
6254
|
Chinai JM, Janakiram M, Chen F, Chen W, Kaplan M, Zang X. New immunotherapies targeting the PD-1 pathway. Trends Pharmacol Sci 2015; 36:587-95. [PMID: 26162965 DOI: 10.1016/j.tips.2015.06.005] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 12/22/2022]
Abstract
Ligands from the B7 family bind to receptors of the CD28 family, which regulate early T cell activation in lymphoid organs and control inflammation and autoimmunity in peripheral tissues. Programmed death-1 (PD-1), a member of the CD28 family, is an inhibitory receptor on T cells and is responsible for their dysfunction in infectious diseases and cancers. The complex mechanisms controlling the expression and signaling of PD-1 and programmed death ligand 1 (PD-L1) are emerging. Recently completed and ongoing clinical trials that target these molecules have shown remarkable success by generating durable clinical responses in some cancer patients. In chronic viral infections, preclinical data reveal that targeting PD-1 and its ligands can improve T cell responses and virus clearance. There is also promise in stimulating this pathway for the treatment of autoimmune and inflammatory disorders.
Collapse
Affiliation(s)
- Jordan M Chinai
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Murali Janakiram
- Department of Oncology, Montefiore Medical Center, New York, NY 10467, USA
| | - Fuxiang Chen
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wantao Chen
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mark Kaplan
- Centers for Therapeutic Innovation, Pfizer Inc., New York, NY 10016, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Oncology, Montefiore Medical Center, New York, NY 10467, USA.
| |
Collapse
|
6255
|
Manipulating the in vivo immune response by targeted gene knockdown. Curr Opin Immunol 2015; 35:63-72. [PMID: 26149459 DOI: 10.1016/j.coi.2015.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/09/2015] [Accepted: 06/19/2015] [Indexed: 02/06/2023]
Abstract
Aptamers, nucleic acids selected for high affinity binding to proteins, can be used to activate or antagonize immune mediators or receptors in a location and cell-type specific manner and to enhance antigen presentation. They can also be linked to other molecules (other aptamers, siRNAs or miRNAs, proteins, toxins) to produce multifunctional compounds for targeted immune modulation in vivo. Aptamer-siRNA chimeras (AsiCs) that induce efficient cell-specific knockdown in immune cells in vitro and in vivo can be used as an immunological research tool or potentially as an immunomodulating therapeutic.
Collapse
|
6256
|
The antigenic landscape of multiple myeloma: mass spectrometry (re)defines targets for T-cell-based immunotherapy. Blood 2015; 126:1203-13. [PMID: 26138685 DOI: 10.1182/blood-2015-04-640532] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/22/2015] [Indexed: 12/16/2022] Open
Abstract
Direct analysis of HLA-presented antigens by mass spectrometry provides a comprehensive view on the antigenic landscape of different tissues/malignancies and enables the identification of novel, pathophysiologically relevant T-cell epitopes. Here, we present a systematic and comparative study of the HLA class I and II presented, nonmutant antigenome of multiple myeloma (MM). Quantification of HLA surface expression revealed elevated HLA molecule counts on malignant plasma cells compared with normal B cells, excluding relevant HLA downregulation in MM. Analyzing the presentation of established myeloma-associated T-cell antigens on the HLA ligandome level, we found a substantial proportion of antigens to be only infrequently presented on primary myelomas or to display suboptimal degrees of myeloma specificity. However, unsupervised analysis of our extensive HLA ligand data set delineated a panel of 58 highly specific myeloma-associated antigens (including multiple myeloma SET domain containing protein) which are characterized by frequent and exclusive presentation on myeloma samples. Functional characterization of these target antigens revealed peptide-specific, preexisting CD8(+) T-cell responses exclusively in myeloma patients, which is indicative of pathophysiological relevance. Furthermore, in vitro priming experiments revealed that peptide-specific T-cell responses can be induced in response-naive myeloma patients. Together, our results serve to guide antigen selection for T-cell-based immunotherapy of MM.
Collapse
|
6257
|
Nelson BH, McAlpine JN. The more tumors change, the more they stay tame: Do T cells keep POLE ultramutated endometrial carcinomas in check? Gynecol Oncol 2015; 138:1-2. [DOI: 10.1016/j.ygyno.2015.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/03/2015] [Indexed: 01/10/2023]
|
6258
|
Ali HR, Glont SE, Blows FM, Provenzano E, Dawson SJ, Liu B, Hiller L, Dunn J, Poole CJ, Bowden S, Earl HM, Pharoah PDP, Caldas C. PD-L1 protein expression in breast cancer is rare, enriched in basal-like tumours and associated with infiltrating lymphocytes. Ann Oncol 2015; 26:1488-93. [PMID: 25897014 DOI: 10.1093/annonc/mdv192] [Citation(s) in RCA: 223] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/13/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Expression of programmed death ligand 1 (PD-L1) in solid tumours has been shown to predict whether patients are likely to respond to anti-PD-L1 therapies. To estimate the therapeutic potential of PD-L1 inhibition in breast cancer, we evaluated the prevalence and significance of PD-L1 protein expression in a large collection of breast tumours. PATIENTS AND METHODS Correlations between CD274 (PD-L1) copy number, transcript and protein levels were evaluated in tumours from 418 patients recruited to the METABRIC genomic study. Immunohistochemistry was used to detect PD-L1 protein in breast tumours in tissue microarrays from 5763 patients recruited to the SEARCH population-based study (N = 4079) and the NEAT randomised, controlled trial (N = 1684). RESULTS PD-L1 protein data was available for 3916 of the possible 5763 tumours from the SEARCH and NEAT studies. PD-L1 expression by immune cells was observed in 6% (235/3916) of tumours and expression by tumour cells was observed in just 1.7% (66/3916). PD-L1 was most frequently expressed in basal-like tumours. This was observed both where tumours were subtyped by combined copy number and expression profiling [39% (17/44) of IntClust 10 i.e. basal-like tumours were PD-L1 immune cell positive; P < 0.001] and where a surrogate IHC-based classifier was used [19% (56/302) of basal-like tumours were PD-L1 immune cell positive; P < 0.001]. Moreover, CD274 (PD-L1) amplification was observed in five tumours of which four were IntClust 10. Expression of PD-L1 by either tumour cells or infiltrating immune cells was positively correlated with infiltration by both cytotoxic and regulatory T cells (P < 0.001). There was a nominally significant association between PD-L1 and improved disease-specific survival (hazard ratio 0.53, 95% confidence interval 0.26-1.07; P = 0.08) in ER-negative disease. CONCLUSIONS Expression of PD-L1 is rare in breast cancer, markedly enriched in basal-like tumours and is correlated with infiltrating lymphocytes. PD-L1 inhibition may benefit the 19% of patients with basal-like tumours in which the protein is expressed. NEAT CLINICALTRIALSGOV NCT00003577.
Collapse
Affiliation(s)
- H R Ali
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge Department of Pathology, University of Cambridge, Cambridge
| | - S-E Glont
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge
| | - F M Blows
- Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge
| | - E Provenzano
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| | - S-J Dawson
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| | - B Liu
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge
| | - L Hiller
- Warwick Clinical Trials Unit, University of Warwick, Coventry
| | - J Dunn
- Warwick Clinical Trials Unit, University of Warwick, Coventry
| | - C J Poole
- Warwick Clinical Trials Unit, University of Warwick, Coventry
| | - S Bowden
- Cancer Research UK Clinical Trials Unit, Institute for Cancer Studies, The University of Birmingham, Edgbaston, Birmingham
| | - H M Earl
- Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| | - P D P Pharoah
- Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge Strangeways Research Laboratory, University of Cambridge, Cambridge, UK
| | - C Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| |
Collapse
|
6259
|
Jamal-Hanjani M, Lee SM. Lung cancer. Lung Cancer Manag 2015. [DOI: 10.2217/lmt.15.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mariam Jamal-Hanjani Mariam Jamal-Hanjani is a Specialist Registrar in Medical Oncology. She studied physics and then medicine at University College London (UCL), and is currently pursuing her PhD in non-small-cell lung cancer tumor evolution and intratumor heterogeneity at the Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute.
Collapse
Affiliation(s)
- Mariam Jamal-Hanjani
- University College London Hospitals NHS Trust, UCL Cancer Institute & CRUK Lung Cancer Centre of Excellence, 250 Euston Road, London, NW1 2PT, UK
| | - Siow-Ming Lee
- University College London Hospitals NHS Trust, UCL Cancer Institute & CRUK Lung Cancer Centre of Excellence, 250 Euston Road, London, NW1 2PT, UK
| |
Collapse
|
6260
|
Swanton C, McGranahan N, Starrett GJ, Harris RS. APOBEC Enzymes: Mutagenic Fuel for Cancer Evolution and Heterogeneity. Cancer Discov 2015; 5:704-12. [PMID: 26091828 PMCID: PMC4497973 DOI: 10.1158/2159-8290.cd-15-0344] [Citation(s) in RCA: 373] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/14/2015] [Indexed: 12/16/2022]
Abstract
UNLABELLED Deep sequencing technologies are revealing the complexities of cancer evolution, casting light on mutational processes fueling tumor adaptation, immune escape, and treatment resistance. Understanding mechanisms driving cancer diversity is a critical step toward developing strategies to attenuate tumor evolution and adaptation. One emerging mechanism fueling tumor diversity and subclonal evolution is genomic DNA cytosine deamination catalyzed by APOBEC3B and at least one other APOBEC family member. Deregulation of APOBEC3 enzymes causes a general mutator phenotype that manifests as diverse and heterogeneous tumor subclones. Here, we summarize knowledge of the APOBEC DNA deaminase family in cancer, and their role as driving forces for intratumor heterogeneity and a therapeutic target to limit tumor adaptation. SIGNIFICANCE APOBEC mutational signatures may be enriched in tumor subclones, suggesting APOBEC cytosine deaminases fuel subclonal expansions and intratumor heterogeneity. APOBEC family members might represent a new class of drug target aimed at limiting tumor evolution, adaptation, and drug resistance.
Collapse
Affiliation(s)
- Charles Swanton
- The Francis Crick Institute, London, United Kingdom. UCL Cancer Institute, CRUK Lung Cancer Centre of Excellence, London, United Kingdom.
| | - Nicholas McGranahan
- The Francis Crick Institute, London, United Kingdom. Centre for Mathematics & Physics in the Life Sciences & Experimental Biology (CoMPLEX), University College London, London, United Kingdom
| | - Gabriel J Starrett
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Reuben S Harris
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
6261
|
Turnis ME, Andrews LP, Vignali DAA. Inhibitory receptors as targets for cancer immunotherapy. Eur J Immunol 2015; 45:1892-905. [PMID: 26018646 PMCID: PMC4549156 DOI: 10.1002/eji.201344413] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 04/25/2015] [Accepted: 05/26/2015] [Indexed: 12/11/2022]
Abstract
Inhibitory receptors expressed on T cells control immune responses while limiting autoimmunity. However, tumors can hijack these "checkpoints" for protection from immune attack. Tumor-specific T cells that exhibit an exhausted, unresponsive phenotype express high levels of inhibitory receptors including CTLA4, PD1, and LAG3, among others. Intratumoral regulatory T cells promote immunosuppression and also express multiple inhibitory receptors. Overcoming this inhibitory receptor-mediated immune tolerance has thus been a major focus of recent cancer immunotherapeutic developments. Here, we review how boosting the host's immune system by blocking inhibitory receptor signaling with antagonistic mAbs restores the capacity of T cells to drive durable antitumor immune responses. Clinical trials targeting the CTLA4 and PD1 pathways have shown durable effects in multiple tumor types. Many combinatorial therapies are currently being investigated with encouraging results that highlight enhanced antitumor immunogenicity and improved patient survival. Finally, we will discuss the ongoing identification and dissection of novel T-cell inhibitory receptor pathways, which could lead to the development of new combinatorial therapeutic approaches.
Collapse
Affiliation(s)
- Meghan E Turnis
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Dario A A Vignali
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
6262
|
Schaue D, McBride WH. Opportunities and challenges of radiotherapy for treating cancer. Nat Rev Clin Oncol 2015; 12:527-40. [PMID: 26122185 DOI: 10.1038/nrclinonc.2015.120] [Citation(s) in RCA: 504] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The past 20 years have seen dramatic changes in the delivery of radiation therapy, but the impact of radiobiology on the clinic has been far less substantial. A major consideration in the use of radiotherapy has been on how best to exploit differences between the tumour and host tissue characteristics, which in the past has been achieved empirically by radiation-dose fractionation. New advances are uncovering some of the mechanistic processes that underlie this success story. In this Review, we focus on how these processes might be targeted to improve the outcome of radiotherapy at the individual patient level. This approach would seem a more productive avenue of treatment than simply trying to increase the radiation dose delivered to the tumour.
Collapse
Affiliation(s)
- Dörthe Schaue
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| | - William H McBride
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| |
Collapse
|
6263
|
Galaine J, Borg C, Godet Y, Adotévi O. Interest of Tumor-Specific CD4 T Helper 1 Cells for Therapeutic Anticancer Vaccine. Vaccines (Basel) 2015; 3:490-502. [PMID: 26350591 PMCID: PMC4586463 DOI: 10.3390/vaccines3030490] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 12/16/2022] Open
Abstract
Nowadays, immunotherapy represents one promising approach for cancer treatment. Recently, spectacular results of cancer immunotherapy clinical trials have confirmed the crucial role of immune system in cancer regression. Therapeutic cancer vaccine represents one widely used immunotherapy strategy to stimulate tumor specific T cell responses but clinical impact remains disappointing in targeting CD8 T cells. Although CD8 T cells have been initially considered to be the main protagonists, it is now clear that CD4 T cells also play a critical role in antitumor response. In this article, we discuss the role of tumor antigen-specific CD4 T cell responses and how we can target these cells to improve cancer vaccines.
Collapse
Affiliation(s)
- Jeanne Galaine
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
| | - Christophe Borg
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
- Department of Medical Oncology, University Hospital of Besançon, Besançon cedex F25020, France.
| | - Yann Godet
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
| | - Olivier Adotévi
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
- Department of Medical Oncology, University Hospital of Besançon, Besançon cedex F25020, France.
| |
Collapse
|
6264
|
Verdeil G, Fuertes Marraco SA, Murray T, Speiser DE. From T cell "exhaustion" to anti-cancer immunity. Biochim Biophys Acta Rev Cancer 2015; 1865:49-57. [PMID: 26123831 DOI: 10.1016/j.bbcan.2015.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 06/18/2015] [Accepted: 06/23/2015] [Indexed: 12/14/2022]
Abstract
The immune system has the potential to protect from malignant diseases for extended periods of time. Unfortunately, spontaneous immune responses are often inefficient. Significant effort is required to develop reliable, broadly applicable immunotherapies for cancer patients. A major innovation was transplantation with hematopoietic stem cells from genetically distinct donors for patients with hematologic malignancies. In this setting, donor T cells induce long-term remission by keeping cancer cells in check through powerful allogeneic graft-versus-leukemia effects. More recently, a long awaited breakthrough for patients with solid tissue cancers was achieved, by means of therapeutic blockade of T cell inhibitory receptors. In untreated cancer patients, T cells are dysfunctional and remain in a state of T cell "exhaustion". Nonetheless, they often retain a high potential for successful defense against cancer, indicating that many T cells are not entirely and irreversibly exhausted but can be mobilized to become highly functional. Novel antibody therapies that block inhibitory receptors can lead to strong activation of anti-tumor T cells, mediating clinically significant anti-cancer immunity for many years. Here we review these new treatments and the current knowledge on tumor antigen-specific T cells.
Collapse
Affiliation(s)
- Grégory Verdeil
- Ludwig Cancer Research Center and Department of Oncology, Clinical Tumor Biology & Immunotherapy Group, Lausanne University Hospital Center (CHUV) and University of Lausanne, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland
| | - Silvia A Fuertes Marraco
- Ludwig Cancer Research Center and Department of Oncology, Clinical Tumor Biology & Immunotherapy Group, Lausanne University Hospital Center (CHUV) and University of Lausanne, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland
| | - Timothy Murray
- Ludwig Cancer Research Center and Department of Oncology, Clinical Tumor Biology & Immunotherapy Group, Lausanne University Hospital Center (CHUV) and University of Lausanne, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland
| | - Daniel E Speiser
- Ludwig Cancer Research Center and Department of Oncology, Clinical Tumor Biology & Immunotherapy Group, Lausanne University Hospital Center (CHUV) and University of Lausanne, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland.
| |
Collapse
|
6265
|
Capitini CM, Romero P. JITC launches a new section: commentary and editorials. J Immunother Cancer 2015; 3:28. [PMID: 26110058 PMCID: PMC4479236 DOI: 10.1186/s40425-015-0073-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 11/10/2022] Open
Affiliation(s)
- Christian M Capitini
- Department of Pediatrics and Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI USA
| | - Pedro Romero
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
6266
|
Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, Daud A, Carlino MS, McNeil C, Lotem M, Larkin J, Lorigan P, Neyns B, Blank CU, Hamid O, Mateus C, Shapira-Frommer R, Kosh M, Zhou H, Ibrahim N, Ebbinghaus S, Ribas A. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med 2015; 372:2521-32. [PMID: 25891173 DOI: 10.1056/nejmoa1503093] [Citation(s) in RCA: 4455] [Impact Index Per Article: 445.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The immune checkpoint inhibitor ipilimumab is the standard-of-care treatment for patients with advanced melanoma. Pembrolizumab inhibits the programmed cell death 1 (PD-1) immune checkpoint and has antitumor activity in patients with advanced melanoma. METHODS In this randomized, controlled, phase 3 study, we assigned 834 patients with advanced melanoma in a 1:1:1 ratio to receive pembrolizumab (at a dose of 10 mg per kilogram of body weight) every 2 weeks or every 3 weeks or four doses of ipilimumab (at 3 mg per kilogram) every 3 weeks. Primary end points were progression-free and overall survival. RESULTS The estimated 6-month progression-free-survival rates were 47.3% for pembrolizumab every 2 weeks, 46.4% for pembrolizumab every 3 weeks, and 26.5% for ipilimumab (hazard ratio for disease progression, 0.58; P<0.001 for both pembrolizumab regimens versus ipilimumab; 95% confidence intervals [CIs], 0.46 to 0.72 and 0.47 to 0.72, respectively). Estimated 12-month survival rates were 74.1%, 68.4%, and 58.2%, respectively (hazard ratio for death for pembrolizumab every 2 weeks, 0.63; 95% CI, 0.47 to 0.83; P=0.0005; hazard ratio for pembrolizumab every 3 weeks, 0.69; 95% CI, 0.52 to 0.90; P=0.0036). The response rate was improved with pembrolizumab administered every 2 weeks (33.7%) and every 3 weeks (32.9%), as compared with ipilimumab (11.9%) (P<0.001 for both comparisons). Responses were ongoing in 89.4%, 96.7%, and 87.9% of patients, respectively, after a median follow-up of 7.9 months. Efficacy was similar in the two pembrolizumab groups. Rates of treatment-related adverse events of grade 3 to 5 severity were lower in the pembrolizumab groups (13.3% and 10.1%) than in the ipilimumab group (19.9%). CONCLUSIONS The anti-PD-1 antibody pembrolizumab prolonged progression-free survival and overall survival and had less high-grade toxicity than did ipilimumab in patients with advanced melanoma. (Funded by Merck Sharp & Dohme; KEYNOTE-006 ClinicalTrials.gov number, NCT01866319.).
Collapse
|
6267
|
Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Duffy SM, Goldberg RM, de la Chapelle A, Koshiji M, Bhaijee F, Huebner T, Hruban RH, Wood LD, Cuka N, Pardoll DM, Papadopoulos N, Kinzler KW, Zhou S, Cornish TC, Taube JM, Anders RA, Eshleman JR, Vogelstein B, Diaz LA. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med 2015; 372:2509-20. [PMID: 26028255 PMCID: PMC4481136 DOI: 10.1056/nejmoa1500596] [Citation(s) in RCA: 7184] [Impact Index Per Article: 718.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Somatic mutations have the potential to encode "non-self" immunogenic antigens. We hypothesized that tumors with a large number of somatic mutations due to mismatch-repair defects may be susceptible to immune checkpoint blockade. METHODS We conducted a phase 2 study to evaluate the clinical activity of pembrolizumab, an anti-programmed death 1 immune checkpoint inhibitor, in 41 patients with progressive metastatic carcinoma with or without mismatch-repair deficiency. Pembrolizumab was administered intravenously at a dose of 10 mg per kilogram of body weight every 14 days in patients with mismatch repair-deficient colorectal cancers, patients with mismatch repair-proficient colorectal cancers, and patients with mismatch repair-deficient cancers that were not colorectal. The coprimary end points were the immune-related objective response rate and the 20-week immune-related progression-free survival rate. RESULTS The immune-related objective response rate and immune-related progression-free survival rate were 40% (4 of 10 patients) and 78% (7 of 9 patients), respectively, for mismatch repair-deficient colorectal cancers and 0% (0 of 18 patients) and 11% (2 of 18 patients) for mismatch repair-proficient colorectal cancers. The median progression-free survival and overall survival were not reached in the cohort with mismatch repair-deficient colorectal cancer but were 2.2 and 5.0 months, respectively, in the cohort with mismatch repair-proficient colorectal cancer (hazard ratio for disease progression or death, 0.10 [P<0.001], and hazard ratio for death, 0.22 [P=0.05]). Patients with mismatch repair-deficient noncolorectal cancer had responses similar to those of patients with mismatch repair-deficient colorectal cancer (immune-related objective response rate, 71% [5 of 7 patients]; immune-related progression-free survival rate, 67% [4 of 6 patients]). Whole-exome sequencing revealed a mean of 1782 somatic mutations per tumor in mismatch repair-deficient tumors, as compared with 73 in mismatch repair-proficient tumors (P=0.007), and high somatic mutation loads were associated with prolonged progression-free survival (P=0.02). CONCLUSIONS This study showed that mismatch-repair status predicted clinical benefit of immune checkpoint blockade with pembrolizumab. (Funded by Johns Hopkins University and others; ClinicalTrials.gov number, NCT01876511.).
Collapse
Affiliation(s)
- Dung T Le
- From the Swim Across America Laboratory (D.T.L., J.N.U., B.R.B., L.A.D.), Sidney Kimmel Comprehensive Cancer Center (D.T.L., J.N.U., H.W., H.K., A.D.E., A.D.S., B.S.L., N.S.A., D.L., B.B., R.C.D., D.M.P., N.P., K.W.K., S.Z., B.V., L.A.D.), Ludwig Center and Howard Hughes Medical Institute (B.R.B., A.D.S., N.P., K.W.K., S.Z., B.V., L.A.D.), and the Departments of Radiology (A.Z.) and Pathology (F.B., T.H., R.H.H., L.D.W., N.C., T.C.C., J.M.T., R.A.A., J.R.E.), Johns Hopkins University School of Medicine, Baltimore; Department of Medicine, Stanford University School of Medicine, Stanford, CA (G.A.F.); Providence Cancer Center at Providence Health and Services, Portland, OR (T.S.C.); Department of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh (J.J.L.); Bon Secours Cancer Institute, Richmond, VA (S.M.D.); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center-James Cancer Center and Solove Research Institute, and Human Cancer Genetics Program, Ohio State University Comprehensive Cancer Center, Columbus (R.M.G., A.C.); and Merck, Kenilworth, NJ, and North Wales, PA (M.K.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6268
|
Delamarre L, Mellman I, Yadav M. Cancer immunotherapy. Neo approaches to cancer vaccines. Science 2015; 348:760-1. [PMID: 25977539 DOI: 10.1126/science.aab3465] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
| | - Ira Mellman
- Genentech, South San Francisco, CA 94080, USA.
| | | |
Collapse
|
6269
|
Carbognin L, Pilotto S, Milella M, Vaccaro V, Brunelli M, Caliò A, Cuppone F, Sperduti I, Giannarelli D, Chilosi M, Bronte V, Scarpa A, Bria E, Tortora G. Differential Activity of Nivolumab, Pembrolizumab and MPDL3280A according to the Tumor Expression of Programmed Death-Ligand-1 (PD-L1): Sensitivity Analysis of Trials in Melanoma, Lung and Genitourinary Cancers. PLoS One 2015; 10:e0130142. [PMID: 26086854 PMCID: PMC4472786 DOI: 10.1371/journal.pone.0130142] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/18/2015] [Indexed: 12/13/2022] Open
Abstract
Background The potential predictive role of programmed death-ligand-1 (PD-L1) expression on tumor cells in the context of solid tumor treated with checkpoint inhibitors targeting the PD-1 pathway represents an issue for clinical research. Methods Overall response rate (ORR) was extracted from phase I-III trials investigating nivolumab, pembrolizumab and MPDL3280A for advanced melanoma, non-small cell lung cancer (NSCLC) and genitourinary cancer, and cumulated by adopting a fixed and random-effect model with 95% confidence interval (CI). Interaction test according to tumor PD-L1 was accomplished. A sensitivity analysis according to adopted drug, tumor type, PD-L1 cut-off and treatment line was performed. Results Twenty trials (1,475 patients) were identified. A significant interaction (p<0.0001) according to tumor PD-L1 expression was found in the overall sample with an ORR of 34.1% (95% CI 27.6-41.3%) in the PD-L1 positive and 19.9% (95% CI 15.4-25.3%) in the PD-L1 negative population. ORR was significantly higher in PD-L1 positive in comparison to PD-L1 negative patients for nivolumab and pembrolizumab, with an absolute difference of 16.4% and 19.5%, respectively. A significant difference in activity of 22.8% and 8.7% according to PD-L1 was found for melanoma and NSCLC, respectively, with no significant difference for genitourinary cancer. Conclusion Overall, the three antibodies provide a significant differential effect in terms of activity according to PD-L1 expression on tumor cells. The predictive value of PD-L1 on tumor cells seems to be more robust for anti-PD-1 antibody (nivolumab and pembrolizumab), and in the context of advanced melanoma and NSCLC.
Collapse
Affiliation(s)
- Luisa Carbognin
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Sara Pilotto
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Michele Milella
- Medical Oncology, Regina Elena National Cancer Institute, Roma, Italy
| | - Vanja Vaccaro
- Medical Oncology, Regina Elena National Cancer Institute, Roma, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Anna Caliò
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | | | | | - Marco Chilosi
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Vincenzo Bronte
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Aldo Scarpa
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
- ARC-NET Center for Applied Research on Cancer, Verona, Italy
| | - Emilio Bria
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
- * E-mail:
| | - Giampaolo Tortora
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| |
Collapse
|
6270
|
Qian J, Zou Y, Wang J, Zhang B, Massion PP. Global gene expression profiling reveals a suppressed immune response pathway associated with 3q amplification in squamous carcinoma of the lung. GENOMICS DATA 2015; 5:272-4. [PMID: 26484266 PMCID: PMC4583673 DOI: 10.1016/j.gdata.2015.06.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 06/01/2015] [Indexed: 11/20/2022]
Abstract
Chromosome 3q26–28 is a critical region of genomic amplification in non-small cell lung cancer (NSCLC), particularly lung squamous cell carcinomas (SCCs). No molecular therapeutic target has shown clinical utility for SCC, in contrast with adenocarcinomas of the lung. To identify novel candidate drivers in this region, we performed both Array Comparative Genomic Hybridization (array CGH, Agilent Human Genome CGH 244A oligo-microarrays) and Gene Expression Microarray (Agilent Human Gene Expression 4 × 44 K microarray) on 24 untreated lung SCC specimens. Using our previously published integrative genomics approach, we identified 12 top amplified driver genes within this region that are highly correlated and overexpressed in lung SCC. We further demonstrated one of the 12 top amplified driver Fragile X mental retardation-related protein 1 (FXR1) as a novel cancer gene in NSCLC and FXR1 executes its regulatory function by forming a novel complex with two other oncogenes, protein kinase C, iota ( PRKCI) and epithelial cell transforming 2 (ECT2) within the same amplicon in lung cancer cell. Here we report that immune response pathways are significantly suppressed in lung SCC and negatively associated with 3q driver gene expression, implying a potential role of 3q drivers in cancer immune-surveillance. In light of the attractive immunotherapy strategy using blockade of negative regulators of T cell function for multiple human cancer including lung SCC, our findings may provide a rationale for targeting 3q drivers in combination of immunotherapies for human tumors harboring the 3q amplicon. The data have been deposited in NCBI's Gene Expression Omnibus and are accessible through GEO Series accession number GSE40089.
Collapse
Affiliation(s)
- Jun Qian
- Thoracic Program at the Vanderbilt Ingram Cancer Center, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Nashville, TN, USA
| | - Yong Zou
- Thoracic Program at the Vanderbilt Ingram Cancer Center, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Nashville, TN, USA
| | - Jing Wang
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - Bing Zhang
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - Pierre P. Massion
- Thoracic Program at the Vanderbilt Ingram Cancer Center, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Nashville, TN, USA
- Veterans Affairs Medical Center, Nashville, TN, USA
- Corresponding author at: Thoracic Program, Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, Preston Research Building 640, Nashville, TN 37232-6838, USA. Tel.: + 1 615 936 2256; fax: + 1 615 936 1790.
| |
Collapse
|
6271
|
Abstract
Scientific advances in the last decade have demonstrated the critical role of host immune system in the elimination and suppression of cancer cells. Better knowledge of signaling pathways has enabled the development of new cancer immunotherapy. The discovery of negative feedback mechanisms following the lymphocyte activation has promoted the development of new antibodies targeting molecule inhibitors such as PD1, PDL1 or CTLA-4. Dramatic results were obtained with melanoma. Checkpoint inhibitors (pembrolizumab and ipilimumab) have many advantages in terms of rate of objective response and overall survival. Recent studies in translational research aimed to understand and analyze mechanisms of action of anti-PD1/anti-PDL1. Expression of PDL1 in the tumor is associated with a significantly greater objective response rate (immunohistochemistry). Nevertheless, limits with tumor immunohistochemical analysis encourage new biomarkers research. Other immunotherapy approaches, such as cell and gene therapies using engineered T cells call for further advancements to broaden their applicability. However, these therapies are very expensive and their manufacturing process very restrictive, which could lately limit their use in case of inefficiency of checkpoint inhibitors or when lymphocytic infiltration in tumor is absent. In this case, the objective would be to engineer ex vivo the patient's immune system by restoring the ability of T cells to identify and suppress tumor cells. Currently, two gene-reprogramming tools are under development: chimeric antigen receptor and TCR modified T cells.
Collapse
|
6272
|
Huang J, Wang L, Cong Z, Amoozgar Z, Kiner E, Xing D, Orsulic S, Matulonis U, Goldberg MS. The PARP1 inhibitor BMN 673 exhibits immunoregulatory effects in a Brca1(-/-) murine model of ovarian cancer. Biochem Biophys Res Commun 2015; 463:551-6. [PMID: 26047697 DOI: 10.1016/j.bbrc.2015.05.083] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/21/2022]
Abstract
Familial breast and ovarian cancer are often caused by inherited mutations of BRCA1. While current prognoses for such patients are rather poor, inhibition of poly-ADP ribose polymerase 1 (PARP1) induces synthetic lethality in cells that are defective in homologous recombination. BMN 673 is a potent PARP1 inhibitor that is being clinically evaluated for treatment of BRCA-mutant cancers. Using the Brca1-deficient murine epithelial ovarian cancer cell line BR5FVB1-Akt, we investigated whether the antitumor effects of BMN 673 extend beyond its known pro-apoptotic function. Administration of modest amounts of BMN 673 greatly improved the survival of mice bearing subcutaneous or intraperitoneal tumors. We thus hypothesized that BMN 673 may influence the composition and function of immune cells in the tumor microenvironment. Indeed, BMN 673 significantly increases the number of peritoneal CD8(+) T cells and NK cells as well as their production of IFN-γ and TNF-α. These data suggest that the cell stress caused by BMN 673 induces not only cancer cell-intrinsic apoptosis but also cancer cell-extrinsic antitumor immune effects in a syngeneic murine model of ovarian cancer. BMN 673 may therefore serve as a promising adjuvant therapy to immunotherapy to achieve durable responses among patients whose tumors harbor defects in homologous recombination.
Collapse
Affiliation(s)
- Jing Huang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Lei Wang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Zhongyi Cong
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Zohreh Amoozgar
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Evgeny Kiner
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Deyin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Sandra Orsulic
- Women's Cancer Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ursula Matulonis
- Gynecologic Oncology Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael S Goldberg
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
6273
|
Abstract
The efficacy of cancer vaccines has long been hampered by insufficient definition of tumor-specific antigens. A recent study by Kreiter et al. published in Nature has provided a blueprint for a patient-tailored approach to develop individualized RNA vaccines.
Collapse
Affiliation(s)
- Michael Platten
- 1] DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany [2] Neurology Clinic, University Hospital Heidelberg and National Center for Tumor Diseases, Heidelberg, Germany
| | - Rienk Offringa
- 1] Dept. Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany [2] European Pancreas Center, Surgery Clinic, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
6274
|
Sunshine J, Taube JM. PD-1/PD-L1 inhibitors. Curr Opin Pharmacol 2015; 23:32-8. [PMID: 26047524 DOI: 10.1016/j.coph.2015.05.011] [Citation(s) in RCA: 441] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/06/2015] [Accepted: 05/15/2015] [Indexed: 12/26/2022]
Abstract
Tumors may adopt normal physiologic checkpoints for immunomodulation leading to an imbalance between tumor growth and host surveillance. Antibodies targeting the PD-1/PD-L1 checkpoint have shown dynamic and durable tumor regressions, suggesting a rebalancing of the host-tumor interaction. Nivolumab and pembrolizumab are the anti-PD-1 antibodies that are currently the furthest in clinical development, and anti-PD-L1 agents under investigation include MPDL3280A, MEDI4736, and BMS-936559. These agents have been used to treat advanced melanoma, non-small cell lung cancer, renal cell carcinoma, bladder cancer and Hodgkin lymphoma, amongst other tumor types. In this article, we review the updated response results for early clinical trials, note recent FDA actions regarding this class of agents, and summarize results across trials looking at PD-L1 status as a predictor of response to anti-PD-1/PD-L1.
Collapse
Affiliation(s)
- Joel Sunshine
- Department of Dermatology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Janis M Taube
- Department of Dermatology, Johns Hopkins Medical Institutions, Baltimore, MD, USA; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA; Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
6275
|
Karachaliou N, Cao MG, Teixidó C, Viteri S, Morales-Espinosa D, Santarpia M, Rosell R. Understanding the function and dysfunction of the immune system in lung cancer: the role of immune checkpoints. Cancer Biol Med 2015; 12:79-86. [PMID: 26175923 PMCID: PMC4493378 DOI: 10.7497/j.issn.2095-3941.2015.0029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/10/2015] [Indexed: 12/17/2022] Open
Abstract
Survival rates for metastatic lung cancer, including non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), are poor with 5-year survivals of less than 5%. The immune system has an intricate and complex relationship with tumorigenesis; a groundswell of research on the immune system is leading to greater understanding of how cancer progresses and presenting new ways to halt disease progress. Due to the extraordinary power of the immune system-with its capacity for memory, exquisite specificity and central and universal role in human biology-immunotherapy has the potential to achieve complete, long-lasting remissions and cures, with few side effects for any cancer patient, regardless of cancer type. As a result, a range of cancer therapies are under development that work by turning our own immune cells against tumors. However deeper understanding of the complexity of immunomodulation by tumors is key to the development of effective immunotherapies, especially in lung cancer.
Collapse
Affiliation(s)
- Niki Karachaliou
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Maria Gonzalez Cao
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Cristina Teixidó
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Santiago Viteri
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Daniela Morales-Espinosa
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Mariacarmela Santarpia
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Rafael Rosell
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| |
Collapse
|
6276
|
Hellmann M, Rizvi N, Wolchok JD, Chan TA. Genomic profile, smoking, and response to anti-PD-1 therapy in non-small cell lung carcinoma. Mol Cell Oncol 2015; 3:e1048929. [PMID: 27308563 DOI: 10.1080/23723556.2015.1048929] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 05/02/2015] [Accepted: 05/04/2015] [Indexed: 01/05/2023]
Abstract
The recent successes of immune checkpoint therapies have established a new era for the treatment of patients with cancer, yet the predictors of response remain largely undetermined. We recently demonstrated that the genomic landscape of lung cancers substantially influences the response to programmed cell death 1 receptor (PD-1) blockade, providing new insights into the molecular determinants of the response to immunotherapy.
Collapse
Affiliation(s)
- Matthew Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY
| | - Naiyer Rizvi
- Division of Hematology/Oncology, New York Presbyterian/Columbia University Hospital , New York, NY
| | - Jedd D Wolchok
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center , New York, NY
| |
Collapse
|
6277
|
Parmiani G, Maccalli C, Maio M. Integrating Immune Checkpoint Blockade with Anti-Neo/Mutated Antigens Reactivity to Increase the Clinical Outcome of Immunotherapy. Vaccines (Basel) 2015; 3:420-8. [PMID: 26343195 PMCID: PMC4494352 DOI: 10.3390/vaccines3020420] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 12/21/2022] Open
Abstract
Antibodies to immune checkpoints have entered the clinical arena and have been shown to provide a clinical benefit for metastatic melanoma and, possibly, for other tumors as well. In this review paper we summarize this therapeutic activity and underline the functional mechanisms that may be involved. Among them, we discuss the so far neglected role of tumor-associated antigens (TAAs) deriving from tumor somatic mutations and summarize the results of recent trials showing the immunogenic strength of such TAAs which can be specifically targeted by T cells activated by immune checkpoint antibodies. Finally we discuss new immunotherapy approaches that involve the combination of self/shared- or neo-TAAs-based vaccines and immune checkpoint blockade antibodies, to increase the clinical response of metastatic melanoma patients.
Collapse
Affiliation(s)
- Giorgio Parmiani
- Italian Network for Bio-therapy of Tumors-(NIBIT)-Laboratory, c/o University Hospital of Siena, V. le Bracci, 16, Siena 53100, Italy.
- Division of Medical Oncology and Immunotherapy, University Hospital of Siena, V. le Bracci, 16, Siena 53100, Italy.
| | - Cristina Maccalli
- Italian Network for Bio-therapy of Tumors-(NIBIT)-Laboratory, c/o University Hospital of Siena, V. le Bracci, 16, Siena 53100, Italy.
- Division of Medical Oncology and Immunotherapy, University Hospital of Siena, V. le Bracci, 16, Siena 53100, Italy.
| | - Michele Maio
- Division of Medical Oncology and Immunotherapy, University Hospital of Siena, V. le Bracci, 16, Siena 53100, Italy.
| |
Collapse
|
6278
|
Pietanza MC, Byers LA, Minna JD, Rudin CM. Small cell lung cancer: will recent progress lead to improved outcomes? Clin Cancer Res 2015; 21:2244-55. [PMID: 25979931 PMCID: PMC4497796 DOI: 10.1158/1078-0432.ccr-14-2958] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy with a unique natural history characterized by a short doubling time, high growth fraction, and early development of widespread metastases. Although a chemotherapy- and radiation-sensitive disease, SCLC typically recurs rapidly after primary treatment, with only 6% of patients surviving 5 years from diagnosis. This disease has been notable for the absence of major improvements in its treatment: Nearly four decades after the introduction of a platinum-etoposide doublet, therapeutic options have remained virtually unchanged, with correspondingly little improvement in survival rates. Here, we summarize specific barriers and challenges inherent to SCLC research and care that have limited progress in novel therapeutic development to date. We discuss recent progress in basic and translational research, especially in the development of mouse models, which will provide insights into the patterns of metastasis and resistance in SCLC. Opportunities in clinical research aimed at exploiting SCLC biology are reviewed, with an emphasis on ongoing trials. SCLC has been described as a recalcitrant cancer, for which there is an urgent need for accelerated progress. The NCI convened a panel of laboratory and clinical investigators interested in SCLC with a goal of defining consensus recommendations to accelerate progress in the treatment of SCLC, which we summarize here.
Collapse
Affiliation(s)
- M Catherine Pietanza
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York.
| | - Lauren Averett Byers
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Charles M Rudin
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| |
Collapse
|
6279
|
Bergmann L, Brugger W, Herr W, Mackensen A, Multhoff G. [What opportunities does Immuno-oncology indicate for overarching long-term survival?]. Oncol Res Treat 2015; 38 Suppl 3:6-11. [PMID: 25966812 DOI: 10.1159/000381363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Lothar Bergmann
- Medizinische Klinik II: Hämatologie/Onkologie, Universitätsklinikum Frankfurt, Frankfurt/M., Deutschland
| | | | | | | | | |
Collapse
|
6280
|
Abstract
Immune checkpoint therapy, which targets regulatory pathways in T cells to enhance antitumor immune responses, has led to important clinical advances and provided a new weapon against cancer. This therapy has elicited durable clinical responses and, in a fraction of patients, long-term remissions where patients exhibit no clinical signs of cancer for many years. The way forward for this class of novel agents lies in our ability to understand human immune responses in the tumor microenvironment. This will provide valuable information regarding the dynamic nature of the immune response and regulation of additional pathways that will need to be targeted through combination therapies to provide survival benefit for greater numbers of patients.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Immunology, M.D. Anderson Cancer Center, Houston, TX, USA. Genitourinary Medical Oncology, M.D. Anderson Cancer Center, Houston, TX, USA.
| | - James P Allison
- Department of Immunology, M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
6281
|
Abstract
The clinical relevance of T cells in the control of a diverse set of human cancers is now beyond doubt. However, the nature of the antigens that allow the immune system to distinguish cancer cells from noncancer cells has long remained obscure. Recent technological innovations have made it possible to dissect the immune response to patient-specific neoantigens that arise as a consequence of tumor-specific mutations, and emerging data suggest that recognition of such neoantigens is a major factor in the activity of clinical immunotherapies. These observations indicate that neoantigen load may form a biomarker in cancer immunotherapy and provide an incentive for the development of novel therapeutic approaches that selectively enhance T cell reactivity against this class of antigens.
Collapse
Affiliation(s)
- Ton N Schumacher
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, Netherlands.
| | - Robert D Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
6282
|
Kreiter S, Vormehr M, van de Roemer N, Diken M, Löwer M, Diekmann J, Boegel S, Schrörs B, Vascotto F, Castle JC, Tadmor AD, Schoenberger SP, Huber C, Türeci Ö, Sahin U. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 2015; 520:692-6. [PMID: 25901682 DOI: 10.1038/nature14426] [Citation(s) in RCA: 969] [Impact Index Per Article: 96.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/30/2015] [Indexed: 12/23/2022]
Abstract
Tumour-specific mutations are ideal targets for cancer immunotherapy as they lack expression in healthy tissues and can potentially be recognized as neo-antigens by the mature T-cell repertoire. Their systematic targeting by vaccine approaches, however, has been hampered by the fact that every patient's tumour possesses a unique set of mutations ('the mutanome') that must first be identified. Recently, we proposed a personalized immunotherapy approach to target the full spectrum of a patient's individual tumour-specific mutations. Here we show in three independent murine tumour models that a considerable fraction of non-synonymous cancer mutations is immunogenic and that, unexpectedly, the majority of the immunogenic mutanome is recognized by CD4(+) T cells. Vaccination with such CD4(+) immunogenic mutations confers strong antitumour activity. Encouraged by these findings, we established a process by which mutations identified by exome sequencing could be selected as vaccine targets solely through bioinformatic prioritization on the basis of their expression levels and major histocompatibility complex (MHC) class II-binding capacity for rapid production as synthetic poly-neo-epitope messenger RNA vaccines. We show that vaccination with such polytope mRNA vaccines induces potent tumour control and complete rejection of established aggressively growing tumours in mice. Moreover, we demonstrate that CD4(+) T cell neo-epitope vaccination reshapes the tumour microenvironment and induces cytotoxic T lymphocyte responses against an independent immunodominant antigen in mice, indicating orchestration of antigen spread. Finally, we demonstrate an abundance of mutations predicted to bind to MHC class II in human cancers as well by employing the same predictive algorithm on corresponding human cancer types. Thus, the tailored immunotherapy approach introduced here may be regarded as a universally applicable blueprint for comprehensive exploitation of the substantial neo-epitope target repertoire of cancers, enabling the effective targeting of every patient's tumour with vaccines produced 'just in time'.
Collapse
Affiliation(s)
- Sebastian Kreiter
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany
| | - Mathias Vormehr
- Research Center for Immunotherapy (FZI), Langenbeckstrasse 1, Building 708, 55131 Mainz, Germany
| | - Niels van de Roemer
- Research Center for Immunotherapy (FZI), Langenbeckstrasse 1, Building 708, 55131 Mainz, Germany
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany
| | - Martin Löwer
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany
| | - Jan Diekmann
- 1] TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany [2] Biopharmaceutical New Technologies (BioNTech) Corporation, An der Goldgrube 12, 55131 Mainz, Germany
| | - Sebastian Boegel
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany
| | - Barbara Schrörs
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany
| | - Fulvia Vascotto
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany
| | - John C Castle
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany
| | - Arbel D Tadmor
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany
| | - Stephen P Schoenberger
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | - Christoph Huber
- Research Center for Immunotherapy (FZI), Langenbeckstrasse 1, Building 708, 55131 Mainz, Germany
| | - Özlem Türeci
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany
| | - Ugur Sahin
- 1] TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, 55131 Mainz, Germany [2] Research Center for Immunotherapy (FZI), Langenbeckstrasse 1, Building 708, 55131 Mainz, Germany [3] Biopharmaceutical New Technologies (BioNTech) Corporation, An der Goldgrube 12, 55131 Mainz, Germany
| |
Collapse
|
6283
|
Snyder A, Makarov V, Hellmann M, Rizvi N, Merghoub T, Wolchok JD, Chan TA. Genetics and immunology: reinvigorated. Oncoimmunology 2015; 4:e1029705. [PMID: 26451299 DOI: 10.1080/2162402x.2015.1029705] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 03/11/2015] [Indexed: 10/23/2022] Open
Abstract
Immune checkpoint blockade therapy is changing oncology by improving the outcome of patients with advanced malignancies. Our research has revealed the genetic features of tumors present in patients who initiate a successful antitumor immune response and derive clinical benefit from immune checkpoint blockade therapy versus non-responders.
Collapse
Affiliation(s)
- Alexandra Snyder
- Dept. of Medicine; 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
| | - Matthew Hellmann
- Dept. of Medicine; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| | - Naiyer Rizvi
- Division of Hematology/Oncology; New York Presbyterian/Columbia University Hospital ; New York, NY USA
| | - Taha Merghoub
- Dept. of Medicine; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| | - Jedd D Wolchok
- Dept. of Medicine; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| |
Collapse
|
6284
|
Pierce RH, Campbell JS, Pai SI, Brody JD, Kohrt HEK. In-situ tumor vaccination: Bringing the fight to the tumor. Hum Vaccin Immunother 2015; 11:1901-9. [PMID: 26055074 PMCID: PMC4635874 DOI: 10.1080/21645515.2015.1049779] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/17/2015] [Accepted: 05/05/2015] [Indexed: 12/14/2022] Open
Abstract
After decades of development in the shadow of traditional cancer treatment, immunotherapy has come into the spotlight. Treatment of metastatic tumors with monoclonal antibodies to T cell checkpoints like programed cell death 1 (PD-1) or its ligand, (PD-L1), have resulted in significant clinical responses across multiple tumor types. However, these therapies fail in the majority of patients with solid tumors, in particular those who lack PD1(+)CD8(+) tumor-infiltrating lymphocytes within their tumors. Intratumoral "in situ vaccination" approaches seek to enhance immunogenicity, generate tumor infiltrating lymophcytes (TIL) and drive a systemic anti-tumor immune response, directed against "unvaccinated," disseminated tumors. Given the emerging picture of intratumoral immunotherapy as safe and capable of delivering systemic efficacy, it is anticipated that these approaches will become integrated into future multi-modality therapy.
Collapse
Affiliation(s)
| | | | - Sara I Pai
- Department of Surgery; Harvard Medical School; Massachusetts General Hospital; Boston, MA USA
| | - Joshua D Brody
- Division of Hematology/Oncology; Icahn School of Medicine at Mount Sinai; Hess Center for Science and Medicine; New York, NY USA
| | - Holbrook EK Kohrt
- Divisions of Hematology and Oncology; Stanford University Center for Clinical Sciences Research; Stanford, CA USA
| |
Collapse
|
6285
|
Abstract
Metastatic disease is responsible for 90% of death from solid tumors. However, only a minority of metastasis-specific targets has been exploited therapeutically, and effective prevention and suppression of metastatic disease is still an elusive goal. In this review, we will first summarize the current state of knowledge about the molecular features of the disease, with particular focus on steps and targets potentially amenable to therapeutic intervention. We will then discuss the reasons underlying the paucity of metastatic drugs in the current oncological arsenal and potential ways to overcome this therapeutic gap. We reason that the discovery of novel promising targets, an increased understanding of the molecular features of the disease, the effect of disruptive technologies, and a shift in the current preclinical and clinical settings have the potential to create more successful drug development endeavors.
Collapse
Affiliation(s)
- Yari Fontebasso
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Steven M Dubinett
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
6286
|
Abstract
Modern cancer genomics has emerged from the combination of the Human Genome Reference, massively parallel sequencing, and the comparison of tumor to normal DNA sequences, revealing novel insights into the cancer genome and its amazing diversity. Recent developments in applying our knowledge of cancer genomics have focused on the utility of these data for clinical applications. The emergent results of this translation into the clinical setting already are changing the clinical care and monitoring of cancer patients.
Collapse
Affiliation(s)
- Elaine Mardis
- McDonnell Genome Institute, Washington University School of Medicine, Campus Box 8501, 4444 Forest Park Avenue, St. Louis, MO, 63108, USA
| |
Collapse
|
6287
|
Jabbour SK, Berman AT, Simone CB. Integrating immunotherapy into chemoradiation regimens for medically inoperable locally advanced non-small cell lung cancer. Transl Lung Cancer Res 2007; 6:113-118. [PMID: 28529894 DOI: 10.21037/tlcr.2017.04.02] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
For patients with inoperable stage II-III non-small cell lung cancer (NSCLC), the backbone of curative intent therapy is concurrent chemoradiotherapy (CRT). As checkpoint inhibitors have shown clinical benefit in the setting of metastatic NSCLC, additional study is necessary to understand their role in patients receiving CRT. When integrating immunotherapy with radiotherapy (RT) for cure, clinicians will need to consider synergy, timing, doses, and safety among the combination of therapies. This article seeks to review data evaluating interactions, temporal sequencing, fractionation, and overlapping toxicity profiles of thoracic chemoradiation and immunotherapy.
Collapse
Affiliation(s)
- Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Abigail T Berman
- Department of Radiation Oncology, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Charles B Simone
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6288
|
Patel SH, Rimner A, Cohen RB. Combining immunotherapy and radiation therapy for small cell lung cancer and thymic tumors. Transl Lung Cancer Res 2007; 6:186-195. [PMID: 28529901 DOI: 10.21037/tlcr.2017.03.04] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent work with immunotherapy has shown promising results with treatment of several solid malignancies, and there are several reports of good systemic responses with the combination of immunotherapy and radiation therapy (RT), most notably in advanced melanoma. Given the rapid increase in the use of checkpoint blockade as well as anti-tumor vaccines, we review here the preclinical rationale and ongoing clinical work in combining immunotherapy with RT for small cell lung cancer (SCLC) and thymic tumors. While there are several reports of promising results with the combination of immunotherapy and conventional systemic treatment, we focus here on the ongoing clinical studies that combine immunotherapy with RT, and highlight the emerging data for this multimodality approach as well as key preclinical and clinical issues that remain to be addressed. With regards to SCLC, trials exploring to the combination of immunotherapy and RT are already ongoing, but clinical studies for this combination in thymoma are lacking.
Collapse
Affiliation(s)
- Suchit H Patel
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Roger B Cohen
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, Philadelphia, USA
| |
Collapse
|
6289
|
Campbell AM, Decker RH. Mini-review of conventional and hypofractionated radiation therapy combined with immunotherapy for non-small cell lung cancer. Transl Lung Cancer Res 2007; 6:220-229. [PMID: 28529904 DOI: 10.21037/tlcr.2017.03.02] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A successful antitumoral response requires immunological activation as well as an antigenic pool capable of stimulating both the innate and the adaptive immune system. Recent advances in immunotherapy have been aimed at boosting the activation status of the innate and adaptive immune system, including cytokine administration, monoclonal antibodies engineered to target high yield elements in oncogenic signaling pathways, cancer vaccines, and checkpoint inhibitors. Herein, we examine the ways that radiation therapy induced cell death provides a pool of stimulus antigen, and draw parallels from the immunobiology of autoimmunity to explore how the immunogenicity of antigen derived from radiation-induced cell death might augment the antitumoral response. We also review basic research into the ability of different radiation dose fractionation schedules to induce an antitumoral response. After a discussion of basic immunotherapeutic principles, we review the published literature in the field of non-small cell lung cancer (NSCLC) and examine the ways that combining radiation and immunotherapy have begun to change the therapeutic terrain. We provide a summary of ongoing clinical trials aimed at combining immunotherapy and radiation therapy in NSCLC while emphasizing the need for identification of biomarkers with predictive power and the assessment of efficacy as a function of fractionation strategy.
Collapse
Affiliation(s)
- Allison M Campbell
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Roy H Decker
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
6290
|
Perales Palacios I, García Campos F, Michaus Oquiñena L, Blanco Guzmán S, Lantero Benedito M. [Isolation of Plesiomonas shigelloides in a case of gastroenteritis]. Rev Clin Esp 1984; 15:353-365. [PMID: 6658089 DOI: 10.1038/s41571-018-0002-6] [Citation(s) in RCA: 371] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|