1
|
Kabirian R, Tredan O, Marmé F, Paoletti X, Eberst L, Lebreton C, La Motte Rouge TD, Sabatier R, Angelergues A, Fabbro M, Gorp TV, Mansi L, Gladieff L, Kaczmarek E, Alexandre J, Grellety T, Favier L, Welz J, Frenel JS, Leary A. TEDOVA: vaccine OSE2101 +/- pembrolizumab as maintenance in platinum-sensitive recurrent ovarian cancer. Future Oncol 2024:1-10. [PMID: 39155847 DOI: 10.1080/14796694.2024.2386922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/29/2024] [Indexed: 08/20/2024] Open
Abstract
Ovarian cancer is a leading cause of death from gynecological cancers worldwide. Platinum-based chemotherapy provides the cornerstone of the medical management. In first line and subsequent relapses, maintenance strategies are offered to prolong intervals between lines of chemotherapy. Current maintenance options involve bevacizumab and poly ADP-ribose polymerase inhibitors, but these lines of therapy can only be used once in the disease course. Patients in first or second platinum sensitive relapse after poly ADP-ribose polymerase inhibitors and bevacizumab represent an area of unmet medical need. This academic sponsored, international Phase II randomized trial is evaluating the combination of a therapeutic cancer vaccine (OSE2101) with anti-PD1 (pembrolizumab) as maintenance therapy, in patients with platinum-sensitive recurrence regardless of number of prior lines and no progression after platinum-based chemotherapy.Clinical Trial Registration: NCT04713514 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Rayan Kabirian
- Department of Medical Oncology, Gustave Roussy, Villejuif, & GINECO, France
| | | | - Frederik Marmé
- Medical Faculty Mannheim, Heidelberg University & AGO Study Group, Germany
| | | | - Lauriane Eberst
- Institut de Cancérologie de Strasbourg Europe, ICANS, Strasbourg, & GINECO, France
| | | | | | - Renaud Sabatier
- Aix-Marseille Univ, CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Department of Medical Oncology, Marseille, & GINECO, France
| | | | - Michel Fabbro
- Institut du Cancer de Montpellier (ICM), Montpellier, & GINECO, France
| | - Toon Van Gorp
- Leuven Cancer Institute, Division of Gynaecological Oncology, University Hospital Leuven, BGOG,Belgium
| | - Laura Mansi
- CHU Besançon - Hôpital jean Minjoz, Besançon, & GINECO, France
| | | | | | - Jérôme Alexandre
- Université de Paris Cité, AP-HP, Hôpital Cochin, Paris, & GINECO, France
| | - Thomas Grellety
- Centre Hospitalier de la Côte Basque, Bayonne, & GINECO, France
| | - Laure Favier
- Centre Georges François Leclerc, Dijon, & GINECO, France
| | - Julia Welz
- Evang. Kliniken Essen-Mitte - ESSEN & AGO Study Group, Germany
| | - Jean-Sébastien Frenel
- Institut de Cancérologie de l'Ouest, Centre René Gauducheau, 44800, Saint Herblain, & GINECO,France
| | - Alexandra Leary
- Department of Medical Oncology, Gustave Roussy, Villejuif, & GINECO, France
| |
Collapse
|
2
|
Besse B, Felip E, Garcia Campelo R, Cobo M, Mascaux C, Madroszyk A, Cappuzzo F, Hilgers W, Romano G, Denis F, Viteri S, Debieuvre D, Galetta D, Baldini E, Razaq M, Robinet G, Maio M, Delmonte A, Roch B, Masson P, Schuette W, Zer A, Remon J, Costantini D, Vasseur B, Dziadziuszko R, Giaccone G. Randomized open-label controlled study of cancer vaccine OSE2101 versus chemotherapy in HLA-A2-positive patients with advanced non-small-cell lung cancer with resistance to immunotherapy: ATALANTE-1. Ann Oncol 2023; 34:920-933. [PMID: 37704166 DOI: 10.1016/j.annonc.2023.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Patients with advanced non-small-cell lung cancer (NSCLC) treated with immune checkpoint blockers (ICBs) ultimately progress either rapidly (primary resistance) or after durable benefit (secondary resistance). The cancer vaccine OSE2101 may invigorate antitumor-specific immune responses after ICB failure. The objective of ATALANTE-1 was to evaluate its efficacy and safety in these patients. PATIENTS AND METHODS ATALANTE-1 was a two-step open-label study to evaluate the efficacy and safety of OSE2101 compared to standard-of-care (SoC) chemotherapy (CT). Patients with human leukocyte antigen (HLA)-A2-positive advanced NSCLC without actionable alterations, failing sequential or concurrent CT and ICB were randomized (2 : 1) to OSE2101 or SoC (docetaxel or pemetrexed). Primary endpoint was overall survival (OS). Interim OS futility analysis was planned as per Fleming design. In April 2020 at the time of interim analysis, a decision was taken to prematurely stop the accrual due to coronavirus disease 2019 (COVID-19). Final analysis was carried out in all patients and in the subgroup of patients with ICB secondary resistance defined as failure after ICB monotherapy second line ≥12 weeks. RESULTS Two hundred and nineteen patients were randomized (139 OSE2101, 80 SoC); 118 had secondary resistance to sequential ICB. Overall, median OS non-significantly favored OSE2101 over SoC {hazard ratio (HR) [95% confidence interval (CI)] 0.86 [0.62-1.19], P = 0.36}. In the secondary resistance subgroup, OSE2101 significantly improved median OS versus SoC [11.1 versus 7.5 months; HR (95% CI) 0.59 (0.38-0.91), P = 0.017], and significantly improved post-progression survival (HR 0.46, P = 0.004), time to Eastern Cooperative Oncology Group (ECOG) performance status deterioration (HR 0.43, P = 0.006) and Quality of Life Questionnaire Core 30 (QLQ-C30) global health status compared to SoC (P = 0.045). Six-month disease control rates and progression-free survival were similar between groups. Grade ≥3 adverse effects occurred in 11.4% of patients with OSE2101 and 35.1% in SoC (P = 0.002). CONCLUSIONS In HLA-A2-positive patients with advanced NSCLC and secondary resistance to immunotherapy, OSE2101 increased survival with better safety compared to CT. Further evaluation in this population is warranted.
Collapse
Affiliation(s)
- B Besse
- Paris-Saclay University, Cancer Medicine Department, Institut Gustave Roussy, Villejuif, France.
| | - E Felip
- Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Barcelona
| | - R Garcia Campelo
- Medical Oncology Department, Complejo Hospitalario Universitario A Coruña, Biomedical Research Institute, INIBIC, A Coruña
| | - M Cobo
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria University Hospitals, IBIMA, Málaga, Spain
| | - C Mascaux
- Pneumology Department, Hôpitaux Universitaires de Strasbourg-Nouvel Hôpital Civil, Strasbourg
| | - A Madroszyk
- Medical Oncology Department, IPC-Institut Paoli-Calmettes, Marseille, France
| | - F Cappuzzo
- Oncology Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - W Hilgers
- Medical Oncology Department, Sainte Catherine Cancer Center, Avignon, France
| | - G Romano
- Medical Oncology Department, Ospedale Vito Fazzi-ASL Lecce, Lecce, Italy
| | - F Denis
- Medical Oncology Department, Institut Inter-Régional de Cancérologie Jean Bernard-Elsan, Le Mans, France
| | - S Viteri
- Medical Oncology Department, Instituto Oncológico Dr. Rosell, Hospital Universitario Dexeus, Grupo Quironsalud, Barcelona, Spain
| | - D Debieuvre
- Pneumology Department, Groupe Hospitalier de la Région Mulhouse Sud Alsace, Mulhouse, France
| | - D Galetta
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari
| | - E Baldini
- Oncology Department, Ospedale San Luca, Lucca, Italy
| | - M Razaq
- Oncology Department, Stephenson Cancer Center, Oklahoma City, USA
| | - G Robinet
- Oncology Department, Centre Hospitalier Régional Universitaire Morvan, Brest, France
| | - M Maio
- Department of Oncology, University of Siena and Center for Immuno-Oncology, University Hospital, Siena
| | - A Delmonte
- Thoracic Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" (IRST), Meldola, Italy
| | - B Roch
- Thoracic Oncology Unit, Montpellier University, University Hospital of Montpellier, Montpellier
| | - P Masson
- Pneumology Department, Centre Hospitalier de Cholet, Cholet, France
| | - W Schuette
- Medical Oncology Department, Hospital Martha-Maria Halle-Doelau, Halle, Germany
| | - A Zer
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - J Remon
- Paris-Saclay University, Cancer Medicine Department, Institut Gustave Roussy, Villejuif, France
| | - D Costantini
- Medical Development Department, OSE Immunotherapeutics, Paris, France
| | - B Vasseur
- Medical Development Department, OSE Immunotherapeutics, Paris, France
| | - R Dziadziuszko
- Oncology and Radiotherapy Department and Early Phase Clinical Trials Centre, Medical University of Gdansk, Gdansk, Poland
| | - G Giaccone
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
| |
Collapse
|
3
|
PSRTTCA: A new approach for improving the prediction and characterization of tumor T cell antigens using propensity score representation learning. Comput Biol Med 2023; 152:106368. [PMID: 36481763 DOI: 10.1016/j.compbiomed.2022.106368] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 10/19/2022] [Accepted: 11/25/2022] [Indexed: 11/27/2022]
Abstract
Despite the arsenal of existing cancer therapies, the ongoing recurrence and new cases of cancer pose a serious health concern that necessitates the development of new and effective treatments. Cancer immunotherapy, which uses the body's immune system to combat cancer, is a promising treatment option. As a result, in silico methods for identifying and characterizing tumor T cell antigens (TTCAs) would be useful for better understanding their functional mechanisms. Although few computational methods for TTCA identification have been developed, their lack of model interpretability is a major drawback. Thus, developing computational methods for the effective identification and characterization of TTCAs is a critical endeavor. PSRTTCA, a new machine learning (ML)-based approach for improving the identification and characterization of TTCAs based on their primary sequences, is proposed in this study. Specifically, we introduce a new propensity score representation learning algorithm that allows one to generate various sets of propensity scores of amino acids, dipeptides, and g-gap dipeptides to be TTCAs. To enhance the predictive performance, optimal sets of variant propensity scores were determined and fed into the final meta-predictor (PSRTTCA). Benchmarking results revealed that PSRTTCA was a more precise and promising tool for the identification and characterization of TTCAs than conventional ML classifiers and existing methods. Furthermore, PSR-derived propensities of amino acids in becoming TTCAs are used to reveal the relationship between TTCAs and their informative physicochemical properties in order to provide insights into TTCA characteristics. Finally, a user-friendly online computational platform of PSRTTCA is publicly available at http://pmlabstack.pythonanywhere.com/PSRTTCA. The PSRTTCA predictor is anticipated to facilitate community-wide efforts in accelerating the discovery of novel TTCAs for cancer immunotherapy and other clinical applications.
Collapse
|
4
|
Ram Kumar PS, Rencilin CF, Sundar K. Emerging nanomaterials for cancer immunotherapy. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy is a unique approach to treat cancer that targets tumours besides triggering the immune cells. It attempts to harness the supremacy and specificity of immune cells for the regression of malignancy. The key strategy of immunotherapy is that it boosts the natural defence and manipulates the immune system at both cellular and molecular levels. Long-lasting anti-tumour response, reduced metastasis, and recurrence can be achieved with immunotherapy than conventional treatments. For example, targeting cytotoxic T-lymphocyte antigen-4 (CTLA4) by monoclonal antibody is reported as an effective strategy against cancer progression in vivo and chimeric antigen receptor (CAR) modified T-cells are known to express a stronger anti-tumour activity. CTLA4 and CAR are, therefore, beneficial in cancer immunotherapy; however, in clinical settings, both are expensive and cause adverse side effects. Nanomaterials have augmented advantages in cancer immunotherapy, besides their utility in effective delivery and diagnostics. In particular, materials based on lipids, polymers, and metals have been sought-after for delivery technologies. Moreover, the surface of nanomaterials can be engineered using ligands, antigens, and antibodies to target immune cells. In this sense, checkpoint inhibitors, cytokines, agonistic antibodies, surface receptors, and engineered T-cells are promising to regulate the immune system against tumours. Therefore, emerging nanomaterials that can be used for the treatment of cancer is the prime focus of this review. The correlation of mode of administration and biodistribution of various nanomaterials is reviewed here. Besides, the acute and chronic side effects and outcome of clinical trials in the context of cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Pandian Sureshbabu Ram Kumar
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Clayton Fernando Rencilin
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Krishnan Sundar
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| |
Collapse
|
5
|
Su Q, Song H, Huang P, Zhang C, Yang J, Kong D, Wang W. Supramolecular co-assembly of self-adjuvanting nanofibrious peptide hydrogel enhances cancer vaccination by activating MyD88-dependent NF-κB signaling pathway without inflammation. Bioact Mater 2021; 6:3924-3934. [PMID: 33997486 PMCID: PMC8080410 DOI: 10.1016/j.bioactmat.2021.03.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/10/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
Peptide vaccine targeting tumor-specific antigens is a promising cancer treatment regimen. However, peptide vaccines are commonly low-immunogenic, leading to suboptimal antitumor T-cell responses. Current peptide vaccination approaches are challenged by the variability of peptide physicochemical characters and vaccine formulations, flexibility, and the broad feasibility. Here, the supramolecular co-assembly of antigen epitope-conjugated peptides (ECPs) targeting CD8 or CD4 T-cell receptors was used to engineer a nanofibrious hydrogel vaccine platform. This approach provided precise and tunable loading of peptide antigens in nanofibers, which notably increased the antigen uptake, cross-presentation, and activation of dendritic cells (DCs). Immunization in mice indicated that the co-assembled peptide hydrogel did not induce local inflammation responses and elicited significantly promoted T-cell immunity by activating the MyD88-dependent NF-κB signaling pathway in DCs. Vaccination of mice using co-assembled peptide vaccine stimulated both enhanced CD8 and CD4 T cells against EG.7-OVA tumors without additional immunoadjuvants or delivery systems, and resulted in a more remarkable cancer immunotherapy efficacy, compared with free peptide vaccine or aluminum-adjuvanted peptide formulation. Altogether, peptide co-assembly demonstrated by three independent pairs of ECPs is a facile, customizable, and chemically defined approach for co-delivering peptide antigens in self-adjuvanting hydrogel vaccines that could induce stronger anticancer T-cell responses. Supramolecular co-assembly is a facile approach for manufacturing peptide vaccine. Peptide vaccine activates DCs through the MyD88-dependent NF-κB signaling pathway. Vaccination of co-assembled peptide hydrogel augments antitumor T-cell responses. Co-assembled peptide vaccine shows therapeutic cancer immunotherapy efficacy.
Collapse
Affiliation(s)
- Qi Su
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Huijuan Song
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Jing Yang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
6
|
Fridman A, Finnefrock AC, Peruzzi D, Pak I, La Monica N, Bagchi A, Casimiro DR, Ciliberto G, Aurisicchio L. An efficient T-cell epitope discovery strategy using in silico prediction and the iTopia assay platform. Oncoimmunology 2021; 1:1258-1270. [PMID: 23243589 PMCID: PMC3518498 DOI: 10.4161/onci.21355] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Functional T-cell epitope discovery is a key process for the development of novel immunotherapies, particularly for cancer immunology. In silico epitope prediction is a common strategy to try to achieve this objective. However, this approach suffers from a significant rate of false-negative results and epitope ranking lists that often are not validated by practical experience. A high-throughput platform for the identification and prioritization of potential T-cell epitopes is the iTopia(TM) Epitope Discovery System(TM), which allows measuring binding and stability of selected peptides to MHC Class I molecules. So far, the value of iTopia combined with in silico epitope prediction has not been investigated systematically. In this study, we have developed a novel in silico selection strategy based on three criteria: (1) predicted binding to one out of five common MHC Class I alleles; (2) uniqueness to the antigen of interest; and (3) increased likelihood of natural processing. We predicted in silico and characterized by iTopia 225 candidate T-cell epitopes and fixed-anchor analogs from three human tumor-associated antigens: CEA, HER2 and TERT. HLA-A2-restricted fragments were further screened for their ability to induce cell-mediated responses in HLA-A2 transgenic mice. The iTopia binding assay was only marginally informative while the stability assay proved to be a valuable experimental screening method complementary to in silico prediction. Thirteen novel T-cell epitopes and analogs were characterized and additional potential epitopes identified, providing the basis for novel anticancer immunotherapies. In conclusion, we show that combination of in silico prediction and an iTopia-based assay may be an accurate and efficient method for MHC Class I epitope discovery among tumor-associated antigens.
Collapse
|
7
|
Khalili JS, Hanson RW, Szallasi Z. In silico prediction of tumor antigens derived from functional missense mutations of the cancer gene census. Oncoimmunology 2021; 1:1281-1289. [PMID: 23243591 PMCID: PMC3518500 DOI: 10.4161/onci.21511] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Antigen-specific immune responses against peptides derived from missense gene mutations have been identified in multiple cancers. The application of personalized peptide vaccines based on the tumor mutation repertoire of each cancer patient is a near-term clinical reality. These peptides can be identified for pre-validation by leveraging the results of massive gene sequencing efforts in cancer. In this study, we utilized NetMHC 3.2 to predict nanomolar peptide binding affinity to 57 human HLA-A and B alleles. All peptides were derived from 5,685 missense mutations in 312 genes annotated as functionally relevant in the Cancer Genome Project. Of the 26,672,189 potential 8-11 mer peptide-HLA pairs evaluated, 0.4% (127,800) display binding affinities < 50 nM, predicting high affinity interactions. These peptides can be segregated into two groups based on the binding affinity to HLA proteins relative to germline-encoded sequences: peptides for which both the mutant and wild-type forms are high affinity binders, and peptides for which only the mutant form is a high affinity binder. Current evidence directs the attention to mutations that increase HLA binding affinity, as compared with cognate wild-type peptide sequences, as these potentially are more relevant for vaccine development from a clinical perspective. Our analysis generated a database including all predicted HLA binding peptides and the corresponding change in binding affinity as a result of point mutations. Our study constitutes a broad foundation for the development of personalized peptide vaccines that hone-in on functionally relevant targets in multiple cancers in individuals with diverse HLA haplotypes.
Collapse
Affiliation(s)
- Jahan S Khalili
- Departments of Melanoma Medical Oncology and Systems Biology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | | | | |
Collapse
|
8
|
Islam MSB, Miah M, Hossain ME, Kibria KMK. A conserved multi-epitope-based vaccine designed by targeting hemagglutinin protein of highly pathogenic avian H5 influenza viruses. 3 Biotech 2020; 10:546. [PMID: 33251084 PMCID: PMC7682764 DOI: 10.1007/s13205-020-02544-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 11/03/2020] [Indexed: 11/29/2022] Open
Abstract
The highly pathogenic avian H5N1 influenza viruses have been recognized as a potential pandemic threat to humans, and to the poultry industry since 1997. H5 viruses consist of a high mutation rate, so universal vaccine designing is very challenging. Here, we describe a vaccinomics approach to design a novel multi-epitope influenza vaccine, based on the highly conserved regions of surface glycoprotein, Hemagglutinin (HA). Initially, the HA protein sequences from Bangladeshi origin were retrieved and aligned by ClustalW. The sequences of 100% conserved regions extracted and analyzed to select the highest potential T-cell and B-cell epitope. The HTL and CTL analyses using IEDB tools showed that DVWTYNAELLVLMEN possesses the highest affinity with MHC class I and II alleles, and it has the highest population coverage. The docking simulation study suggests that this epitope has the potential to interact with both MHC class I and MHC class II. The B-cell epitope prediction provides a potential peptide, GAIAGFIEGGWQGM. We further retrieved HA sequences of 3950 avian and 250 human H5 isolates from several populations of the world, where H5 was an epidemic. Surprisingly, these epitopes are more than 98% conserved in those regions which indicate their potentiality as a conserved vaccine. We have proposed a multi-epitope vaccine using these sequences and assess its stability and potentiality to induce B-cell immunity. In vivo study is necessary to corroborate this epitope as a vaccine, however, setting forth groundwork for wet-lab studies essential to mitigate pandemic threats and provide cross-protection of both avian and humans against H5 influenza viruses.
Collapse
Affiliation(s)
- Md. Shaid Bin Islam
- Department of Biotechnology and Genetic Engineering, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail, 1902 Bangladesh
| | - Mojnu Miah
- Infectious Diseases Division, International Centre for Diarrheal Diseases Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mohammad Enayet Hossain
- Emerging Infections, Infectious Diseases Division, International Centre for Diarrheal Diseases Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - K. M. Kaderi Kibria
- Department of Biotechnology and Genetic Engineering, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail, 1902 Bangladesh
| |
Collapse
|
9
|
Koşaloğlu-Yalçın Z, Lanka M, Frentzen A, Logandha Ramamoorthy Premlal A, Sidney J, Vaughan K, Greenbaum J, Robbins P, Gartner J, Sette A, Peters B. Predicting T cell recognition of MHC class I restricted neoepitopes. Oncoimmunology 2018; 7:e1492508. [PMID: 30377561 PMCID: PMC6204999 DOI: 10.1080/2162402x.2018.1492508] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/31/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
Epitopes that arise from a somatic mutation, also called neoepitopes, are now known to play a key role in cancer immunology and immunotherapy. Recent advances in high-throughput sequencing have made it possible to identify all mutations and thereby all potential neoepitope candidates in an individual cancer. However, most of these neoepitope candidates are not recognized by T cells of cancer patients when tested in vivo or in vitro, meaning they are not immunogenic. Especially in patients with a high mutational load, usually hundreds of potential neoepitopes are detected, highlighting the need to further narrow down this candidate list. In our study, we assembled a dataset of known, naturally processed, immunogenic neoepitopes to dissect the properties that make these neoepitopes immunogenic. The tools to use and thresholds to apply for prioritizing neoepitopes have so far been largely based on experience with epitope identification in other settings such as infectious disease and allergy. Here, we performed a detailed analysis on our dataset of curated immunogenic neoepitopes to establish the appropriate tools and thresholds in the cancer setting. To this end, we evaluated different predictors for parameters that play a role in a neoepitope's immunogenicity and suggest that using binding predictions and length-rescaling yields the best performance in discriminating immunogenic neoepitopes from a background set of mutated peptides. We furthermore show that almost all neoepitopes had strong predicted binding affinities (as expected), but more surprisingly, the corresponding non-mutated peptides had nearly as high affinities. Our results provide a rational basis for parameters in neoepitope filtering approaches that are being commonly used. Abbreviations: SNV: single nucleotide variant; nsSNV: nonsynonymous single nucleotide variant; ROC: receiver operating characteristic; AUC: area under ROC curve; HLA: human leukocyte antigen; MHC: major histocompatibility complex; PD-1: Programmed cell death protein 1; PD-L1 or CTLA-4: cytotoxic T-lymphocyte associated protein 4.
Collapse
Affiliation(s)
- Zeynep Koşaloğlu-Yalçın
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Manasa Lanka
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Angela Frentzen
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Kerrie Vaughan
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Jason Greenbaum
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Paul Robbins
- Surgery Branch, National Cancer Institute, Bethesda, MD, USA
| | - Jared Gartner
- Surgery Branch, National Cancer Institute, Bethesda, MD, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
10
|
Emambux S, Tachon G, Junca A, Tougeron D. Results and challenges of immune checkpoint inhibitors in colorectal cancer. Expert Opin Biol Ther 2018; 18:561-573. [DOI: 10.1080/14712598.2018.1445222] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sheik Emambux
- Department of Medical Oncology, Poitiers University Hospital, Poitiers, France
| | - Gaelle Tachon
- Department of Cancer biology, Poitiers University Hospital, Poitiers, France
- INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, France
- University of Poitiers, Faculty of medicine, Poitiers France
| | - Audelaure Junca
- University of Poitiers, Faculty of medicine, Poitiers France
- Department of Pathology, Poitiers University Hospital, Poitiers, France
| | - David Tougeron
- Department of Medical Oncology, Poitiers University Hospital, Poitiers, France
- University of Poitiers, Faculty of medicine, Poitiers France
- Department of Gastroenterology, Poitiers University Hospital, Poitiers, France
| |
Collapse
|
11
|
Pan Q, Zhang Q, Chu J, Pais R, Liu S, He C, Eko FO. Chlamydia abortus Pmp18.1 Induces IL-1β Secretion by TLR4 Activation through the MyD88, NF-κB, and Caspase-1 Signaling Pathways. Front Cell Infect Microbiol 2017; 7:514. [PMID: 29326885 PMCID: PMC5741698 DOI: 10.3389/fcimb.2017.00514] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/29/2017] [Indexed: 12/16/2022] Open
Abstract
The polymorphic membrane protein D (Pmp18D) is a 160-kDa outer membrane protein that is conserved and plays an important role in Chlamydia abortus pathogenesis. We have identified an N-terminal fragment of Pmp18D (designated Pmp18.1) as a possible subunit vaccine antigen. In this study, we evaluated the vaccine potential of Pmp18.1 by investigating its ability to induce innate immune responses in dendritic cells and the signaling pathway(s) involved in rPmp18.1-induced IL-1β secretion. We next investigated the immunomodulatory impact of VCG, in comparison with the more established Th1-promoting adjuvants, CpG and FL, on rPmp18.1-mediated innate immune activation. Finally, the effect of siRNA targeting TLR4, MyD88, NF-κB p50, and Caspase-1 mRNA in DCs on IL-1β cytokine secretion was also investigated. Bone marrow-derived dendritic cells (BMDCs) were stimulated with rPmp18.1 in the presence or absence of VCG or CpG or FL and the magnitude of cytokines produced was assessed using a multiplex cytokine ELISA assay. Expression of costimulatory molecules and Toll-like receptors (TLRs) was analyzed by flow cytometry. Quantitation of intracellular levels of myeloid differentiation factor 88 (MyD88), nuclear factor kappa beta (NF-κB p50/p65), and Caspase-1 was evaluated by Western immunoblotting analysis while NF-κB p65 nuclear translocation was assessed by confocal microscopy. The results showed DC stimulation with rPmp18.1 provoked the secretion of proinflammatory cytokines and upregulated expression of TLRs and co-stimulatory molecules associated with DC maturation. These responses were significantly (p ≤ 0.001) enhanced by VCG but not CpG or FL. In addition, rPmp18.1 activated the expression of MyD88, NF-κB p50, and Caspase-1 as well as the nuclear expression of NF-κB p65 in treated DCs. Furthermore, targeting TLR4, MyD88, NF-κB p50, and Caspase-1 mRNA in BMDCs with siRNA significantly reduced their expression levels, resulting in decreased IL-1β cytokine secretion, strongly suggesting their involvement in the rPmp18.1-induced IL-1β cytokine secretion. Taken together, these results indicate that C. abortus Pmp18.1 induces IL-1β secretion by TLR4 activation through the MyD88, NF-κB as well as the Caspase-1 signaling pathways and may be a potential C. abortus vaccine candidate. The vaccine potential of Pmp18.1 will subsequently be evaluated in an appropriate animal model, using VCG as an immunomodulator, following immunization and challenge.
Collapse
Affiliation(s)
- Qing Pan
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States.,Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qiang Zhang
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jun Chu
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Roshan Pais
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Shanshan Liu
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Cheng He
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Francis O Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
12
|
Chen Y, Xue SA, Behboudi S, Mohammad GH, Pereira SP, Morris EC. Ex Vivo PD-L1/PD-1 Pathway Blockade Reverses Dysfunction of Circulating CEA-Specific T Cells in Pancreatic Cancer Patients. Clin Cancer Res 2017; 23:6178-6189. [PMID: 28710313 PMCID: PMC5683391 DOI: 10.1158/1078-0432.ccr-17-1185] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/20/2017] [Accepted: 07/10/2017] [Indexed: 12/27/2022]
Abstract
Purpose: Carcinoembryonic antigen (CEA) is a candidate target for cellular immunotherapy of pancreatic cancer. In this study, we have characterized the antigen-specific function of autologous cytotoxic T lymphocytes (CTL) specific for the HLA-A2-restricted peptide, pCEA691-699, isolated from the peripheral T-cell repertoire of pancreatic cancer patients and sought to determine if ex vivo PD-L1 and TIM-3 blockade could enhance CTL function.Experimental Design: CD8+ T-cell lines were generated from peripheral blood mononuclear cells of 18 HLA-A2+ patients with pancreatic cancer and from 15 healthy controls. In vitro peptide-specific responses were evaluated by flow cytometry after staining for intracellular cytokine production and carboxy fluorescein succinimydyl ester cytotoxicity assays using pancreatic cancer cell lines as targets.Results: Cytokine-secreting functional CEA691-specific CTL lines were successfully generated from 10 of 18 pancreatic cancer patients, with two CTL lines able to recognize and kill both CEA691 peptide-loaded T2 cells and CEA+ HLA-A2+ pancreatic cancer cell lines. In the presence of ex vivo PD-L1 blockade, functional CEA691-specific CD8+ T-cell responses, including IFNγ secretion and proliferation, were enhanced, and this effect was more pronounced on Ag-specific T cells isolated from tumor draining lymph nodes.Conclusions: These data demonstrate that CEA691-specific CTL can be readily expanded from the self-restricted T-cell repertoire of pancreatic cancer patients and that their function can be enhanced by PD-L1 blockade. Clin Cancer Res; 23(20); 6178-89. ©2017 AACR.
Collapse
Affiliation(s)
- Yuan Chen
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Shao-An Xue
- Institute of Immunity and Transplantation, University College London, London, United Kingdom.,Genetic Engineering Laboratory, School of Biological and Environmental Engineering, Xi'An University, Xi'An, P. R. China
| | | | - Goran H Mohammad
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Chemistry Department, College of Science, University of Sulaimani, Sulaimanyah, Kurdistan Region, Iraq
| | - Stephen P Pereira
- Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Emma C Morris
- Institute of Immunity and Transplantation, University College London, London, United Kingdom.
| |
Collapse
|
13
|
Winograd EK, Ciesielski MJ, Fenstermaker RA. Novel vaccines for glioblastoma: clinical update and perspective. Immunotherapy 2017; 8:1293-1308. [PMID: 27993092 DOI: 10.2217/imt-2016-0059] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma is the most common primary brain cancer. Aggressive treatment with surgery, radiation therapy and chemotherapy provides limited overall survival benefit. Glioblastomas have a formidable tumor microenvironment that is hostile to immunological effector cells and these cancers produce profound systemic immunosuppression. However, surgical resection of these tumors creates conditions that favor the use of immunotherapeutic strategies. Therefore, extensive surgical resection, when feasible, will remain part of the equation to provide an environment in which active specific immunotherapy has the greatest chance of working. Toward that end, a number of vaccination protocols are under investigation. Vaccines studied to date have produced cellular and humoral antitumor responses, but unequivocal clinical efficacy has yet to be demonstrated. In addition, focus is shifting toward the prospect of therapies involving vaccines in combination with immune checkpoint inhibitors and other immunomodulatory agents so that effector cells remain active against their targets systemically and within the tumor microenvironment.
Collapse
Affiliation(s)
- Evan K Winograd
- Department of Neurosurgery, State University of New York at Buffalo, Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY 14260, USA
| | - Michael J Ciesielski
- Department of Neurosurgery, State University of New York at Buffalo, Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY 14260, USA.,Department of Neurosurgery, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA.,Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Robert A Fenstermaker
- Department of Neurosurgery, State University of New York at Buffalo, Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY 14260, USA.,Department of Neurosurgery, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA.,Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
14
|
Peres LDP, da Luz FAC, Pultz BDA, Brígido PC, de Araújo RA, Goulart LR, Silva MJB. Peptide vaccines in breast cancer: The immunological basis for clinical response. Biotechnol Adv 2015; 33:1868-77. [PMID: 26523780 DOI: 10.1016/j.biotechadv.2015.10.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 10/15/2015] [Accepted: 10/29/2015] [Indexed: 02/07/2023]
Abstract
This review discusses peptide-based vaccines in breast cancer, immune responses and clinical outcomes, which include studies on animal models and phase I, phase I/II, phase II and phase III clinical trials. Peptide-based vaccines are powerful neoadjuvant immunotherapies that can directly target proteins expressed in tumor cells, mainly tumor-associated antigens (TAAs). The most common breast cancer TAA epitopes are derived from MUC1, HER2/neu and CEA proteins. Peptides derived from TAAs could be successfully used to elicit CD8 and CD4 T cell-specific responses. Thus, choosing peptides that adapt to natural variations of human leukocyte antigen (HLA) genes is critical. The most attractive advantage is that the target response is more specific and less toxic than for other therapies and vaccines. Prominent studies on NeuVax - E75 (epitope for HER2/neu and GM-CSF) in breast cancer and DPX-0907 (HLA-A2-TAAs) expressed in breast cancer, ovarian and prostate cancer have shown the efficacy of peptide-based vaccines as neoadjuvant immunotherapy against cancer. Future peptide vaccine strategies, although a challenge to be applied in a broad range of breast cancers, point to the development of degenerate multi-epitope immunogens against multiple targets.
Collapse
Affiliation(s)
- Lívia de Paula Peres
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil.
| | - Felipe Andrés Cordero da Luz
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil
| | - Brunna dos Anjos Pultz
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil
| | - Paula Cristina Brígido
- Laboratório de Tripanossomatídeos, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil
| | | | - Luiz Ricardo Goulart
- Laboratório de Nanobiotecnologia - Universidade Federal de Uberlândia - UFU, (INGEB), Uberlândia, MG, Brazil
| | - Marcelo José Barbosa Silva
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil.
| |
Collapse
|
15
|
Wang L, Xu Y, Luo C, Sun J, Zhang J, Lee MW, Bai A, Chen G, Frenz CM, Li Z, Huang W. MAGEA10 gene expression in non-small cell lung cancer and A549 cells, and the affinity of epitopes with the complex of HLA-A(∗)0201 alleles. Cell Immunol 2015; 297:10-8. [PMID: 26058806 DOI: 10.1016/j.cellimm.2015.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/11/2015] [Accepted: 05/14/2015] [Indexed: 12/15/2022]
Abstract
MAGEA10, a cancer/testis antigens expressed in tumors but not in normal tissues with the exception of testis and placenta, represents an attractive target for cancer immunotherapy. However, suppressive cytoenvironment and requirement of specific HLA-alleles presentation frequently led to immunotherapy failure. In this study MAGEA10 was scarcely expressed in cancer patients, but enhanced by viili polysaccharides, which indicates a possibility of increasing epitopes presentation. Furthermore the correlation of gene expression with methylation, indicated by R(2) value for MAGEA10 that was 3 times higher than the value for other MAGE genes tested, provides an explanation of why MAGEA10 was highly inhibited, this is also seen by Kaplan-Meier analysis because MAGEA10 did not change the patients' lifespan. By using Molecular-Docking method, 3 MAGEA10 peptides were found binding to the groove position of HLA-A(∗)0210 as same as MAGEA4 peptide co-crystallized with HLA-A(∗)0210, which indicates that they could be promising for HLA-A(∗)0201 presentation in immunotherapy.
Collapse
Affiliation(s)
- Likui Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China.
| | - Yuefang Xu
- Department of pharmacy, First Hospital Affiliated to PLA General Hospital, Beijing 100037, PR China.
| | - Cheng Luo
- Department of Biotechnology, University of Tartu, Riia 23, Tartu 51010, Estonia.
| | - Jian Sun
- Department of Chemistry, The University of Hong Kong, Hong Kong.
| | - Jinlu Zhang
- School of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, PR China.
| | - Ming-Wei Lee
- School of Medical Laboratory and Biotechnology, Chung-Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., Taichung City 402, Taiwan.
| | - Aiping Bai
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 02215, USA.
| | - Guanhua Chen
- Department of Chemistry, The University of Hong Kong, Hong Kong.
| | - Christopher M Frenz
- Department of Mathematics and Computer Science, Mercy College, Dobbs Ferry, NY, USA.
| | - Zhengguo Li
- School of Life Science, Chongqing University, Chongqing 400030, China.
| | - Wenlin Huang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China.
| |
Collapse
|
16
|
Lendvai N, Cohen AD, Cho HJ. Beyond consolidation: auto-SCT and immunotherapy for plasma cell myeloma. Bone Marrow Transplant 2015; 50:770-80. [PMID: 25751647 DOI: 10.1038/bmt.2015.5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 12/31/2014] [Indexed: 12/15/2022]
Abstract
Autologous hematopoietic cell transplantation (auto-HCT) is the standard consolidation therapy for plasma cell myeloma patients following induction therapy. Auto-HCT improves disease-free survival (DFS), but is generally not curative. The allogeneic HCT experience demonstrated that T-cell immunotherapy can confer long-term DFS. Preclinical and clinical data indicate that myeloma-associated Ags elicit humoral and cellular immune responses (IRs) in myeloma patients. These findings strongly suggest that the immunotherapeutic strategies, including immune checkpoint inhibitors, therapeutic cancer vaccines and adoptive cellular therapies, are promising avenues of clinical research that may be most applicable in the minimal residual disease state following auto-HCT. These strategies are designed to prime or augment antimyeloma IRs and promote a 'host-vs-myeloma' effect that may result in durable DFS. Innovative clinical trials investigating immune checkpoint inhibitors and cancer vaccines have demonstrated that robust immunity against myeloma-associated Ags can be elicited in the setting of auto-HCT. A diverse array of immunotherapeutic strategies have entered clinical trials in myeloma, including PD-1/PD-L1 inhibitors, DC/myeloma cell fusion vaccines and adoptive chimeric Ag receptor T-cell therapy, and further investigation of combinations of immunologic and pharmaceutical agents are expected in the near future. In this review, we will discuss the preclinical data supporting immunotherapy in auto-HCT for myeloma, clinical investigation of these strategies and the future prospects of immunotherapy in pursuit of the goal of curative therapy.
Collapse
Affiliation(s)
- N Lendvai
- 1] Myeloma Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA [2] Department of Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | - A D Cohen
- Division of Hematology/Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - H J Cho
- Multiple Myeloma Service, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
17
|
Khodadoust MS, Alizadeh AA. Tumor antigen discovery through translation of the cancer genome. Immunol Res 2015; 58:292-9. [PMID: 24718952 DOI: 10.1007/s12026-014-8505-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cancer cells harbor unique mutations that theoretically create corresponding unique tumor-specific antigens. This class of mutated antigens represents an attractive target for cancer immunotherapy, but their identification has been cumbersome. By combining cancer genome sequencing with computational analysis of MHC binding, it is possible to predict and rank all of the possible mutated tumor antigens. This form of antigen screen is being combined with high throughput methods to measure the immune response to each candidate mutated antigen. Using these techniques, it is possible to systematically test each mutated tumor antigens for an associated immune response. Only a small fraction of the putative mutated antigens tested in this manner have been found to elicit an immune response, yet these responses appear to be both robust and durable. It is becoming increasingly clear that these mutated tumor antigens are an important target in the antitumor response. Studies incorporating this approach promise to improve our understanding of the inherent immunogenicity of individual cancers, potentially providing an explanation for the varying clinical responses to novel immunotherapeutic agents.
Collapse
Affiliation(s)
- Michael S Khodadoust
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | | |
Collapse
|
18
|
Fenstermaker RA, Ciesielski MJ. Challenges in the development of a survivin vaccine (SurVaxM) for malignant glioma. Expert Rev Vaccines 2014; 13:377-85. [PMID: 24521310 DOI: 10.1586/14760584.2014.881255] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is growing interest in immunotherapy for malignant gliomas. This interest stems from a number of immunological observations, together with the failure of conventional therapeutic agents to produce broad and clinically meaningful improvements in survival and quality of life. The challenges faced in translating laboratory-based immunological observations to Phase I and II clinical trials for immunotherapy of gliomas are substantial. Nevertheless, as our understanding of the effects of active specific vaccination in glioma patients grows, results support optimism that such methods may eventually prove useful as an adjunctive treatment for these cancers. This paper highlights a number of barriers encountered in the translational development of a survivin-targeted peptide vaccine (SurVaxM) for patients with malignant gliomas.
Collapse
Affiliation(s)
- Robert A Fenstermaker
- Department of Neurosurgery, Roswell Park Cancer Institute and State University of New York School of Medicine and Biomedical Sciences, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | |
Collapse
|
19
|
Lanitis E, Smith JB, Dangaj D, Flingai S, Poussin M, Xu S, Czerniecki BJ, Li YF, Robbins PF, Powell DJ. A human ErbB2-specific T-cell receptor confers potent antitumor effector functions in genetically engineered primary cytotoxic lymphocytes. Hum Gene Ther 2014; 25:730-9. [PMID: 25003657 PMCID: PMC4137348 DOI: 10.1089/hum.2014.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 06/20/2014] [Indexed: 01/10/2023] Open
Abstract
The ErbB2 protein is a member of the tyrosine kinase family of growth factor receptors that is overexpressed in cancers of the breast, ovary, stomach, kidney, colon, and lung, and therefore represents an attractive candidate antigen for targeted cancer immunotherapy. Cytotoxic T lymphocytes specific for various immunogenic ErbB2 peptides have been described, but they often exhibit both poor functional avidity and tumor reactivity. In order to generate potent CD8(+) T cells with specificity for the ErbB2(369-377) peptide, we performed one round of in vitro peptide stimulation of CD8(+) T cells isolated from an HLA-A2(+) patient who was previously vaccinated with autologous dendritic cells pulsed with HLA class I ErbB2 peptides. Using this approach, we enriched highly avid ErbB2-reactive T cells with strong ErbB2-specific, antitumor effector functions. We then stimulated these ErbB2-reactive T cells with ErbB2(+) HLA-A2(+) tumor cells in vitro and sorted tumor-activated ErbB2(369-377) peptide T cells, which allowed for the isolation of a novel T-cell receptor (TCR) with ErbB2(369-377) peptide specificity. Primary human CD8(+) T cells genetically modified to express this ErbB2-specific TCR specifically bound ErbB2(369-377) peptide containing HLA-A2 tetramers, and efficiently recognized target cells pulsed with low nanomolar concentrations of ErbB2(369-377) peptide as well as nonpulsed ErbB2(+) HLA-A2(+) tumor cell lines in vitro. In a novel xenograft model, ErbB2-redirected T cells also significantly delayed progression of ErbB2(+) HLA-A2(+) human tumor in vivo. Together, these results support the notion that redirection of normal T-cell specificity by TCR gene transfer can have potential applications in the adoptive immunotherapy of ErbB2-expressing malignancies.
Collapse
Affiliation(s)
- Evripidis Lanitis
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Jenessa B. Smith
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Denarda Dangaj
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Seleeke Flingai
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Mathilde Poussin
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Shuwen Xu
- Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104
| | - Brian J. Czerniecki
- Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104
| | - Yong F. Li
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Paul F. Robbins
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Daniel J. Powell
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
- Abramson Cancer Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
20
|
Comber JD, Philip R. MHC class I antigen presentation and implications for developing a new generation of therapeutic vaccines. THERAPEUTIC ADVANCES IN VACCINES 2014; 2:77-89. [PMID: 24790732 DOI: 10.1177/2051013614525375] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Major histocompatibility complex class I (MHC-I) presented peptide epitopes provide a 'window' into the changes occurring in a cell. Conventionally, these peptides are generated by proteolysis of endogenously synthesized proteins in the cytosol, loaded onto MHC-I molecules, and presented on the cell surface for surveillance by CD8(+) T cells. MHC-I restricted processing and presentation alerts the immune system to any infectious or tumorigenic processes unfolding intracellularly and provides potential targets for a cytotoxic T cell response. Therefore, therapeutic vaccines based on MHC-I presented peptide epitopes could, theoretically, induce CD8(+) T cell responses that have tangible clinical impacts on tumor eradication and patient survival. Three major methods have been used to identify MHC-I restricted epitopes for inclusion in peptide-based vaccines for cancer: genetic, motif prediction and, more recently, immunoproteomic analysis. Although the first two methods are capable of identifying T cell stimulatory epitopes, these have significant disadvantages and may not accurately represent epitopes presented by a tumor cell. In contrast, immunoproteomic methods can overcome these disadvantages and identify naturally processed and presented tumor associated epitopes that induce more clinically relevant tumor specific cytotoxic T cell responses. In this review, we discuss the importance of using the naturally presented MHC-I peptide repertoire in formulating peptide vaccines, the recent application of peptide-based vaccines in a variety of cancers, and highlight the pros and cons of the current state of peptide vaccines.
Collapse
Affiliation(s)
| | - Ramila Philip
- Immunotope, Inc., Pennsylvania Biotechnology Center, 3805 Old Easton Road, Doylestown, PA 18902, USA
| |
Collapse
|
21
|
|
22
|
Abstract
Recent studies have underlined the close link between immune response and prognosis of patients with colorectal cancer (CRC). Immune response understanding combined with biotechnology progress of the last years has allowed development of immunotherapy strategies in CRC. Immunotherapy strategies are divided in "active" or "passive" strategies (patients immune system stimulation or not) and considering the activation of antigen specific immune response or not. These immunotherapy strategies are well tolerated and induced cellular and humoral response correlated with clinical response. Many monoclonal antibodies targeting signalisation pathways or angiogenic growth factors have demonstrated their efficacy in CRC. Multiple vaccine strategies, using different tumour associated antigens, have demonstrated a biological efficacy but with poor clinical results. Results are more promising in adjuvant setting but need to be confirmed by randomized trials. Adoptive immunotherapy with transfer of tumour associated antigen specific T cell is probably the most promising strategy. Actually, except monoclonal antibodies, immunotherapy is not used in clinical practice in CRC due to the lack of results and absence of standardisation.
Collapse
|
23
|
Schaft N, Coccoris M, Drexhage J, Knoop C, de Vries IJM, Adema GJ, Debets R. An Altered gp100 Peptide Ligand with Decreased Binding by TCR and CD8α Dissects T Cell Cytotoxicity from Production of Cytokines and Activation of NFAT. Front Immunol 2013; 4:270. [PMID: 24027572 PMCID: PMC3762364 DOI: 10.3389/fimmu.2013.00270] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 08/21/2013] [Indexed: 12/14/2022] Open
Abstract
Altered peptide ligands (APLs) provide useful tools to study T cell activation and potentially direct immune responses to improve treatment of cancer patients. To better understand and exploit APLs, we studied the relationship between APLs and T cell function in more detail. Here, we tested a broad panel of gp100280–288 APLs with respect to T cell cytotoxicity, production of cytokines, and activation of Nuclear Factor of Activated T cells (NFAT) by human T cells gene-engineered with a gp100-HLA-A2-specific TCRαβ. We demonstrated that gp100-specific cytotoxicity, production of cytokines, and activation of NFAT were not affected by APLs with single amino acid substitutions, except for an APL with an amino acid substitution at position 3 (APL A3), which did not elicit any T cell response. A gp100 peptide with a double amino acid mutation (APL S4S6) elicited T cell cytotoxicity and production of IFNγ, and to a lesser extent TNFα, IL-4, and IL-5, but not production of IL-2 and IL-10, or activation of NFAT. Notably, T cell receptor (TCR)-mediated functions showed decreases in sensitivities for S4S6 versus gp100 wild-type (wt) peptide, which were minor for cytotoxicity but at least a 1000-fold more prominent for the production of cytokines. TCR-engineered T cells did not bind A3-HLA-A2, but did bind S4S6-HLA-A2 although to a lowered extent compared to wt peptide-HLA-A2. Moreover, S4S6-induced T cell function demonstrated an enhanced dependency on CD8α. Taken together, most gp100 APLs functioned as agonists, but A3 and S4S6 peptides acted as a null ligand and partial agonist, respectively. Our results further suggest that TCR-mediated cytotoxicity can be dissected from production of cytokines and activation of NFAT, and that the agonist potential of peptide mutants relates to the extent of binding by TCR and CD8α. These findings may facilitate the design of APLs to advance the study of T cell activation and their use for therapeutic applications.
Collapse
Affiliation(s)
- Niels Schaft
- Laboratory of Experimental Tumor Immunology, Department Medical Oncology, Erasmus MC Cancer Institute , Rotterdam , Netherlands
| | | | | | | | | | | | | |
Collapse
|
24
|
Buhrman JD, Slansky JE. Improving T cell responses to modified peptides in tumor vaccines. Immunol Res 2013; 55:34-47. [PMID: 22936035 DOI: 10.1007/s12026-012-8348-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Immune recognition and elimination of cancerous cells is the primary goal of cancer immunotherapy. However, obstacles including immune tolerance and tumor-induced immunosuppression often limit beneficial immune responses. Vaccination is one proposed intervention that may help to overcome these issues and is an active area of study in cancer immunotherapy. Immunizing with tumor antigenic peptides is a promising, straight-forward vaccine strategy hypothesized to boost preexisting antitumor immunity. However, tumor antigens are often weak T cell agonists, attributable to several mechanisms, including immune self-tolerance and poor immunogenicity of self-derived tumor peptides. One strategy for overcoming these mechanisms is vaccination with mimotopes, or peptide mimics of tumor antigens, which alter the antigen presentation and/or T cell activation to increase the expansion of tumor-specific T cells. Evaluation of mimotope vaccine strategies has revealed that even subtle alterations in peptide sequence can dramatically alter antigen presentation and T cell receptor recognition. Most of this research has been performed using T cell clones, which may not be accurate representations of the naturally occurring antitumor response. The relationship between clones generated after mimotope vaccination and the polyclonal T cell repertoire is unclear. Our work with mimotopes in a mouse model of colon carcinoma has revealed important insights into these issues. We propose that the identification of mimotopes based on stimulation of the naturally responding T cell repertoire will dramatically improve the efficacy of mimotope vaccination.
Collapse
Affiliation(s)
- Jonathan D Buhrman
- Integrated Department of Immunology, University of Colorado School of Medicine, National Jewish Health, Denver, CO 80206, USA
| | | |
Collapse
|
25
|
Abstract
Immune response has a crucial role in the control of tumoral progression in colorectal cancer (CRC). A close link between the rate of tumor infiltrating lymphocytes and prognosis has been found. Indeed, recent studies have shattered our dogmas as the rate of tumor infiltrating lymphocytes seems to be a better prognostic factor than TNM classification. This review is focused on immune response in CRC. Specific cell-mediated immunity is important for the control of tumoral growth. Specific T cell activation is induced by tumoral antigens process by dendritic cells. Among the numerous tumoral antigens that could induce an immune response in CRC, the carcinoembryonic antigen is the most studied but its immunogenicity remains low. In CRC with microsatellite instability, several immunogenic neo-antigens have been identified and could explain the high level of tumor infiltrating lymphocytes and the better prognosis of this type of tumour. However, CRC can escape immune response by immune response modulation or tumoral cells modifications conducting to immune resistance. These data should be considered for the treatment of CRC or the development of immunotherapy strategies.
Collapse
|
26
|
|
27
|
Wu X, Xu X, Gu R, Wang Z, Chen H, Xu K, Zhang M, Hutton J, Yang T. Prediction of HLA class I-restricted T-cell epitopes of islet autoantigen combined with binding and dissociation assays. Autoimmunity 2012; 45:176-85. [PMID: 22260783 DOI: 10.3109/08916934.2011.622014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Identification of cognate peptides recognized by human leucocyte antigen (HLA)/T cell receptor (TCR) complex provides insight into the pathogenic process of type 1 diabetes (T1D). We hypothesize that HLA-binding assays alone are inadequate metrics for the affinity of peptides. Zinc transporter-8 (ZnT8) has emerged in recent years as a novel, major, human autoantigen. Therefore, we aim to identify the HLA-A2-restricted ZnT8 epitopes using both binding and dissociation assays. HLA class I peptide affinity algorithms were used to predict candidate ZnT8 peptides that bind to HLA-A2. We analyzed 15 reported epitopes of seven β-cell candidate autoantigens and eight predicted candidate ZnT8 peptides using binding and dissociation assays. Using IFN-γ ELISpot assay, we tested peripheral blood mononuclear cells (PBMCs) from recent-onset T1D patients and healthy controls for reactivity to seven reported epitopes and eight candidate ZnT8 peptides directly ex vivo. We found five of seven recently reported epitopes in Chinese T1D patients. Of the eight predicted ZnT8 peptides, ZnT8(153-161) had a strong binding affinity and the lowest dissociation rate to HLA-A*0201. We identified it as a novel HLA-A*0201-restricted T-cell epitope in three of eight T1D patients. We conclude that ZnT8(153-161) is a novel HLA-A*0201-restricted T-cell epitope. We did not observe a significant correlation (P = 0.3, R = - 0.5) between cytotoxic T cell (CTL) response and peptide/HLA*0201 complex stability. However, selection of peptides based on affinity and their dissociation rate may be helpful for the identification of candidate CTL epitopes. Thus, we can minimize the number of experiments for the identification of T-cell epitopes from interesting antigens.
Collapse
Affiliation(s)
- Xiangmei Wu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Identification of HLA-A∗02:01-restricted CTL epitopes in Trypanosoma cruzi heat shock protein-70 recognized by Chagas disease patients. Microbes Infect 2011; 13:1025-32. [PMID: 21704723 DOI: 10.1016/j.micinf.2011.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 05/12/2011] [Accepted: 05/24/2011] [Indexed: 02/07/2023]
Abstract
CD8(+) cytotoxic T lymphocyte (CTL) response is critical for controlling the infection of the protozoan parasite Trypanosoma cruzi, the causative agent of Chagas disease. Since only a few CD8 antigens have been described in Chagas disease patients, the identification of new class I-restricted epitopes is urgently needed for the development of immunotherapies against T. cruzi infection. In this study, bioinformatic methods were used to predict HLA-A∗02:01-binders, and 30 peptides were selected, synthesized and tested for HLA-A∗02:01 binding. Among them, sixteen peptides with medium-to-high affinity were assayed for their recognition by CTL from HSP70-immunized or T. cruzi-infected transgenic B6-A2/K(b) mice. Our results show that four immunodominant epitopes (HSP70(210-8), HSP70(255-63), HSP70(316-24) and HSP70(345-53)) are contained in the T. cruzi HSP70 antigen. Indeed two of them (HSP70(210-8) and HSP70(316-24)) were also recognized by CTL of HLA-A∗02:01(+) Chagas disease patients, indicating that these peptides are processed and displayed as MHC class I epitopes during the natural history of T. cruzi infection. The HLA-A∗02:01 restriction was evidenced using peptide-pulsed K562-A2 cells as antigen-presenting cells. Both cytotoxic and cytokine-secreting activities were detected in response to the former two peptides and, moreover, 10/12 patients (83%) recognized at least one of these two HSP70-derived CD8(+) epitopes.
Collapse
|
29
|
Abstract
Seasonal influenza is a common and highly transmissible disease, characterized by frequent and unpredictable mutations occurring in the viral envelope glycoproteins. Owing to this high variability, annual reformulation and immunization are required and still, the vaccine is not effective enough when there is an antigenic mismatch with circulating strains. A solution could come from the construction of a universal vaccine that would be based on highly conserved antigens and would be effective against many strains: some universal vaccine developers focus on the Matrix 2 protein, whereas others use additional conserved proteins, such as the nucleoprotein and Matrix 1, or even a range of peptides from these proteins and others to induce cross-strain immunity. This article aims to highlight recent significant advances in the development of a universal vaccine against influenza and focuses mainly on studies using the epitope-based approach that have also entered the clinical trial stage; it includes a brief summary of current vaccines against influenza as well as the ongoing efforts to develop a universal vaccine.
Collapse
Affiliation(s)
- Tamar Ben-Yedidia
- BiondVax Pharmaceuticals Ltd, 14 Einstein Street, Ness Ziona, Israel
| |
Collapse
|
30
|
Miles KM, Miles JJ, Madura F, Sewell AK, Cole DK. Real time detection of peptide-MHC dissociation reveals that improvement of primary MHC-binding residues can have a minimal, or no, effect on stability. Mol Immunol 2010; 48:728-32. [PMID: 21130497 PMCID: PMC3032881 DOI: 10.1016/j.molimm.2010.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 11/01/2010] [Accepted: 11/01/2010] [Indexed: 12/02/2022]
Abstract
The majority of known major histocompatibility complex class I (MHCI)-associated tumor-derived peptide antigens do not contain an optimal motif for MHCI binding. As a result, anchor residue-modified ‘heteroclitic’ peptides have been widely used in therapeutic cancer vaccination trials in order to enhance immune responsiveness. In general, the improved stability of these heteroclitic complexes has been inferred from their improved immunogenicity but has not been formally assessed. Here, we investigated the binding of 4 HLA A*0201-restricted tumor-derived peptides and their commonly used heteroclitic variants. We utilized a cell surface binding assay and a novel robust method for testing the durability of soluble recombinant pMHCI in real time by surface plasmon resonance. Surprisingly, we show that heteroclitic peptides designed with optimal MHC binding motifs do not always form pMHCs that are substantially more stable than their wildtype progenitors. These findings, combined with our recent discovery that TCRs can distinguish between wildtype peptides and those altered at a primary buried MHC anchor residue, suggest that altered TCR binding may account for a large part of the increased immune response that can be generated by anchor residue-modified ligands. Our results further highlight the fact that heteroclitic peptide-based immune interventions require careful evaluation to ensure that wildtype antigen specificity is maintained in vivo.
Collapse
Affiliation(s)
- Kim M. Miles
- Cardiff University, School of Medicine, Heath Park, Cardiff, UK
| | - John J. Miles
- Cardiff University, School of Medicine, Heath Park, Cardiff, UK
- Cellular Immunology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
| | - Florian Madura
- Cardiff University, School of Medicine, Heath Park, Cardiff, UK
| | | | - David K. Cole
- Cardiff University, School of Medicine, Heath Park, Cardiff, UK
- Corresponding author at: Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park, Cardiff CF14 4XN, UK. Tel.: +442920687006; fax: +4402920687007.
| |
Collapse
|
31
|
Abstract
BACKGROUND Harnessing the immune response in treating breast cancer would potentially offer a less toxic, more targeted approach to eradicating residual disease. Breast cancer vaccines are being developed to effectively train cytotoxic T cells to recognize and kill transformed cells while sparing normal ones. However, achieving this goal has been problematic due to the ability of established cancers to suppress and evade the immune response. METHODS A review of the literature on vaccines and breast cancer treatment was conducted, specifically addressing strategies currently available, as well as appropriate settings, paradigms for vaccine development and response monitoring, and challenges with immunosuppression. RESULTS Multiple issues need to be addressed in order to optimize the benefits offered by breast cancer vaccines. Primary issues include the following: (1) cancer vaccines will likely work better in a minimal residual disease state, (2) clinical trial design for immunotherapy should incorporate recommendations from expert groups such as the Cancer Vaccine Working Group and use standardized immune response measurements, (3) the presently available cancer vaccine approaches, including dendritic cell-based, tumor-associated antigen peptide-based, and whole cell-based, have various pros and cons, (4) to date, no one approach has been shown to be superior to another, and (5) vaccines will need to be combined with immunoregulatory agents to overcome tumor-related immunosuppression. CONCLUSIONS Combining a properly optimized cancer vaccine with novel immunomodulating agents that overcome tumor-related immunosuppression in a well-designed clinical trial offers the best hope for developing an effective breast cancer vaccine strategy.
Collapse
Affiliation(s)
- Hatem Soliman
- Department of Women's Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
32
|
Cole DK, Edwards ESJ, Wynn KK, Clement M, Miles JJ, Ladell K, Ekeruche J, Gostick E, Adams KJ, Skowera A, Peakman M, Wooldridge L, Price DA, Sewell AK. Modification of MHC anchor residues generates heteroclitic peptides that alter TCR binding and T cell recognition. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:2600-10. [PMID: 20639478 PMCID: PMC3024538 DOI: 10.4049/jimmunol.1000629] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Improving T cell Ags by altering MHC anchor residues is a common strategy used to enhance peptide vaccines, but there has been little assessment of how such modifications affect TCR binding and T cell recognition. In this study, we use surface plasmon resonance and peptide-MHC tetramer binding at the cell surface to demonstrate that changes in primary peptide anchor residues can substantially and unpredictably alter TCR binding. We also demonstrate that the ability of TCRs to differentiate between natural and anchor-modified heteroclitic peptides distinguishes T cells that exhibit a strong preference for either type of Ag. Furthermore, we show that anchor-modified heteroclitic peptides prime T cells with different TCRs compared with those primed with natural Ag. Thus, vaccination with heteroclitic peptides may elicit T cells that exhibit suboptimal recognition of the intended natural Ag and, consequently, impaired functional attributes in vivo. Heteroclitic peptide-based immune interventions therefore require careful evaluation to ensure efficacy in the clinic.
Collapse
MESH Headings
- Amino Acid Sequence
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Cytokines/immunology
- Cytokines/metabolism
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Flow Cytometry
- HLA-A Antigens/genetics
- HLA-A Antigens/immunology
- HLA-A Antigens/metabolism
- HLA-A2 Antigen
- Humans
- Mutation
- Oligopeptides/genetics
- Oligopeptides/immunology
- Oligopeptides/metabolism
- Peptide Library
- Protein Binding/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Surface Plasmon Resonance
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- David K Cole
- Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Targeting the epidermal growth factor receptor (HER) family by T cell receptor gene-modified T lymphocytes. J Mol Med (Berl) 2010; 88:1113-21. [PMID: 20700725 DOI: 10.1007/s00109-010-0660-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 06/18/2010] [Accepted: 07/13/2010] [Indexed: 10/19/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) has been successfully targeted as a breast cancer-associated antigen by various strategies. HER2 is also overexpressed in other solid tumors such as stomach cancer, as well as in hematological malignancies such as acute lymphoblastic leukemia. HER2-targeted therapies are currently under clinical investigation for a panel of malignancies. In this study, we isolated the T cell receptor (TCR) genes of a HER2-reactive allo-human leukocyte antigen-A2-restricted CTL clone and introduced the TCRα- and β-chain genes into the retrovirus vector MP71. Murinization and codon optimization of the HER2-reactive TCR was required for efficient TCR expression in primary human T cells. The tumor recognition efficiency of HER2-TCR gene-modified T cells was similar to the parental CTL clone from which the TCR genes were isolated. The known cross-reactivity of the HER2-reactive TCR with HER3 and HER4 was retained when the TCR was transduced into primary T cells. Our results could contribute to the development of a TCR-based approach for the treatment of HER2-positive breast cancer, as well as of other malignancies expressing HER2, HER3, and/or HER4.
Collapse
|
34
|
Singh R, Rothman AL, Potts J, Guirakhoo F, Ennis FA, Green S. Sequential immunization with heterologous chimeric flaviviruses induces broad-spectrum cross-reactive CD8+ T cell responses. J Infect Dis 2010; 202:223-33. [PMID: 20536361 PMCID: PMC2903744 DOI: 10.1086/653486] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Flavivirus vaccines based on ChimeriVax technology contain the nonstructural genes of the yellow fever vaccine and the premembrane and envelope genes of heterologous flaviviruses, such as Japanese encephalitis and West Nile viruses. These chimeric vaccines induce both humoral and cell-mediated immunity. Mice were vaccinated with yellow fever, chimeric Japanese encephalitis virus (YF/JE), or chimeric West Nile virus (YF/WN) vaccines, followed by a secondary homologous or heterologous vaccination; the hierarchy and function of CD8(+) T cell responses to a variable envelope epitope were then analyzed and compared with those directed against a conserved immunodominant yellow fever virus NS3 epitope. Sequential vaccination with heterologous chimeric flaviviruses generated a broadly cross-reactive CD8(+) T cell response dependent on both the sequence of infecting viruses and epitope variant. The enhanced responses to variant epitopes after heterologous vaccination were not related to preexisting antibody or to higher virus titers. These results demonstrate that the sequence of vaccination affects the expansion of cross-reactive CD8(+) T cells after heterologous chimeric flavivirus challenge.
Collapse
Affiliation(s)
- Rekha Singh
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Alan L. Rothman
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - James Potts
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | | | - Francis A. Ennis
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Sharone Green
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| |
Collapse
|
35
|
Antitumor cytotoxic T-cell response induced by a survivin peptide mimic. Cancer Immunol Immunother 2010; 59:1211-21. [PMID: 20422411 DOI: 10.1007/s00262-010-0845-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 03/01/2010] [Indexed: 12/23/2022]
Abstract
Survivin is a tumor-associated antigen with significant potential as a cancer vaccine target. We have identified a survivin peptide mimic containing human MHC class I epitopes and a potential class II ligand that induces a potent antitumor response in C57BL/6 mice with GL261 cerebral gliomas. This peptide is able to elicit both CD8+ CTL and T helper cell responses in C57BL/6 mice. The corresponding region of the human survivin molecule represented by peptide SVN53-67 is 100% homologous to the murine protein, but SVN53-67 is weakly immunogenic in man. We evaluated several amino acid substitutions in putative human MHC I anchor positions in SVN53-67 to identify potential peptide mimics that could provide an enhanced antitumor immune response against human glioma and primary central nervous system lymphoma (PCNSL) cells in culture. We evaluated survivin peptides with predicted binding to human HLA-A*0201 antigen using peptide-loaded dendritic cells from PBMC of patients with these malignancies. One alteration (M57) led to binding to HLA-A*0201 with significantly higher affinity. We compared the ability of autologous dendritic cells loaded with SVN53-67 peptide and SVN53-67/M57 in CTL assays against allomatched and autologous, survivin-expressing, human malignant glioma and PCNSL cells. Both SVN53-67 and SVN53-67/M57 produced CTL-mediated killing of malignant target cells; however, SVN53-67/M57 was significantly more effective than SVN53-67. Thus, SVN53-67/M57 may act as a peptide mimic to induce an enhanced antitumor CTL response in tumor patients. The use of SVN53-67/M57 as a cancer vaccine might have application for cancer vaccine therapy.
Collapse
|
36
|
Aurisicchio L, Ciliberto G. Patented cancer vaccines: the promising leads. Expert Opin Ther Pat 2010; 20:647-60. [DOI: 10.1517/13543771003720483] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
37
|
Karanikas V, Zamanakou M, Soukou F, Kerenidi T, Gourgoulianis KI, Germenis AE. Naturally occurring tumor‐specific CD8
+
T‐cell precursors in individuals with and without cancer. Immunol Cell Biol 2010; 88:575-85. [DOI: 10.1038/icb.2010.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Vaios Karanikas
- Cancer Immunology Unit, Department of Immunology & Histocompatibility, School of Medicine, University of Thessaly Larissa Greece
| | - Maria Zamanakou
- Cancer Immunology Unit, Department of Immunology & Histocompatibility, School of Medicine, University of Thessaly Larissa Greece
| | - Faye Soukou
- Cancer Immunology Unit, Department of Immunology & Histocompatibility, School of Medicine, University of Thessaly Larissa Greece
| | - Theodora Kerenidi
- Department of Respiratory Medicine, School of Medicine, University of Thessaly, University Hospital of Larissa Larissa Greece
| | - Konstantinos I Gourgoulianis
- Department of Respiratory Medicine, School of Medicine, University of Thessaly, University Hospital of Larissa Larissa Greece
| | - Anastasios E Germenis
- Cancer Immunology Unit, Department of Immunology & Histocompatibility, School of Medicine, University of Thessaly Larissa Greece
| |
Collapse
|
38
|
Karyampudi L, Formicola C, Erskine CL, Maurer MJ, Ingle JN, Krco CJ, Wettstein PJ, Kalli KR, Fikes JD, Beebe M, Hartmann LC, Disis ML, Ferrone S, Ishioka G, Knutson KL. A degenerate HLA-DR epitope pool of HER-2/neu reveals a novel in vivo immunodominant epitope, HER-2/neu88-102. Clin Cancer Res 2010; 16:825-34. [PMID: 20103660 DOI: 10.1158/1078-0432.ccr-09-2781] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Over the past two decades, there has been significant interest in targeting HER-2/neu in immune-based approaches for the treatment of HER-2/neu+ cancers. For example, peptide vaccination using a CD8 T cell-activating HER-2/neu epitope (amino acids 369-377) is an approach that is being considered in advanced phase clinical trials. Studies have suggested that the persistence of HER-2/neu-specific CD8 T cells could be improved by incorporating human leukocyte antigen (HLA) class II epitopes in the vaccine. Our goal in this study was to identify broad coverage HLA-DR epitopes of HER-2/neu, an antigen that is highly expressed in a variety of carcinomas. EXPERIMENTAL DESIGN A combination of algorithms and HLA-DR-binding assays was used to identify HLA-DR epitopes of HER-2/neu antigen. Evidence of preexistent immunity in cancer patients against the identified epitopes was determined using IFN-gamma enzyme-linked immunosorbent spot (ELIspot) assay. RESULTS Eighty-four HLA-DR epitopes of HER-2/neu were predicted, 15 of which had high binding affinity for > or =11 common HLA-DR molecules. A degenerate pool of four HLA-DR-restricted 15-amino acid epitopes (p59, p88, p422, and p885) was identified, against which >58% of breast and ovarian cancer patients had preexistent T-cell immunity. All four epitopes are naturally processed by antigen-presenting cells. Hardy-Weinberg analysis showed that the pool is useful in approximately 84% of population. Lastly, in this degenerate pool, we identified a novel in vivo immunodominant HLA-DR epitope, HER-2/neu(88-102) (p88). CONCLUSION The broad coverage and natural immunity to this epitope pool suggests potential usefulness in HER-2/neu-targeting, immune-based therapies such as vaccines.
Collapse
|
39
|
Vaccine Therapy for Lung Cancer. Lung Cancer 2010. [DOI: 10.1007/978-1-60761-524-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
40
|
Abstract
Advances in the understanding of the immunoregulatory functions of dendritic cells (DCs) in animal models and humans have led to their exploitation as anticancer vaccines. Although DC-based immunotherapy has proven clinically safe and efficient to induce tumor-specific immune responses, only a limited number of objective clinical responses have been reported in cancer patients. These relatively disappointing results have prompted the evaluation of multiple approaches to improve the efficacy of DC vaccines. The topic of this review focuses on personalized DC-based anticancer vaccines, which in theory have the potential to present to the host immune system the entire repertoire of antigens harbored by autologous tumor cells. We also discuss the implementation of these vaccines in cancer therapeutic strategies, their limitations and the future challenges for effective immunotherapy against cancer.
Collapse
Affiliation(s)
- Nona Janikashvili
- Department of Pediatrics, Steele Children’s Research Center, Arizona 85724, USA
| | - Nicolas Larmonier
- Department of Pediatrics, Steele Children’s Research Center, Arizona 85724, USA
- Department of Immunobiology, BIO5 Institute & Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Emmanuel Katsanis
- Department of Pediatrics, Steele Children’s Research Center, Arizona 85724, USA
- Department of Immunobiology, BIO5 Institute & Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
- University of Arizona, Department of Pediatrics, 1501 N Campbell Ave, PO Box 245073, Tucson, AZ 85724-85073, USA
| |
Collapse
|
41
|
A Novel Mouse Model for Evaluation and Prediction of HLA-A2-restricted CEA Cancer Vaccine Responses. J Immunother 2009; 32:744-54. [DOI: 10.1097/cji.0b013e3181aee1b6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Seavey MM, Pan ZK, Maciag PC, Wallecha A, Rivera S, Paterson Y, Shahabi V. A novel human Her-2/neu chimeric molecule expressed by Listeria monocytogenes can elicit potent HLA-A2 restricted CD8-positive T cell responses and impact the growth and spread of Her-2/neu-positive breast tumors. Clin Cancer Res 2009; 15:924-32. [PMID: 19188163 DOI: 10.1158/1078-0432.ccr-08-2283] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The aim of this study was to efficiently design a novel vaccine for human Her-2/neu-positive (hHer-2/neu) breast cancer using the live, attenuated bacterial vector Listeria monocytogenes. EXPERIMENTAL DESIGN Three recombinant L. monocytogenes-based vaccines were generated that could express and secrete extracellular and intracellular fragments of the hHer-2/neu protein. In addition, we generated a fourth construct fusing selected portions of each individual fragment that contained most of the human leukocyte antigen (HLA) epitopes as a combination vaccine (L. monocytogenes-hHer-2/neu chimera). RESULTS Each individual vaccine was able to either fully regress or slow tumor growth in a mouse model for Her-2/neu-positive tumors. All three vaccines could elicit immune responses directed toward human leukocyte antigen-A2 epitopes of hHer-2/neu. The L. monocytogenes-hHer-2/neu chimera was able to mimic responses generated by the three separate vaccines and prevent spontaneous outgrowth of tumors in an autochthonous model for Her-2/neu-positive breast cancer, induce tumor regression in transplantable models, and prevent seeding of experimental lung metastases in a murine model for metastatic breast cancer. CONCLUSION This novel L. monocytogenes-hHer-2/neu chimera vaccine proves to be just as effective as the individual vaccines but combines the strength of all three in a single vaccination. These encouraging results support future clinical trials using this chimera vaccine and may be applicable to other cancer types expressing the Her-2/neu molecule such as colorectal and pancreatic cancer.
Collapse
Affiliation(s)
- Matthew M Seavey
- Department of Microbiology, University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Adar Y, Singer Y, Levi R, Tzehoval E, Perk S, Banet-Noach C, Nagar S, Arnon R, Ben-Yedidia T. A universal epitope-based influenza vaccine and its efficacy against H5N1. Vaccine 2009; 27:2099-107. [DOI: 10.1016/j.vaccine.2009.02.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 01/25/2009] [Accepted: 02/02/2009] [Indexed: 01/21/2023]
|
44
|
Shepard HM, Brdlik CM, Schreiber H. Signal integration: a framework for understanding the efficacy of therapeutics targeting the human EGFR family. J Clin Invest 2009; 118:3574-81. [PMID: 18982164 DOI: 10.1172/jci36049] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The human EGFR (HER) family is essential for communication between many epithelial cancer cell types and the tumor microenvironment. Therapeutics targeting the HER family have demonstrated clinical success in the treatment of diverse epithelial cancers. Here we propose that the success of HER family-targeted monoclonal antibodies in cancer results from their ability to interfere with HER family consolidation of signals initiated by a multitude of other receptor systems. Ligand/receptor systems that initiate these signals include cytokine receptors, chemokine receptors, TLRs, GPCRs, and integrins. We further extrapolate that improvements in cancer therapeutics targeting the HER family are likely to incorporate mechanisms that block or reverse stromal support of malignant progression by isolating the HER family from autocrine and stromal influences.
Collapse
|
45
|
Vojdani A. Antibodies as predictors of complex autoimmune diseases and cancer. Int J Immunopathol Pharmacol 2008; 21:553-66. [PMID: 18831922 DOI: 10.1177/039463200802100308] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The pathologic role of autoantibodies in many autoimmune diseases is widely accepted. An enzyme immunoassay was used for measurement of antibodies against disease-specific antigens and etiologic agents for cross-reactive antigens associated with them. This antibody assay was applied to a panel of antigens for the detection of different neuroautoimmune diseases that included multiple sclerosis, motor peripheral neuropathies, multifocal motor neuropathy, amyotrophic lateral sclerosis, pediatric autoimmune neuropsychiatric disorder associated with streptococcal infection. We studied women with pregnancies complicated by neural tube defect, neuroborreliosis, autism and patients with possible somatic hypermutation. Antibodies were also measured against antigens and etiologic agents associated with primary biliary cirrhosis and chronic obstructive pulmonary disease. And, finally, antibodies were measured against several tumor antigens or peptides which are expressed in prostatic, breast and colon tissues. This panel of different autoantibodies was applied to 290 patients with neuroautoimmune disorders, cancer, and possible somatic hypermutation. The levels of these antibodies against different tissue-specific antigens and etiologic agents associated with them were significantly elevated in patients versus controls. We hope that this novel 96 antigen-specific ELISA will be used in additional studies that will prove its clinical efficacy, not only for the early diagnosis of many neuroautoimmune, liver and lung autoimmune disorders, but also for prognosis and the implementation of preventive steps for many complex diseases.
Collapse
Affiliation(s)
- A Vojdani
- Immunosciences Lab., Inc., Beverly Hills, CA, USA.
| |
Collapse
|
46
|
Barve M, Bender J, Senzer N, Cunningham C, Greco FA, McCune D, Steis R, Khong H, Richards D, Stephenson J, Ganesa P, Nemunaitis J, Ishioka G, Pappen B, Nemunaitis M, Morse M, Mills B, Maples PB, Sherman J, Nemunaitis JJ. Induction of Immune Responses and Clinical Efficacy in a Phase II Trial of IDM-2101, a 10-Epitope Cytotoxic T-Lymphocyte Vaccine, in Metastatic Non–Small-Cell Lung Cancer. J Clin Oncol 2008; 26:4418-25. [DOI: 10.1200/jco.2008.16.6462] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Generation of broad cytotoxic T-lymphocyte responses against multiple epitopes and tumor-associated antigens (TAAs) may provide effective immunotherapy in patients with cancer. We evaluated a single-vial peptide vaccine consisting of nine HLA-A2 supertype-binding epitopes (two native and seven analog epitopes modified for optimal HLA binding or T-cell receptor stimulation) covering five TAAs and the universal helper pan-DR epitope, formulated as a stable emulsion with incomplete Freund's adjuvant (Montanide ISA 51; Seppic SA, Paris, France). The clinical efficacy, safety, and multiepitope immunogenicity of IDM-2101 was evaluated in patients with stage IIIB or IV non–small-cell lung cancer (NSCLC). Patients and Methods A total of 63 patients were enrolled who were positive for HLA-A2. End points included survival, safety, and immune response. IDM-2101 (previously EP-2101) was administered every 3 weeks for the first 15 weeks, then every 2 months through year 1, then quarterly through year 2, for a total of 13 doses. Epitope-specific cytotoxic and helper T-lymphocyte immunogenic responses were measured by the interferon gamma enzyme-linked immunosorbent spot assay. Results No significant adverse events were noted. Low-grade erythema and pain at the injection site were the most common adverse effects. One-year survival in the treated patients was 60%, and median survival was 17.3 months. One complete and one partial response were identified. Survival was longer in patients demonstrating an immune response to epitope peptides (P < .001). Conclusion IDM-2101 was well tolerated, and evidence of efficacy was suggested.
Collapse
Affiliation(s)
- Minal Barve
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - James Bender
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Neil Senzer
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Casey Cunningham
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - F. Anthony Greco
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - David McCune
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Ronald Steis
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Hung Khong
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Donald Richards
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Joe Stephenson
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Prasanthi Ganesa
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Jackie Nemunaitis
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Glenn Ishioka
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Beena Pappen
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Michael Nemunaitis
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Michael Morse
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Bonnie Mills
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Phillip B. Maples
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Jeffrey Sherman
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - John J. Nemunaitis
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| |
Collapse
|
47
|
Conrad H, Gebhard K, Krönig H, Neudorfer J, Busch DH, Peschel C, Bernhard H. CTLs Directed against HER2 Specifically Cross-React with HER3 and HER4. THE JOURNAL OF IMMUNOLOGY 2008; 180:8135-45. [DOI: 10.4049/jimmunol.180.12.8135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
48
|
Kobayashi H, Celis E. Peptide epitope identification for tumor-reactive CD4 T cells. Curr Opin Immunol 2008; 20:221-7. [PMID: 18499419 DOI: 10.1016/j.coi.2008.04.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Revised: 03/20/2008] [Accepted: 04/25/2008] [Indexed: 10/22/2022]
Abstract
Because T lymphocytes have the capacity to recognize tumor cells, significant efforts are being devoted towards the development of T cell-based immunotherapy for cancer. Most of this work has centered in the induction of anti-tumor CD8 T cells, which exhibit cytolytic activity towards tumor cells expressing tumor-specific or tumor associated antigens. Unfortunately to this day, T cell-based immunotherapy for cancer remains suboptimal. One of the possible explanations is that these immunotherapies have ignored the role that CD4 T helper lymphocytes play in the generation and persistence of CD8 T cell responses. Thus, we believe that in order to obtain clinical benefits T cell-based immunotherapy must stimulate both CD8 and CD4 tumor-reactive T cell responses. During the past seven years our group has focused on the identification of CD4 T cell epitopes from tumor-associated and tumor-specific antigens that could be used to complement the already identified CD8 T cell epitopes to produce effective vaccination strategies against numerous tumor types. We will describe here the strategy we used that resulted in the identification and characterization of numerous CD4 T cell epitopes that are applicable to developing therapies against hematological malignancies and solid tumors.
Collapse
Affiliation(s)
- Hiroya Kobayashi
- Department of Pathology, Asahikawa Medical College, Asahikawa, Japan
| | | |
Collapse
|
49
|
Nemunaitis JJ. Are Vaccines Making a Comeback in Non–Small-Cell Lung Cancer? J Clin Oncol 2008; 26:1402-3. [DOI: 10.1200/jco.2007.15.2843] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- John J. Nemunaitis
- Mary Crowley Cancer Research Centers; Texas Oncology PA; and Sammons Cancer Center, Baylor University Medical Center, Dallas, TX
| |
Collapse
|
50
|
Hou Y, Kavanagh B, Fong L. Distinct CD8+ T cell repertoires primed with agonist and native peptides derived from a tumor-associated antigen. THE JOURNAL OF IMMUNOLOGY 2008; 180:1526-34. [PMID: 18209048 DOI: 10.4049/jimmunol.180.3.1526] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Heteroclitic peptides are used to enhance the immunogenicity of tumor-associated Ags to break T cell tolerance to these self-proteins. One such altered peptide ligand (Cap1-6D) has been derived from an epitope in human carcinoembryonic Ag, CEA(605-613) (Cap1). Clinical responses have been seen in colon cancer patients receiving a tumor vaccine comprised of this altered peptide. Whether Cap1-6D serves as a T cell agonist for Cap1-specific T cells or induces different T cells is unknown. We, therefore, examined the T cell repertoires elicited by Cap1-6D and Cap1. Human CTL lines and clones were generated with either Cap1-6D peptide (6D-CTLs) or Cap1 peptide (Cap1-CTLs). The TCR Vbeta usage and functional avidity of the T cells induced in parallel against these target peptides were assessed. The predominant CTL repertoire induced by agonist Cap1-6D is limited to TCR Vbeta1-J2 with homogenous CDR3 lengths. In contrast, the majority of Cap1-CTLs use different Vbeta1 genes and also had diverse CDR3 lengths. 6D-CTLs produce IFN-gamma in response to Cap1-6D peptide with high avidity, but respond with lower avidity to the native Cap1 peptide when compared with the Cap1-CTLs. Nevertheless, 6D-CTLs could still lyse targets bearing the native epitope. Consistent with these functional results, 6D-CTLs possess TCRs that bind Cap-1 peptide/MHC tetramer with higher intensity than Cap1-CTLs but form less stable interactions with peptide/MHC as measured by tetramer decay. These results demonstrate that priming with this CEA-derived altered peptide ligand can induce distinct carcinoembryonic Ag-reactive T cells with different functional capacities.
Collapse
Affiliation(s)
- Yafei Hou
- Division of Hematology/Oncology, Department of Medicine, University of California-San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | | | | |
Collapse
|