51
|
Afridi S, Hoessli DC, Hameed MW. Mechanistic understanding and significance of small peptides interaction with MHC class II molecules for therapeutic applications. Immunol Rev 2017; 272:151-68. [PMID: 27319349 DOI: 10.1111/imr.12435] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Major histocompatibility complex (MHC) class II molecules are expressed by antigen-presenting cells and stimulate CD4(+) T cells, which initiate humoral immune responses. Over the past decade, interest has developed to therapeutically impact the peptides to be exposed to CD4(+) T cells. Structurally diverse small molecules have been discovered that act on the endogenous peptide exchanger HLA-DM by different mechanisms. Exogenously delivered peptides are highly susceptible to proteolytic cleavage in vivo; however, it is only when successfully incorporated into stable MHC II-peptide complexes that these peptides can induce an immune response. Many of the small molecules so far discovered have highlighted the molecular interactions mediating the formation of MHC II-peptide complexes. As potential drugs, these small molecules open new therapeutic approaches to modulate MHC II antigen presentation pathways and influence the quality and specificity of immune responses. This review briefly introduces how CD4(+) T cells recognize antigen when displayed by MHC class II molecules, as well as MHC class II-peptide-loading pathways, structural basis of peptide binding and stabilization of the peptide-MHC complexes. We discuss the concept of MHC-loading enhancers, how they could modulate immune responses and how these molecules have been identified. Finally, we suggest mechanisms whereby MHC-loading enhancers could act upon MHC class II molecules.
Collapse
Affiliation(s)
- Saifullah Afridi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Daniel C Hoessli
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Muhammad Waqar Hameed
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
52
|
Li Q, Liu Q. Noncoding RNAs in Cancer Immunology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 927:243-64. [PMID: 27376738 DOI: 10.1007/978-981-10-1498-7_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cancer immunology is the study of interaction between cancer cells and immune system by the application of immunology principle and theory. With the recent approval of several new drugs targeting immune checkpoints in cancer, cancer immunology has become a very attractive field of research and is thought to be the new hope to conquer cancer. This chapter introduces the aberrant expression and function of noncoding RNAs, mainly microRNAs and long noncoding RNAs, in tumor-infiltrating immune cells, and their significance in tumor immunity. It also illustrates how noncoding RNAs are shuttled between tumor cells and immune cells in tumor microenvironments via exosomes or other microvesicles to modulate tumor immunity.
Collapse
Affiliation(s)
- Qian Li
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107, Yanjiang West Road, Guangzhou, 510120, China
| | - Qiang Liu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107, Yanjiang West Road, Guangzhou, 510120, China.
| |
Collapse
|
53
|
Immunotherapy in colorectal cancer: What have we learned so far? Clin Chim Acta 2016; 460:78-87. [PMID: 27350293 DOI: 10.1016/j.cca.2016.06.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 12/13/2022]
Abstract
After decades of progress based on chemotherapy and targeted agents, patients with metastatic colorectal cancer still have low long-term survival, with more than 500,000 deaths occurring worldwide every year. Recent results showing clinical evidence of efficacy using immunotherapy in other types of tumors, such as melanoma and lung cancer, have also made this a viable therapy for evaluation in colorectal cancer in clinical trials. The development of cancer immunotherapies is progressing quickly, with a variety of technological approaches. This review summarizes the current status of clinical trials testing immunotherapy in colorectal cancer and discusses what has been learned based on previous results. Immunotherapy strategies, such as various models of vaccines, effector-cell therapy and checkpoint inhibitor antibodies, provide protection against progression for a limited subset of patients diagnosed with colorectal cancer. A better understanding of particular immune cell types and pathways in each patient is still needed. These findings will enable the development of novel biomarkers to select the appropriate subset of patients to be treated with a particular immunotherapy, and the tendencies determined from recent results can guide clinical practice for oncologists in this new therapeutic area and in the design of the next round of clinical trials.
Collapse
|
54
|
Hirayama M, Nishimura Y. The present status and future prospects of peptide-based cancer vaccines. Int Immunol 2016; 28:319-28. [PMID: 27235694 DOI: 10.1093/intimm/dxw027] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/20/2016] [Indexed: 12/22/2022] Open
Abstract
Tumor cells commonly express several antigens, such as tumor-associated antigens (TAAs) or mutation-derived antigens (neoantigens), that can be regarded as foreign antigens and elicit anti-tumor immune responses in cancer patients. Various TAAs or neoantigens expressed in cancer cells have been identified and utilized as targets for cancer vaccines. One approach to elicit tumor-specific immune responses is termed peptide-based cancer vaccination; it involves administrating TAAs or neoantigen-derived peptide for treatment of cancers. There have been several forms of peptide-based cancer vaccines depending on which effector cells, such as CTLs or CD4(+) T-helper cells, are targeted to be activated. Many phase I and II clinical trials of peptide-based cancer vaccines using TAA-derived CTL epitopes, T-helper cell epitopes or dendritic cells loaded with TAA-derived peptides for various malignant tumors have been conducted and provide clinical benefits in a small fraction of patients. Nowadays, to improve the efficiency of peptide-based cancer vaccines, combination immunotherapy of peptide-based cancer vaccines with the immune-checkpoint blockade therapies using mAbs specific for CTLA-4, programmed cell death 1 (PD-1), or PD-1 ligand 1 (PD-L1) have been developed for clinical application. Furthermore, along with the recent technological progress in genetic and bioinformatic analysis, it has become easier to identify neoantigens from individual cancer patients. It is expected that peptide-based cancer vaccines targeting neoantigens as a personalized cancer immunotherapy will be developed.
Collapse
Affiliation(s)
- Masatoshi Hirayama
- Department of Immunogenetics and Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto 860-8556, Japan
| | | |
Collapse
|
55
|
Lee SR, Park YK, Shin BA, Park HR. Effects of tumor vaccine expressing Granulocyte-Macrophage Colony Stimulating Factor and interleukin-18 fusion on cancer cells and its possible application for cancer immunotherapy. Cytokine 2016; 89:143-154. [PMID: 26868088 DOI: 10.1016/j.cyto.2016.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 11/19/2022]
Abstract
To access antitumor effects of a combined Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF) and interleukin-18 (IL-18), cDNA fusion of murine GM-CSF and mature IL-18 (GMIL-18) was constructed and transfected in mammalian cells. GMIL-18 fusion protein was highly secreted and displayed bifunctional activities, possessing immune response initiation and cytokine roles, including IFN-γ induction in mouse splenocytes and increased proliferation of GM-CSF-dependent cells, M-NSF-60. The GMIL-18 secreting tumor vaccine was generated and it strongly stimulated differentiation of dendrite cells (DCs) and effusive CD8+ and CD4+ cell infiltration into tumor mice. Moreover, growth of CT26 mouse colon cancer cells was significantly retarded by GMIL-18 (CT26GMIL-18), but not by CT26GM-CSF- or CT26IL-18. The efficiency of prophylactic vaccination was greater than that of therapeutic vaccination in terms of tumor size and its inhibitory role in proliferation. In micrometastasis analysis of tumor models, γ-ray irradiated GMIL-18 tumor vaccine showed a smaller number of liver-meta tumor nodules in mouse liver cells. We concluded that bifunctional GMIL-18 fusion protein could be applied as an immune therapy for cancer treatments.
Collapse
Affiliation(s)
- Sang Rok Lee
- Department of Molecular Medicine, Chonnam National University, Gwangju 501-757, Republic of Korea
| | - Young Kyu Park
- Department of Surgery, Chonnam National University Medical School, Gwangju 501-757, Republic of Korea
| | - Boo Ahn Shin
- Deprtment of Microbiology, Chonnam National University Medical School, Gwangju 501-757, Republic of Korea
| | - Hae-Ryoung Park
- Division of Liberal Arts & Teacher Training, Kwangju Women's University, Gwangju 506-713, Republic of Korea.
| |
Collapse
|
56
|
Kakhi Z, Frisch B, Bourel-Bonnet L, Hemmerlé J, Pons F, Heurtault B. Airway administration of a highly versatile peptide-based liposomal construct for local and distant antitumoral vaccination. Int J Pharm 2015; 496:1047-56. [PMID: 26602294 DOI: 10.1016/j.ijpharm.2015.11.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/13/2015] [Accepted: 11/14/2015] [Indexed: 01/28/2023]
Abstract
With the discovery of tumor-associated antigens such as ErbB2, vaccination is considered as a promising strategy to prevent the development of cancer or treat the existing disease. Among routes of immunization, the respiratory route provides the opportunity to develop non-invasive approach for vaccine delivery. In the current study, this administration route was used in order to investigate the potency of a highly versatile di-epitopic liposomal construct to exhibit local or distant antitumoral efficiency after prophylactic or therapeutic vaccination in mice. Well-characterized liposomes, containing the ErbB2 (p63-71) TCD8(+) and HA (p307-319) TCD4(+) peptide epitopes and the Pam2CAG adjuvant, were formulated and administered into the airway of naïve BALB/c mice. The nanoparticle vaccine candidate induced local and specific systemic immune response, as measured by immune cell infiltration and chemokine and cytokine production in BALF or lung tissue, and by spleen T-cell activation ex vivo, respectively. This potent immune response resulted in an efficient antitumor activity against both lung and solid s.c. tumors. Interestingly, the antitumor efficacy was observed after both prophylactic and therapeutic vaccinations, which are the most judicious ones to fight cancer. Our data showed an undeniable interest of liposomal peptide-based vaccines in antitumor vaccination by the respiratory route, opening new perspectives for cancer treatment.
Collapse
Affiliation(s)
- Zahra Kakhi
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Benoît Frisch
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Line Bourel-Bonnet
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | | | - Françoise Pons
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France.
| | - Béatrice Heurtault
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France.
| |
Collapse
|
57
|
Chai SJ, Yap YY, Foo YC, Yap LF, Ponniah S, Teo SH, Cheong SC, Patel V, Lim KP. Identification of Four-Jointed Box 1 (FJX1)-Specific Peptides for Immunotherapy of Nasopharyngeal Carcinoma. PLoS One 2015; 10:e0130464. [PMID: 26536470 PMCID: PMC4633155 DOI: 10.1371/journal.pone.0130464] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/20/2015] [Indexed: 01/02/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is highly prevalent in South East Asia and China. The poor outcome is due to late presentation, recurrence, distant metastasis and limited therapeutic options. For improved treatment outcome, immunotherapeutic approaches focusing on dendritic and autologous cytotoxic T-cell based therapies have been developed, but cost and infrastructure remain barriers for implementing these in low-resource settings. As our prior observations had found that four-jointed box 1 (FJX1), a tumor antigen, is overexpressed in NPCs, we investigated if short 9-20 amino acid sequence specific peptides matching to FJX1 requiring only intramuscular immunization to train host immune systems would be a better treatment option for this disease. Thus, we designed 8 FJX1-specific peptides and implemented an assay system to first, assess the binding of these peptides to HLA-A2 molecules on T2 cells. After, ELISPOT assays were used to determine the peptides immunogenicity and ability to induce potential cytotoxicity activity towards cancer cells. Also, T-cell proliferation assay was used to evaluate the potential of MHC class II peptides to stimulate the expansion of isolated T-cells. Our results demonstrate that these peptides are immunogenic and peptide stimulated T-cells were able to induce peptide-specific cytolytic activity specifically against FJX1-expressing cancer cells. In addition, we demonstrated that the MHC class II peptides were capable of inducing T-cell proliferation. Our results suggest that these peptides are capable of inducing specific cytotoxic cytokines secretion against FJX1-expressing cancer cells and serve as a potential vaccine-based therapy for NPC patients.
Collapse
Affiliation(s)
- San Jiun Chai
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Yoke Yeow Yap
- Department of Surgery, Clinical Campus Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Yoke Ching Foo
- Department of Oncology, Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Lee Fah Yap
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Sathibalan Ponniah
- Department of Surgery, Cancer Vaccine Development Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Soo Hwang Teo
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Sok Ching Cheong
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Vyomesh Patel
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Kue Peng Lim
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
- * E-mail:
| |
Collapse
|
58
|
Oostvogels R, Lokhorst HM, Mutis T. Minor histocompatibility Ags: identification strategies, clinical results and translational perspectives. Bone Marrow Transplant 2015; 51:163-71. [PMID: 26501766 DOI: 10.1038/bmt.2015.256] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 08/11/2015] [Accepted: 08/15/2015] [Indexed: 12/14/2022]
Abstract
Allogeneic stem cell transplantation (allo-SCT) and donor lymphocyte infusion are effective treatment modalities for various hematological malignancies. Their therapeutic effect, the graft-versus-tumor (GvT) effect, is based mainly on an alloimmune response of donor T cells directed at tumor cells, in which differences in the expression of minor histocompatibility Ags (mHags) on the cells of the patient and donor have a crucial role. However, these differences are also responsible for induction of sometimes detrimental GvHD. As relapse and development of GvHD pose major threats for a large proportion of allotransplanted patients, additional therapeutic strategies are required. To augment the GvT response without increasing the risk of GvHD, specific mHag-directed immunotherapeutic strategies have been developed. Over the past years, much effort has been put into the identification of therapeutically relevant mHags to enable these strategies for a substantial proportion of patients. Currently, the concept of mHag-directed immunotherapy is tested in clinical trials on feasibility, safety and efficacy. In this review, we will summarize the recent developments in mHag identification and the clinical data on mHag-specific immune responses and mHag-directed therapies in patients with hematological malignancies. Finally, we will outline the current challenges and future prospectives in the field.
Collapse
Affiliation(s)
- R Oostvogels
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - H M Lokhorst
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - T Mutis
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
59
|
Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M. Peptide-based vaccines for cancer therapy. Hum Vaccin Immunother 2015; 10:3175-8. [PMID: 25483658 DOI: 10.4161/hv.29418] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Interest for cancer vaccination started more than 30 years ago after the demonstration that both in animal models and later on in patients it is possible to generate anti-tumor immune responses. The clinical application of this knowledge, however, was disappointing. In this review we summarize results on peptides epitopes recognized by T cells that have been studied thanks to their easy synthesis and the lack of significant side effects when administered in-vivo. To improve the clinical efficacy, peptides were modified in their aminoacid sequence to augment their immunogenicity. Peptides vaccines were recently shown to induce a high frequency of immune response in patients that were accompanied by clinical efficacy. These data are discussed at the light of recent progression of immunotherapy caused by the addition of check-point antibodies thus providing a general picture of the potential therapeutic efficacy of the peptide-based vaccines and their combination with other biological agents.
Collapse
|
60
|
Tagliamonte M, Petrizzo A, Tornesello ML, Buonaguro FM, Buonaguro L. Antigen-specific vaccines for cancer treatment. Hum Vaccin Immunother 2015; 10:3332-46. [PMID: 25483639 DOI: 10.4161/21645515.2014.973317] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Vaccines targeting pathogens are generally effective and protective because based on foreign non-self antigens which are extremely potent in eliciting an immune response. On the contrary, efficacy of therapeutic cancer vaccines is still disappointing. One of the major reasons for such poor outcome, among others, is the difficulty of identifying tumor-specific target antigens which should be unique to the tumors or, at least, overexpressed on the tumors as compared to normal cells. Indeed, this is the only option to overcome the peripheral immune tolerance and elicit a non toxic immune response. New and more potent strategies are now available to identify specific tumor-associated antigens for development of cancer vaccine approaches aiming at eliciting targeted anti-tumor cellular responses. In the last years this aspect has been addressed and many therapeutic vaccination strategies based on either whole tumor cells or specific antigens have been and are being currently evaluated in clinical trials. This review summarizes the current state of cancer vaccines, mainly focusing on antigen-specific approaches.
Collapse
Key Words
- APCs, antigen-presenting cell
- BCG, Bacille Calmette-Guerin
- BCR, B-cell receptor
- CDCA1, cell division cycle associated 1
- CRC, colorectal cancer
- CT, Cancer-testis
- CTL, cytotoxic T-lympocites
- DCs, dendritic cells
- EGT, electro-gene-transfer
- FDA, Food & drug administration
- GB, glioblastoma
- GM-CSF, granulocyte macrophage-colony stimulating factor
- HER2, human epidermal growth factor receptor 2
- HLA, human leukocyte antigen
- HPV, human papillomavirus
- HSPs, stress/heat shock proteins
- IFNg, interferon gamma
- Ig Id, immunoglobulin idiotype
- LPs, long peptides
- MAGE-A1, Melanoma-associated antigen 1
- MHC, major histocompatibility complex
- MS, mass spectrometry
- MVA, modified vaccinia strain Ankara
- NSCLC, non-small-cell lung carcinoma
- PAP, prostatic acid phosphatase
- PRRs, Pattern Recognition Receptors
- PSA, Prostate-specific antigen
- RCR, renal cell cancer
- SSX-2, Synovial sarcoma X breakpoint 2
- TAAs, tumor-associated antigens
- TACAs, Tumor-associated carbohydrate antigens
- TARP, T-cell receptor gamma alternate reading frame protein
- TLRs, Toll-Like Receptors
- TPA, transporter associated with antigen processing
- WES, whole exome sequencing
- WGS, whole genome sequencing
- cancer vaccine
- clinical trials
- epitopes
- hTERT, human Telomerase reverse transcriptase
- immunotherapeutics
- mCRPC, metastatic castrate-resistant prostate cancer
- tumor-associated antigens
Collapse
Affiliation(s)
- Maria Tagliamonte
- a Laboratory of Molecular Biology and Viral Oncology; Department of Experimental Oncology; Istituto Nazionale per lo Studio e la Cura dei Tumori; "Fondazione Pascale" - IRCCS ; Naples , Italy
| | | | | | | | | |
Collapse
|
61
|
Peptide-Based Treatment: A Promising Cancer Therapy. J Immunol Res 2015; 2015:761820. [PMID: 26568964 PMCID: PMC4629048 DOI: 10.1155/2015/761820] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/14/2014] [Indexed: 12/16/2022] Open
Abstract
Many new therapies are currently being used to treat cancer. Among these new methods, chemotherapy based on peptides has been of great interest due to the unique advantages of peptides, such as a low molecular weight, the ability to specifically target tumor cells, and low toxicity in normal tissues. In treating cancer, peptide-based chemotherapy can be mainly divided into three types, peptide-alone therapy, peptide vaccines, and peptide-conjugated nanomaterials. Peptide-alone therapy may specifically enhance the immune system's response to kill tumor cells. Peptide-based vaccines have been used in advanced cancers to improve patients' overall survival. Additionally, the combination of peptides with nanomaterials expands the therapeutic ability of peptides to treat cancer by enhancing drug delivery and sensitivity. In this review, we mainly focus on the new advances in the application of peptides in treating cancer in recent years, including diagnosis, treatment, and prognosis.
Collapse
|
62
|
Marino F, Bern M, Mommen GP, Leney AC, van Gaans-van den Brink JA, Bonvin AM, Becker C, van Els CA, Heck AJR. Extended O-GlcNAc on HLA Class-I-Bound Peptides. J Am Chem Soc 2015; 137:10922-10925. [PMID: 26280087 PMCID: PMC4603548 DOI: 10.1021/jacs.5b06586] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report unexpected mass spectrometric observations of glycosylated human leukocyte antigen (HLA) class I-bound peptides. Complemented by molecular modeling, in vitro enzymatic assays, and oxonium ion patterns, we propose that the observed O-linked glycans carrying up to five monosaccharides are extended O-GlcNAc's rather than GalNAc-initiated O-glycans. A cytosolic O-GlcNAc modification is normally terminal and does not extend to produce a polysaccharide, but O-GlcNAc on an HLA peptide presents a special case because the loaded HLA class I complex traffics through the endoplasmic reticulum and Golgi apparatus on its way to the cell membrane and is hence exposed to glycosyltransferases. We also report for the first time natural HLA class I presentation of O- and N-linked glycopeptides derived from membrane proteins. HLA class I peptides with centrally located oligosaccharides have been shown to be immunogenic and may thus be important targets for immune surveillance.
Collapse
Affiliation(s)
- Fabio Marino
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | | | - Geert P.M. Mommen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Institute for Translational Vaccinology, Bilthoven, Netherlands
| | - Aneika C. Leney
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | | | - Alexandre M.J.J. Bonvin
- Computational Structural Biology, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | | | - Cécile A.C.M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, The Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
63
|
Kennedy GT, Judy BF, Bhojnagarwala P, Moon EK, Fridlender ZG, Albelda SM, Singhal S. Surgical cytoreduction restores the antitumor efficacy of a Listeria monocytogenes vaccine in malignant pleural mesothelioma. Immunol Lett 2015; 166:28-35. [PMID: 25999306 DOI: 10.1016/j.imlet.2015.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 04/26/2015] [Accepted: 05/11/2015] [Indexed: 10/23/2022]
Abstract
Recent studies suggest that immunotherapy may offer a promising treatment strategy for early-stage malignant pleural mesothelioma (MPM), but advanced tumor burden may limit the efficacy of immunotherapy. Therefore, we hypothesized that surgical cytoreduction could restore the efficacy of vaccine-based immunotherapy for MPM. We developed a murine model of MPM through transduction of a mesothelioma cell line with mesothelin. We used this model to evaluate the efficacy of a Listeria monocytogenes vaccine expressing mesothelin. Tumor growth was significantly inhibited at four weeks in animals vaccinated two weeks prior to tumor cell inoculation as compared to those given an empty vector control (1371 ± 420 mm(3) versus 405 ± 139 mm(3); p < 0.01). Mice vaccinated one week prior to tumor challenge also displayed significant reduction in tumor volume (1227 ± 406 mm(3) versus 309 ± 173 mm(3); p < 0.01). The vaccine had no effect when administered concurrently with tumor challenge, or after tumors were established. Flow cytometry showed reduced mesothelin expression in large tumors, as well as tumor-associated immunosuppression due to increased myeloid derived suppressor cells (MDSCs). These factors may have limited vaccine efficacy for advanced disease. Surgical cytoreduction of established tumors restored the antitumor potency of the therapeutic vaccine, with significantly reduced tumor burden at post-operative day 18 (397 ± 103 mm(3) versus 1047 ± 258 mm(3); p < 0.01). We found that surgery reduced MDSCs to levels comparable to those in tumor-naïve mice. This study demonstrates that cytoreduction surgery restores the efficacy of cancer vaccines for MPM by reducing tumor-related immunosuppression that impairs immunotherapy.
Collapse
Affiliation(s)
- Gregory T Kennedy
- Division of Thoracic Surgery, Department of Surgery, Hospital of the University of Pennsylvania, 6 White 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Brendan F Judy
- Division of Thoracic Surgery, Department of Surgery, Hospital of the University of Pennsylvania, 6 White 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Pratik Bhojnagarwala
- Division of Thoracic Surgery, Department of Surgery, Hospital of the University of Pennsylvania, 6 White 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Edmund K Moon
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Zvi G Fridlender
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Steven M Albelda
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, Hospital of the University of Pennsylvania, 6 White 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
64
|
Yang H, Kim DS. Peptide Immunotherapy in Vaccine Development: From Epitope to Adjuvant. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 99:1-14. [PMID: 26067814 DOI: 10.1016/bs.apcsb.2015.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vaccines are designed to educate the host immune system to prevent infectious disease or to fight against various diseases such as cancers. Peptides were first employed to provide specific immune responses while minimizing unintended allergenic or reactogenic adverse effects. Discoveries of virus or cancer-specific antigens and the advanced knowledge of immunology accelerate the peptide vaccine development. Despite the overwhelming research pipelines, a very few of them reached to market approvals or phase III clinical trials, because of the lack of efficacy. Several strategies for the next generation peptide vaccines are devised to overcome the weak immunogenicity and the poor delivery. In this review, we discuss the new promising strategies of peptide vaccine development which are recently developed in preclinical and/or clinical stage focusing the roles of peptides in the vaccine formulation from epitope to adjuvant. Additionally, we discuss the future perspectives of peptide vaccine and immunotherapy.
Collapse
Affiliation(s)
- Hyun Yang
- Research and Development Center, Peptron, Inc., Daejeon, South Korea
| | - Dong Seok Kim
- Research and Development Center, Peptron, Inc., Daejeon, South Korea.
| |
Collapse
|
65
|
Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol 2015; 35 Suppl:S185-S198. [PMID: 25818339 DOI: 10.1016/j.semcancer.2015.03.004] [Citation(s) in RCA: 1069] [Impact Index Per Article: 106.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 12/27/2022]
Abstract
Cancer immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Although considerable progress has been made in understanding how cancers evade destructive immunity, measures to counteract tumor escape have not kept pace. There are a number of factors that contribute to tumor persistence despite having a normal host immune system. Immune editing is one of the key aspects why tumors evade surveillance causing the tumors to lie dormant in patients for years through "equilibrium" and "senescence" before re-emerging. In addition, tumors exploit several immunological processes such as targeting the regulatory T cell function or their secretions, antigen presentation, modifying the production of immune suppressive mediators, tolerance and immune deviation. Besides these, tumor heterogeneity and metastasis also play a critical role in tumor growth. A number of potential targets like promoting Th1, NK cell, γδ T cell responses, inhibiting Treg functionality, induction of IL-12, use of drugs including phytochemicals have been designed to counter tumor progression with much success. Some natural agents and phytochemicals merit further study. For example, use of certain key polysaccharide components from mushrooms and plants have shown to possess therapeutic impact on tumor-imposed genetic instability, anti-growth signaling, replicative immortality, dysregulated metabolism etc. In this review, we will discuss the advances made toward understanding the basis of cancer immune evasion and summarize the efficacy of various therapeutic measures and targets that have been developed or are being investigated to enhance tumor rejection.
Collapse
|
66
|
Trajanoski Z, Maccalli C, Mennonna D, Casorati G, Parmiani G, Dellabona P. Somatically mutated tumor antigens in the quest for a more efficacious patient-oriented immunotherapy of cancer. Cancer Immunol Immunother 2015; 64:99-104. [PMID: 25164877 PMCID: PMC11028785 DOI: 10.1007/s00262-014-1599-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/11/2014] [Indexed: 01/23/2023]
Abstract
Although cancer immunotherapy shows efficacy with adoptive T cell therapy (ACT) and antibody-based immune checkpoint blockade, efficacious therapeutic vaccination of cancer patients with tumor-associated antigens (TAAs) remains largely unmet. Current cancer vaccines utilize nonmutated shared TAAs that may have suboptimal immunogenicity. Experimental evidence underscores the strong immunogenicity of unique TAAs derived from somatically mutated cancer proteins, whose massive characterization has been precluded until recently by technical limitations. The development of cost-effective, high-throughput DNA sequencing approaches makes now possible the rapid identification of all the somatic mutations contained in a cancer cell genome. This method, combined with robust bioinformatics platforms for T cell epitope prediction and established reverse immunology approaches, provides us with an integrated strategy to identify patient-specific unique TAAs in a relatively short time, compatible with their potential use in the clinic. Hence, it is now for the first time possible to quantitatively define the patient's unique tumor antigenome and exploit it for vaccination, possibly in combination with ACT and/or immune checkpoint blockade to further increase immunotherapy efficacy.
Collapse
Affiliation(s)
- Zlatko Trajanoski
- Biocenter, Division for Bioinformatics, Innsbruck Medical University, Innrain 80, 6020 Innsbruck, Austria
| | - Cristina Maccalli
- NIBIT Laboratory, Division of Medical Oncology and Immunotherapy, Azienda Ospedaliera Universitaria, Siena, Italy
| | - Daniele Mennonna
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy
| | - Giorgio Parmiani
- Solid Tumor Immuno-Biotherapy Unit, Division of Molecular Oncology, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy
| | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy
| |
Collapse
|
67
|
Parmiani G, Pilla L, Corti A, Doglioni C, Cimminiello C, Bellone M, Parolini D, Russo V, Capocefalo F, Maccalli C. A pilot Phase I study combining peptide-based vaccination and NGR-hTNF vessel targeting therapy in metastatic melanoma. Oncoimmunology 2014; 3:e963406. [PMID: 25941591 DOI: 10.4161/21624011.2014.963406] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/04/2014] [Indexed: 11/19/2022] Open
Abstract
Administration of NGR-TNF, a tumor vessel-targeting and tumor necrosis factor α TNFα) peptide conjugate, with immunotherapy has been shown to inhibit tumor growth in mice. Thus, we planned a Phase I pilot clinical trial to assess safety, immune and clinical response of this combination treatment for advanced melanoma. NA17.A2 and MAGE-3.A1 peptides were used as vaccine. HLA-A*0201 or HLA-A*01 metastatic melanoma patients received human NGR-hTNF i.v. alternating with s.c. weekly injections of either of the peptides emulsified in Montanide. The T-cell response was assessed ex-vivo using peripheral blood mononuclear cells (PBMCs) before, during and after therapy. The serum level of chromogranin A (CgA), soluble TNF receptors (sTNFR1/2), vascular endothelial growth factor (VEGF), and MIP-1β and MCP-1 chemokines, was determined. In 3 subjects, pre- and post-treatment tumor lesions were examined by immunohistochemistry. Clinically, chills were observed in 4 patients during NGR-hTNF infusion and erythema at vaccination site was seen in 7 patients. T-cell response against the vaccine or against other melanoma-associated antigens was detectable after treatment in 6 out of 7 tested patients. Low level or reduction of CgA and sTNFR and increase of MIP-1β and MCP-1 were found in patients sera. In the lesions examined the immune infiltrate was scanty but macrophage number increased in post-therapy lesions. From a clinical standpoint, a long term survival (>4 months) was found in 6 out of 8 evaluable patients (4, 4, 7, 11, 23+, 25+, 25+, 29+ months). The combination of NGR-hTNF and vaccine in metastatic melanoma patients was well tolerated, often associated with an ex-vivo T cell response and long-term overall survival. These findings warrant confirmation in a larger group of patients.
Collapse
Key Words
- APC, antigen presenting cell
- CT, cancer/testis
- CgA, chromogranin A
- DFS, disease-free survival
- MAA, melanoma-associated antigens
- MCP-1, macrophage chemoattractant protein 1
- MIP-1β, macrophage inflammatory protein 1β; OS, overall survival
- PBMC, peripheral blood mononuclear cell
- PD, progression of disease
- PFS, progression-free survival
- RR, response rate
- T cells
- TNFα, tumor necrosis factor α
- anti-vascular target therapy
- combination therapy
- inflammatory cytokines
- melanoma
- peptide-based vaccines
- sTNFR, soluble tumor necrosis factor receptor
Collapse
Affiliation(s)
- Giorgio Parmiani
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Lorenzo Pilla
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Angelo Corti
- Unit of Tumor Biology and Vascular Targeting; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Claudio Doglioni
- Unit of Pathology; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Carolina Cimminiello
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Matteo Bellone
- Unit of Cellular Immunology; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Danilo Parolini
- Unit of Gastrointestinal Surgery; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Vincenzo Russo
- Unit of Cancer Gene Therapy; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Filippo Capocefalo
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Cristina Maccalli
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| |
Collapse
|
68
|
Shaib W, Goldstein D, El-Rayes BF. Peptide Vaccines for Treatment of Colon Cancer: Have We Made Progress? CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0250-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
69
|
Microenvironment of tumor-draining lymph nodes: opportunities for liposome-based targeted therapy. Int J Mol Sci 2014; 15:20209-39. [PMID: 25380524 PMCID: PMC4264163 DOI: 10.3390/ijms151120209] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 02/07/2023] Open
Abstract
The World Health Organization (WHO) recently reported that the total number of global cancer cases in 2013 reached 14 million, a 10% rise since 2008, while the total number of cancer deaths reached 8.2 million, a 5.2% increase since 2008. Metastasis is the major cause of death from cancer, accounting for 90% of all cancer related deaths. Tumor-draining lymph nodes (TDLN), the sentinel nodes, are the first organs of metastasis in several types of cancers. The extent of metastasis in the TDLN is often used in disease staging and prognosis evaluation in cancer patients. Here, we describe the microenvironment of the TDLN and review the recent literature on liposome-based therapies directed to immune cells within the TDLN with the intent to target cancer cells.
Collapse
|
70
|
Humar M, Azemar M, Maurer M, Groner B. Adaptive Resistance to Immunotherapy Directed Against p53 Can be Overcome by Global Expression of Tumor-Antigens in Dendritic Cells. Front Oncol 2014; 4:270. [PMID: 25340039 PMCID: PMC4186483 DOI: 10.3389/fonc.2014.00270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/17/2014] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy of cancer utilizes dendritic cells (DCs) for antigen presentation and the induction of tumor-specific immune responses. However, the therapeutic induction of anti-tumor immunity is limited by tumor escape mechanisms. In this study, immortalized dendritic D2SC/1 cells were transduced with a mutated version of the p53 tumor suppressor gene, p53M234I, or p53C132F/E168G, which are overexpressed in MethA fibrosarcoma tumor cells. In addition, D2SC/1 cells were fused with MethA tumor cells to generate a vaccine that potentially expresses a large repertoire of tumor-antigens. Cellular vaccines were transplanted onto Balb/c mice and MethA tumor growth and anti-tumor immune responses were examined in vaccinated animals. D2SC/1–p53M234I and D2SC/1–p53C132F/E168G cells induced strong therapeutic and protective MethA tumor immunity upon transplantation in Balb/c mice. However, in a fraction of immunized mice MethA tumor growth resumed after an extended latency period. Analysis of these tumors indicated loss of p53 expression. Mice, pre-treated with fusion hybrids generated from D2SC/1 and MethA tumor cells, suppressed MethA tumor growth and averted adaptive immune escape. Polyclonal B-cell responses directed against various MethA tumor proteins could be detected in the sera of D2SC/1–MethA inoculated mice. Athymic nude mice and Balb/c mice depleted of CD4+ or CD8+ T-cells were not protected against MethA tumor cell growth after immunization with D2SC/1–MethA hybrids. Our results highlight a potential drawback of cancer immunotherapy by demonstrating that the induction of a specific anti-tumor response favors the acquisition of tumor phenotypes promoting immune evasion. In contrast, the application of DC/tumor cell fusion hybrids prevents adaptive immune escape by a T-cell dependent mechanism and provides a simple strategy for personalized anti-cancer treatment without the need of selectively priming the host immune system.
Collapse
Affiliation(s)
- Matjaz Humar
- Pharmaceutical Biology and Biotechnology, Albert-Ludwigs-University of Freiburg , Freiburg , Germany
| | - Marc Azemar
- Internistische Onkologie, Tumor Biology Center , Freiburg , Germany
| | - Martina Maurer
- Basilea Pharmaceutica International Ltd. , Basel , Switzerland
| | - Bernd Groner
- Institute for Biomedical Research, Georg Speyer Haus , Frankfurt am Main , Germany
| |
Collapse
|
71
|
Frequent expression of zinc-finger protein ZNF165 in human urinary bladder transitional cell carcinoma. Immunobiology 2014; 220:68-73. [PMID: 25214475 DOI: 10.1016/j.imbio.2014.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/21/2014] [Accepted: 08/22/2014] [Indexed: 11/20/2022]
Abstract
The aim of the study is to evaluate mRNA/protein expression of zinc finger protein 165 (ZNF165) in transitional cell carcinomas (TCCs) of urinary bladder and correlate its expression with the clinicopathological characteristics of patients. In this study, the methods of quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) were utilized to evaluate mRNA/protein expression of ZNF165 in TCC. Independent Student's t test, ANOVA and Chi-square (χ(2)) were used to analyze the data statistically. We observed overexpression of ZNF165 mRNA in testis and majority (59.2%) of TCC patients. ZNF165 mRNA expression was also detected in adjacent noncancerous tissues (ANCTs) and some other normal tissues. Relative mean fold expression of ZNF165 mRNA was found to be significantly (p<0.01) higher in muscle-invasive bladder cancer (MIBC) as compared to non-muscle-invasive bladder cancer (NMIBC) patients. (12.11±9.57 vs. 5.72±2.61, p=0.009). ZNF165 protein expression was demonstrated on archival formalin-fixed, paraffin-embedded (FFPE) bladder tissues using IHC and nuclear staining pattern was detected. No significant difference was observed in protein expression of ZNF165 between the two groups (NMIBC and MIBC patients) (61.1% vs. 55.2%, p=0.629). No significant protein expression of ZNF165 was observed among ANCTs and benign prostatic hyperplasia (BPH) used as control. Our study results suggest that ZNF165 mRNA/protein expression was observed in TCC of human urinary bladder and might be used as a novel diagnostic biomarker and as well a vaccine target in development of urinary bladder cancer specific immunotherapy.
Collapse
|
72
|
De Koker S, Fierens K, Dierendonck M, De Rycke R, Lambrecht BN, Grooten J, Remon JP, De Geest BG. Nanoporous polyelectrolyte vaccine microcarriers. A formulation platform for enhancing humoral and cellular immune responses. J Control Release 2014; 195:99-109. [PMID: 25078552 DOI: 10.1016/j.jconrel.2014.07.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/18/2014] [Accepted: 07/20/2014] [Indexed: 11/19/2022]
Abstract
In this paper we report on the design, characterization and immuno-biological evaluation of nanoporous polyelectrolyte microparticles as vaccine carrier. Relative to soluble antigen, formulation of antigen as a sub-10 μm particle can strongly enhance antigen-specific cellular immune responses. The latter is crucial to confer protective immunity against intracellular pathogens and for anti-cancer vaccines. However, a major bottleneck in microparticulate vaccine formulation is the development of generic strategies that afford antigen encapsulation under benign and scalable conditions. Our strategy is based on spray drying of a dilute aqueous solution of antigen, oppositely charged polyelectrolytes and mannitol as a pore-forming component. The obtained solid microparticles can be redispersed in aqueous medium, leading to leaching out of the mannitol, thereby creating a highly porous internal structure. This porous structure enhances enzymatic processing of encapsulated proteins. After optimizing the conditions to process these microparticles we demonstrate that they strongly enhance cross-presentation in vitro by dendritic cells to CD8 T cells. In vivo experiments in mice confirm that this vaccine formulation technology is capable of enhancing cellular immune responses.
Collapse
Affiliation(s)
- Stefaan De Koker
- Department of Pharmaceutics, Ghent University, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Zwijnaarde, Ghent, Belgium
| | - Kaat Fierens
- VIB Inflammation Research Center, University of Ghent, Ghent, Belgium; Department of Respiratory Medicine, University Hospital Ghent, Ghent, Belgium
| | | | - Riet De Rycke
- VIB Inflammation Research Center, University of Ghent, Ghent, Belgium; Department of Plant Systems Biology, VIB, and Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- VIB Inflammation Research Center, University of Ghent, Ghent, Belgium; Department of Respiratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Johan Grooten
- Department of Biomedical Molecular Biology, Ghent University, Zwijnaarde, Ghent, Belgium
| | - Jean Paul Remon
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | | |
Collapse
|
73
|
Cavallo F, Aurisicchio L, Mancini R, Ciliberto G. Xenogene vaccination in the therapy of cancer. Expert Opin Biol Ther 2014; 14:1427-42. [DOI: 10.1517/14712598.2014.927433] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
74
|
Kim HS, Kang D, Moon MH, Kim HJ. Identification of pancreatic cancer-associated tumor antigen from HSP-enriched tumor lysate-pulsed human dendritic cells. Yonsei Med J 2014; 55:1014-27. [PMID: 24954332 PMCID: PMC4075362 DOI: 10.3349/ymj.2014.55.4.1014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/23/2013] [Accepted: 11/04/2013] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Vaccine strategies utilizing dendritic cells (DCs) to elicit anti-tumor immunity are the subject of intense research. Although we have shown that DCs pulsed with heat-treated tumor lysate (HTL) induced more potent anti-tumor immunity than DCs pulsed with conventional tumor lysate (TL), the underlying molecular mechanism is unclear. In order to explore the molecular basis of this approach and to identify potential antigenic peptides from pancreatic cancer, we analyzed and compared the major histocompatibility complex (MHC) ligands derived from TL- and HTL-pulsed dendritic cells by mass spectrophotometry. MATERIALS AND METHODS Human monocyte-derived dendritic cells were pulsed with TL or HTL prior to maturation induction. To delineate differences of MHC-bound peptide repertoire eluted from DCs pulsed with TL or HTL, nanoflow liquid chromatography-electrospray ionization-tandem mass spectrometry (nLC-ESI-MS-MS) was employed. RESULTS HTL, but not TL, significantly induced DC function, assessed by phenotypic maturation, allostimulation capacity and IFN-γ secretion by stimulated allogeneic T cells. DCs pulsed with TL or HTL displayed pancreas or pancreatic cancer-related peptides in context of MHC class I and II molecules. Some of the identified peptides had not been previously reported as expressed in pancreatic cancer or cancer of other tissue types. CONCLUSION Our partial lists of MHC-associated peptides revealed the differences between peptide profiles eluted from HTL-and TL-loaded DCs, implying that induced heat shock proteins in HTL chaperone tumor-derived peptides enhanced their delivery to DCs and promoted cross-presentation by DC. These findings may aid in identifying novel tumor antigens or biomarkers and in designing future vaccination strategies.
Collapse
Affiliation(s)
- Han-Soo Kim
- Innovative Cell and Gene Therapy Center, International St. Mary's Hospital, Incheon, Korea
| | - Dukjin Kang
- Center for Bioanalysis, Division of Metrology for Quality of Life, Korea Research Institute of Standards and Science, Daejeon, Korea
| | | | - Hyung Jik Kim
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea.
| |
Collapse
|
75
|
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
|
76
|
Rein LAM, Chao NJ. WT1 vaccination in acute myeloid leukemia: new methods of implementing adoptive immunotherapy. Expert Opin Investig Drugs 2014; 23:417-26. [DOI: 10.1517/13543784.2014.889114] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
77
|
Ferrone S, Campoli M. A fresh look at an old story: revisiting HLA class II antigen expression by melanoma cells. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.1.6.805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
78
|
Pilla L, Valenti R, Marrari A, Patuzzo R, Santinami M, Parmiani G, Rivoltini L. Vaccination: role in metastatic melanoma. Expert Rev Anticancer Ther 2014; 6:1305-18. [PMID: 16925496 DOI: 10.1586/14737140.6.8.1305] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Based on the poor impact on overall survival obtained by systemic chemotherapy in metastatic melanoma and the identification of many melanoma antigens recognized by T cells, in the last decade many efforts have been devoted to the development of active specific immunotherapy as a promising systemic treatment for this neoplastic disease. A number of Phase I-II clinical trials have been performed with different vaccination approaches that included whole tumor cells, antigen peptides, antigen-pulsed dendritic cells, recombinant viruses, plasmids or naked DNA, and heat-shock proteins. Despite some promising immunological and clinical results obtained in these studies, melanoma-specific vaccines have altogether failed to prove their efficacy in the few large Phase III randomized clinical trials performed. Nonetheless, the possibility of activating the human immune system to recognize and destroy tumor cells remains a challenging investigative field, considering that the new knowledge of the intricate cellular and molecular mechanisms that regulate the immune function and tumor-host interactions may allow the development of new clinically relevant melanoma vaccination strategies.
Collapse
Affiliation(s)
- Lorenzo Pilla
- Istituto Nazionale per lo Studio e la Cura dei Tumori, Unit of Immunotherapy of Human Tumors, Via Venezian 1, 20133 Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
79
|
Abstract
Dendritic cells (DCs) are the most powerful professional antigen-presenting cells and are unique in their capability to initiate, maintain and regulate the intensity of primary immune responses, including specific antitumor responses. Development of practical procedures to prepare sufficient numbers of functional human DCs in culture from the peripheral blood precursors, paved the way for clinical trials to evaluate various DC-based strategies in patients with malignant diseases. However, no definite conclusions regarding the clinical and even immunological efficacy of DC vaccination can be stated, despite the fact that 12 years have passed since the first clinical trial utilizing DCs in cancer patients. Many unanswered questions hamper the development of DC-based vaccines, including the source of DC preparation and protocols for DC generation, activation and loading with tumor antigens, source of tumor antigens, route of vaccine administration and methods of immunomonitoring. Fortunately, in spite of the many obstacles, DC vaccines continue to hold promise for cancer therapy.
Collapse
Affiliation(s)
- Hua Zhong
- Shanghai Jiao Tong University, Shanghai Chest Hospital, 241 Huaihai Road (w), Shanghai 200030, China.
| | | | | |
Collapse
|
80
|
Walden P, Sterry W. New and emerging vaccination strategies for prevention and treatment of dermatological diseases. Expert Rev Vaccines 2014; 3:421-31. [PMID: 15270647 DOI: 10.1586/14760584.3.4.421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Accelerated by the rapid advancements of our understanding of the molecular and cellular pathology of diseases and of the components and mechanisms of cellular and humoral immune responses, new vaccination strategies are being developed and explored for treatment and prevention of infectious diseases, cancer, autoimmune disorders and allergies. Many newly developed vaccination strategies are already in clinical trials, some with very promising results. Although most of these strategies are still at very early stages of their development, it is foreseeable that vaccination will evolve to play an important role in prevention, treatment and management of all the above classes of diseases.
Collapse
Affiliation(s)
- Peter Walden
- Clinical Research Group for Tumor Immunology, Department of Dermatology, Venerology and Allergy, Charite - Universitatsmedizin Berlin, Humboldt University, 10098 Berlin, Germany.
| | | |
Collapse
|
81
|
Chemical castration of melanoma patients does not increase the frequency of tumor-specific CD4 and CD8 T cells after peptide vaccination. J Immunother 2013; 36:276-86. [PMID: 23603862 DOI: 10.1097/cji.0b013e31829419f3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Peptide vaccination against tumor-associated antigens remains one of the most common methods of immunization in cancer vaccine clinical trials. Although peptide vaccination has been reported to increase circulating antigen-specific T-cells, they have had limited clinical efficacy and there is a necessity to increase their capacity to generate strong antitumor responses. We sought to improve the clinical efficacy of peptide-based vaccines in cancer immunotherapy of metastatic melanoma using a LHRH agonist (leuprolide) as adjuvant. Seventy HLA-A*0201 stage IIb-IV melanoma patients were vaccinated with class I HLA-A*0201-restricted gp100209-2M peptide and stratified for HLA-DP4 restriction. HLA-DP4 patients were also vaccinated with class II HLA-DP4-restricted MAGE-3243-258 peptide. Patients from both groups were randomized to receive 2 doses of leuprolide or not. Here we report the increase in PBMC TREC levels at week 24 after peptide vaccination, which was independent of the leuprolide treatment. This change was mirrored by a small increase in the TREC-enriched CD8CD45RAROCD27CD103, but not the TREC-enriched CD4CD45RAROCD31 T-cell population. Serum concentration of 2 important factors for thymopoiesis was measured: insulin growth factor 1 (IGF-1) levels were not changed, whereas a moderate increase in IL-7 levels was noted in the sera of all patients 6 weeks after vaccination. Increased expression of CD127 (IL-7 receptor-α) at week 24, compared with baseline, was only seen in the CD8CD45RAROCD27CD103 T-cell population. Our results suggest that leuprolide has no effect on thymic output when used as peptide vaccine adjuvant, but IFA-based peptide vaccination may unexpectedly affect the thymus by increasing thymic output of new T cells.
Collapse
|
82
|
Fenoglio D, Traverso P, Parodi A, Kalli F, Zanetti M, Filaci G. Generation of more effective cancer vaccines. Hum Vaccin Immunother 2013; 9:2543-7. [PMID: 23978951 DOI: 10.4161/hv.26147] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cancer vaccines represent a promising therapeutic approach for which prime time is imminent. However, clinical efficacy must be improved in order for cancer vaccines to become a valid alternative or complement to traditional cancer treatments. Considerable efforts have been undertaken so far to better understand the fundamental requirements for clinically-effective cancer vaccines. Recent data emphasize that important requirements, among others, are (1) the use of multi-epitope immunogens, possibly deriving from different tumor antigens; (2) the selection of effective adjuvants; (3) the association of cancer vaccines with agents able to counteract the regulatory milieu present in the tumor microenvironment; and (4) the need to choose the definitive formulation and regimen of a vaccine after accurate preliminary tests comparing different antigen formulations. The first requirement deals with issues related to HLA restriction of tumor antigen presentation, as well as usefulness of tumor antigen spreading and counteraction of immune escape phenomena, linked to tumor antigen down-modulation, for an effective anti-cancer immune response. The second point underscores the necessity of optimal activation of innate immunity to achieve an efficient adaptive anti-cancer immune response. The third point focuses on the importance to inhibit subsets of regulatory cells. The last requirement stresses the concept that the regimen and formulation of the vaccine impacts profoundly on cancer vaccine efficacy. A new generation of cancer vaccines, provided with both immunological and clinical efficacy, will hopefully soon address these requirements.
Collapse
Affiliation(s)
- Daniela Fenoglio
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy; Department of Internal Medicine; University of Genoa; Genoa, Italy
| | - Paolo Traverso
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy; Department of Surgical Sciences; University of Genoa; Genoa, Italy
| | - Alessia Parodi
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy
| | - Francesca Kalli
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy
| | - Maurizio Zanetti
- The Laboratory of Immunology; Department of Medicine and Cancer Center; University of California; San Diego, CA USA
| | - Gilberto Filaci
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy; Department of Internal Medicine; University of Genoa; Genoa, Italy
| |
Collapse
|
83
|
Bregy A, Wong TM, Shah AH, Goldberg JM, Komotar RJ. Active immunotherapy using dendritic cells in the treatment of glioblastoma multiforme. Cancer Treat Rev 2013; 39:891-907. [PMID: 23790634 DOI: 10.1016/j.ctrv.2013.05.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 05/20/2013] [Accepted: 05/26/2013] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Glioblastoma multiforme, the most common malignant brain tumor still has a dismal prognosis with conventional treatment. Therefore, it is necessary to explore new and/or adjuvant treatment options to improve patient outcomes. Active immunotherapy is a new area of research that may be a successful treatment option. The focus is on vaccines that consist of antigen presenting cells (APCs) loaded with tumor antigen. We have conducted a systematic review of prospective studies, case reports and clinical trials. The goal of this study was to examine the efficacy and safety in terms of complications, median overall survival (OS), progression free survival (PFS) and quality of life. METHODS A PubMed search was performed to include all relevant studies that reported the characteristics, outcomes and complications of patients with GBM treated with active immunotherapy using dendritic cells. Reported parameters were immune response, radiological findings, median PFS and median OS. Complications were categorized based on association with the craniotomy or with the vaccine itself. RESULTS A total of 21 studies with 403 patients were included in our review. Vaccination with dendritic cells (DCs) loaded with autologous tumor cells resulted in increased median OS in patients with recurrent GBM (71.6-138.0 wks) as well as those newly diagnosed (65.0-230.4 wks) compared to average survival of 58.4 wks. CONCLUSIONS Active immunotherapy, specifically with autologous DCs loaded with autologous tumor cells, seems to have the potential of increasing median OS and prolonged tumor PFS with minimal complications. Larger clinical trials are needed to show the potential benefits of active immunotherapy.
Collapse
Affiliation(s)
- Amade Bregy
- University of Miami Miller School of Medicine, Department of Neurological Surgery, Miami, FL, USA
| | | | | | | | | |
Collapse
|
84
|
Kalli F, Machiorlatti R, Battaglia F, Parodi A, Conteduca G, Ferrera F, Proietti M, Tardito S, Sanguineti M, Millo E, Fenoglio D, De Palma R, Inghirami G, Filaci G. Comparative analysis of cancer vaccine settings for the selection of an effective protocol in mice. J Transl Med 2013; 11:120. [PMID: 23663506 PMCID: PMC3659084 DOI: 10.1186/1479-5876-11-120] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/08/2013] [Indexed: 12/31/2022] Open
Abstract
Background Cancer vaccines are considered a promising therapeutic approach. However, their clinical results are not yet satisfactory. This may be due to the the difficulty of selection of an efficient tumor associated antigen (TAA) and immunization protocol. Indeed, the weak antigenicity of many TAA impairs the design of robust procedures, therefore a systematic analysis to identify the most efficient TAA is mandatory. Here, we performed a study to compare different gp100 vaccination strategies to identify the best strategy to provide a 100% protection against experimental melanoma in a reproducible manner. Methods C57BL/6J mice were challenged subcutaneously with B16F10 melanoma cells, after vaccination with: a) mouse or human gp10025-33 peptide plus CpG adjuvant; b) mouse or human gp100 gene; c) mouse or human gp10025-33 peptide-pulsed dendritic cells (DC). Alternatively, a neutralizing anti-IL-10 monoclonal antibody (mAb) was subcutaneously administered at the site of tumor challenge to counteract regulatory cells. Finally, combinatorial treatment was performed associating human gp10025-33 peptide-pulsed DC vaccination with administration of the anti-IL-10 mAb. Results Vaccination with human gp10025-33 peptide-pulsed DC was the most effective immunization protocol, although not achieving a full protection. Administration of the anti-IL-10 mAb showed also a remarkable protective effect, replicated in mice challenged with a different tumor, Anaplastic Large Cell Lymphoma. When immunization with gp10025-33 peptide-pulsed DC was associated with IL-10 counteraction, a 100% protective effect was consistently achieved. The analysis on the T-cell tumor infiltrates showed an increase of CD4+granzyme+ T-cells and a decreased number of CD4+CD25+Foxp3+ Treg elements from mice treated with either gp10025-33 peptide-pulsed DC vaccination or anti-IL-10 mAb administration. These data suggest that processes of intratumoral re-balance between effector and regulatory T cell subpopulations may play a critical protective role in immunotherapy protocols. Conclusions Here we demonstrate that, in the setting of a cancer vaccine strategy, a comparative analysis of different personalized approaches may favour the unveiling of the most effective protocol. Moreover, our findings suggest that counteraction of IL-10 activity may be critical to revert the intratumoral environment promoting Treg polarization, thus increasing the effects of a vaccination against selected TAA.
Collapse
Affiliation(s)
- Francesca Kalli
- Centre of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV n. 7, 16132, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Akinfieva O, Nabiev I, Sukhanova A. New directions in quantum dot-based cytometry detection of cancer serum markers and tumor cells. Crit Rev Oncol Hematol 2013; 86:1-14. [DOI: 10.1016/j.critrevonc.2012.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 07/18/2012] [Accepted: 09/05/2012] [Indexed: 10/27/2022] Open
|
86
|
Yoshimura K, Uemura H. Role of vaccine therapy for renal cell carcinoma in the era of targeted therapy. Int J Urol 2013; 20:744-55. [PMID: 23521119 DOI: 10.1111/iju.12147] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/21/2013] [Indexed: 12/28/2022]
Abstract
Renal cell carcinoma is the most common malignant tumor originating from the kidney. Compared with other solid tumors, it does not respond to traditional management modalities, such as chemotherapy and radiotherapy. However, it is well known that renal cell carcinoma represents one of the most immune-responsive cancers and several immunotherapeutic strategies have been investigated in the management of renal cell carcinoma with variable degrees of success. The development of immunotherapy with α-interferon or high-dose interleukin-2 is the best established treatment, and is associated with durable disease control. Although the lack of defined antigens in renal cell carcinoma has hindered more specific vaccine development, research regarding vaccination therapy has been of special interest for the treatment of renal cell carcinoma for more than 30 years. At present, there are three types of cell-based vaccines in renal cell carcinoma treatment: autologous tumor-cell vaccines, genetically modified tumor vaccines and dendritic cell-based vaccines. A further type is peptide-based vaccination with tumor-associated antigens as possible targets, such as carbonic anhydrase IX, survivin and telomerase that are overexpressed in renal cell carcinoma. In the present article, we review data from completed clinical trials of vaccine therapy, and discuss future trials to assess the current knowledge and future role of vaccine therapy for renal cell carcinoma in the era of recently developed targeted therapy.
Collapse
Affiliation(s)
- Kazuhiro Yoshimura
- Department of Urology, Faculty of Medicine, Kinki University, Osaka, Japan.
| | | |
Collapse
|
87
|
Silva JM, Videira M, Gaspar R, Préat V, Florindo HF. Immune system targeting by biodegradable nanoparticles for cancer vaccines. J Control Release 2013; 168:179-99. [PMID: 23524187 DOI: 10.1016/j.jconrel.2013.03.010] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 03/11/2013] [Accepted: 03/14/2013] [Indexed: 01/08/2023]
Abstract
The concept of therapeutic cancer vaccines is based on the activation of the immune system against tumor cells after the presentation of tumor antigens. Nanoparticles (NPs) have shown great potential as delivery systems for cancer vaccines as they potentiate the co-delivery of tumor-associated antigens and adjuvants to dendritic cells (DCs), insuring effective activation of the immune system against tumor cells. In this review, the immunological mechanisms behind cancer vaccines, including the role of DCs in the stimulation of T lymphocytes and the use of Toll-like receptor (TLR) ligands as adjuvants will be discussed. An overview of each of the three essential components of a therapeutic cancer vaccine - antigen, adjuvant and delivery system - will be provided with special emphasis on the potential of particulate delivery systems for cancer vaccines, in particular those made of biodegradable aliphatic polyesters, such as poly(lactic-co-glycolic acid) (PLGA) and poly-ε-caprolactone (PCL). Some of the factors that can influence NP uptake by DCs, including size, surface charge, surface functionalization and route of administration, will also be considered.
Collapse
Affiliation(s)
- Joana M Silva
- iMed.UL, Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal
| | | | | | | | | |
Collapse
|
88
|
Dillman RO, Cornforth AN, Nistor G. Cancer stem cell antigen-based vaccines: the preferred strategy for active specific immunotherapy of metastatic melanoma? Expert Opin Biol Ther 2013; 13:643-56. [PMID: 23451922 DOI: 10.1517/14712598.2013.759556] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION There are now two chemotherapy agents, one tyrosine kinase inhibitor and three immunotherapy products approved for the treatment of metastatic melanoma, but an unmet need persists because these options are toxic and of limited therapeutic benefit. Active specific immunotherapy with therapeutic vaccines could be a useful addition to the therapeutic armamentarium, especially in patients whose tumor burden has been reduced by other treatment modalities. AREAS COVERED This article reviews various sources of melanoma antigens, such as peptides, gangliosides, autologous tumor and cancer stem cells including allogeneic and autologous cell lines. The advantages and disadvantages of various antigen sources and allogeneic and autologous approaches are discussed with an emphasis on the theoretical benefits of immunizing against cancer stem cells. The results from published randomized trials testing the benefit of various vaccine approaches are summarized, as well as promising results from three Phase II trials (one randomized) of patient-specific stem cell antigen-based products. EXPERT OPINION Immune responses directed toward the unique neoantigens and stem cell antigens expressed on continuously proliferating, self-renewing, autologous tumor cells could potentially overcome the limitations inherent in these other antigen-based approaches, that to date, have yielded disappointing results in randomized trials.
Collapse
Affiliation(s)
- Robert O Dillman
- Hoag Institute for Research and Education, Hoag Hospital, One Hoag Dr, Bldg 44 Suite 210, Newport Beach, California 92663, USA.
| | | | | |
Collapse
|
89
|
Cancer treatment using peptides: current therapies and future prospects. JOURNAL OF AMINO ACIDS 2012; 2012:967347. [PMID: 23316341 PMCID: PMC3539351 DOI: 10.1155/2012/967347] [Citation(s) in RCA: 317] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 12/07/2012] [Indexed: 12/12/2022]
Abstract
This paper discusses the role of peptides in cancer therapy with special emphasis on peptide drugs which are already approved and those in clinical trials. The potential of peptides in cancer treatment is evident from a variety of different strategies that are available to address the progression of tumor growth and propagation of the disease. Use of peptides that can directly target cancer cells without affecting normal cells (targeted therapy) is evolving as an alternate strategy to conventional chemotherapy. Peptide can be utilized directly as a cytotoxic agent through various mechanisms or can act as a carrier of cytotoxic agents and radioisotopes by specifically targeting cancer cells. Peptide-based hormonal therapy has been extensively studied and utilized for the treatment of breast and prostate cancers. Tremendous amount of clinical data is currently available attesting to the efficiency of peptide-based cancer vaccines. Combination therapy is emerging as an important strategy to achieve synergistic effects in fighting cancer as a single method alone may not be efficient enough to yield positive results. Combining immunotherapy with conventional therapies such as radiation and chemotherapy or combining an anticancer peptide with a nonpeptidic cytotoxic drug is an example of this emerging field.
Collapse
|
90
|
Russo V, Pilla L, Lunghi F, Crocchiolo R, Greco R, Ciceri F, Maggioni D, Fontana R, Mukenge S, Rivoltini L, Rigamonti G, Mercuri SR, Nicoletti R, Maschio AD, Gianolli L, Fazio F, Marchianò A, Florio AD, Maio M, Salomoni M, Gallo-Stampino C, Fiacco MD, Lambiase A, Coulie PG, Patuzzo R, Parmiani G, Traversari C, Bordignon C, Santinami M, Bregni M. Clinical and immunologic responses in melanoma patients vaccinated with MAGE-A3-genetically modified lymphocytes. Int J Cancer 2012; 132:2557-66. [DOI: 10.1002/ijc.27939] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 10/15/2012] [Indexed: 01/31/2023]
|
91
|
Dendritic cells recruitment in melanoma metastasis treated by electrochemotherapy. Clin Exp Metastasis 2012; 30:37-45. [DOI: 10.1007/s10585-012-9505-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 06/13/2012] [Indexed: 10/28/2022]
|
92
|
Wang E, Tomei S, Marincola FM. Reflections upon human cancer immune responsiveness to T cell-based therapy. Cancer Immunol Immunother 2012; 61:761-70. [PMID: 22576055 PMCID: PMC3362724 DOI: 10.1007/s00262-012-1274-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/24/2012] [Indexed: 01/06/2023]
Abstract
Immune-mediated rejection of human cancer is a relatively rare but well-documented phenomenon. Its rate of occurrence progressively increases from the occasional observation of spontaneous regressions to the high rate of complete remissions observed in response to effective treatments. For two decades, our group has focused its interest in understanding this phenomenon by studying humans following an inductive approach. Sticking to a sequential logic, we dissected the phenomenon by studying to the best of our capability both peripheral and tumor samples and reached the conclusion that immune-mediated cancer rejection is a facet of autoimmunity where the target tissue is the cancer itself. As we are currently defining the strategy to effectively identify the mechanisms leading in individual patients to rejection of their own tumors, we considered useful to summarize the thought process that guided us to our own interpretation of the mechanisms of immune responsiveness.
Collapse
Affiliation(s)
- Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bldg 10, Room 1C711, 9000 Rockville Pike, Bethesda, MD 20892 USA
| | - Sara Tomei
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bldg 10, Room 1C711, 9000 Rockville Pike, Bethesda, MD 20892 USA
| | - Francesco M. Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bldg 10, Room 1C711, 9000 Rockville Pike, Bethesda, MD 20892 USA
| |
Collapse
|
93
|
Abstract
Devitalized tumor cells either autologous or allogeneic have been used as anti-cancer vaccines with the purpose of facilitating the induction of an immune response able to destroy growing tumor cells since the identification of tumor antigens was deemed not to be necessary, particularly in the autologous system. Such vaccines were tested first in animal models and then in the clinics as unmodified tumor cells or after insertion of genes coding for factors known to increase the immune response against tumors. These vaccines were usually given by subcutaneous injections along with different immunological adjuvants. Such immunization approaches were found to be effective in mice when carried out in a tumor preventive setting but significantly less in the therapeutic context, that is, in the presence of an established tumor. By analyzing several clinical trials of vaccination using either autologous or allogeneic unmodified and gene-modified tumor cells published in the last 10 to 15 years, we conclude for a lack of sufficient evidence for efficacy of this strategy in inducing both a strong immune response and a therapeutic response. A potential variant of this strategy is the direct intratumoral injection of immunostimulatory genes delivered by vectors in vivo. But even this approach failed to provide a statistically significant clinical benefit for the cancer patients.We also point out the inherent drawbacks of the tumor cell-based vaccine strategy that include (a) a limited frequency by which human tumor lines can be obtained from clinical samples, (b) the low number of available cells for vaccination, (c) the release of immune-suppressive factors by tumor cells, and (d) the cost and time necessary for standardization and collecting/expanding a number of cells according to the approved regulatory requirements. Thus, taking into consideration the new developments in cancer vaccines, we believe that tumor cell-based vaccines should be dismissed as anti-cancer vaccines unless a clear benefit could be demonstrated by the few ongoing trials of combination with new immunomodulating reagents (eg, anti-CTLA4, PD-1, chemotherapy).
Collapse
|
94
|
Cools N, Petrizzo A, Smits E, Buonaguro FM, Tornesello ML, Berneman Z, Buonaguro L. Dendritic cells in the pathogenesis and treatment of human diseases: a Janus Bifrons? Immunotherapy 2012; 3:1203-22. [PMID: 21995572 DOI: 10.2217/imt.11.110] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) represent the bridging cell compartment between a variety of nonself antigens (i.e., microbial, cancer and vaccine antigens) and adaptive immunity, orchestrating the quality and potency of downstream immune responses. Because of the central role of DCs in the generation and regulation of immunity, the modulation of DC function in order to shape immune responses is gaining momentum. In this respect, recent advances in understanding DC biology, as well as the required molecular signals for induction of T-cell immunity, have spurred many experimental strategies to use DCs for therapeutic immunological approaches for infections and cancer. However, when DCs lose control over such 'protective' responses - by alterations in their number, phenotype and/or function - undesired effects leading to allergy and autoimmune clinical manifestations may occur. Novel therapeutic approaches have been designed and currently evaluated in order to address DCs and silence these immunopathological processes. In this article we present recent concepts of DC biology and some medical implications in view of therapeutic opportunities.
Collapse
Affiliation(s)
- Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (Vaxinfectio), University of Antwerp, B-2610 Wilrijk, Belgium
| | | | | | | | | | | | | |
Collapse
|
95
|
Brookman-May S, Burger M, Wieland WF, Rössler W, May M, Denzinger S. Vaccination therapy in renal cell carcinoma: current position and future options in metastatic and localized disease. Expert Rev Vaccines 2011; 10:837-52. [PMID: 21692704 DOI: 10.1586/erv.11.64] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
As renal cell carcinoma represents one of the most immune-responsive cancers, immunotherapy exhibits a suitable treatment basis. Beside nonspecific stimulation via cytokines, passive specific and active immunotherapy are also appropriate options to recognize and destroy tumor cells. For more than 30 years, research regarding vaccination therapy has been of special interest for the treatment of renal cell carcinoma. However, apart from occasional promising results in Phase I and II trials, vaccination therapy is still considered experimental in this tumor entity, especially owing to missing results from Phase III trials demonstrating clinical efficacy. In the present article, we review data from completed clinical trials of vaccination therapy and also discuss scheduled future trials, in order to assess the current position and possible future fields of application of vaccination therapy in renal cell carcinoma in the era of recently developed targeted therapies.
Collapse
Affiliation(s)
- Sabine Brookman-May
- University of Regensburg, Department of Urology, Caritas St. Josef Medical Center, Landshuter Strasse 65, 93053 Regensburg, Germany.
| | | | | | | | | | | |
Collapse
|
96
|
Cancer vaccines. Any future? Arch Immunol Ther Exp (Warsz) 2011; 59:249-59. [PMID: 21644030 DOI: 10.1007/s00005-011-0129-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 02/02/2011] [Indexed: 12/22/2022]
Abstract
The idea that vaccination can be used to fight cancer is not new. Approximately 100 years ago, researchers attempted to stimulate a tumor-specific, therapeutic immune response to tumors by injecting patients with cells and extracts from their own tumors, or tumors of the same type from different individuals. During the last decade, great efforts have been made to develop immunotherapeutic approaches for the treatment of malignant diseases as alternatives to traditional chemo- and radiotherapy. A quintessential goal of immunotherapy in cancer is treatment with vaccines that elicit potent anti-tumor immune responses without side effects. In this article, we have attempted to review some of the most problematic issues facing the development of cancer vaccines. With the prospect of immunosuppression, an ill-designed cancer vaccine can be more harmful than a no-benefit therapy. We have noted that "immunoediting" and "immunodominance" are the premier setbacks in peptide-based vaccines and therefore it appears necessary not only to manipulate the activity of a vast number of principal components but also to finely tune their concentrations in time and space. In the face of all these quandaries, it is at least doubtful that any reliable anti-cancer vaccine strategy will emerge in the near future.
Collapse
|
97
|
Kovjazin R, Volovitz I, Kundel Y, Rosenbaum E, Medalia G, Horn G, Smorodinsky NI, Brenner B, Carmon L. ImMucin: a novel therapeutic vaccine with promiscuous MHC binding for the treatment of MUC1-expressing tumors. Vaccine 2011; 29:4676-86. [PMID: 21570434 DOI: 10.1016/j.vaccine.2011.04.103] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Revised: 02/21/2011] [Accepted: 04/27/2011] [Indexed: 10/18/2022]
Abstract
An optimal cancer vaccine should be able to induce highly potent, long-lasting, tumor-specific responses in the majority of the cancer patient population. One approach for achieving this is to use synthetic peptide vaccines derived from widely expressed tumor-associated antigens, that promiscuously bind multiple MHC class I and class II alleles. MUC1-SP-L (ImMucin, VXL100) is a 21mer peptide encoding the complete signal peptide domain of MUC1, a tumor-associated antigen expressed by over 90% of solid and non-solid tumors. MUC1-SP-L was predicted in silico to bind various MHC class I and MHC class II alleles, covering the majority of the Caucasian population. PBLs obtained from 13 naïve donors all proliferated, with a Stimulation Index (SI≥2), to the MUC1-SP-L peptide, producing mixed CD4+ and CD8+ responses. Similar results were manifested by MUC1-SP-L in PBLs derived from 9 of 10 cancer patients with MUC1 positive tumors. CD4+ and CD8+ T cell populations exhibited CD45RO memory markers and secreted IFN-gamma and IL-2 following stimulation with MUC1-SP-L. These T cells also exhibited proliferation to the MUC1-SP-L inner 9mer epitopes and cytotoxicity against tumor cell lines expressing MUC1 and a concordant MHC class I allele. Cytotoxicity to MUC1-expressing human and murine tumors was shown also in T cells obtained from HLA-A2 transgenic mice and BALB/c syngeneic mice immunized with the MUC1-SP-L and GM-CSF. In an immunotherapy model, BALB/c mice inoculated with metastatic MUC1 transfected murine DA3 mammary tumor cells, exhibited significantly prolonged survival following vaccination with MUC1-SP-L. Our results indicate superior immunological and anti-tumor properties of MUC1-SP-L compared to previously published MUC1-derived epitopes.
Collapse
Affiliation(s)
- Riva Kovjazin
- Vaxil BioTherapeutics Ltd., 13 Einstein Street, Weizmann Science Park, Nes-Ziona 74036, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Nierkens S, Janssen EM. Harnessing dendritic cells for tumor antigen presentation. Cancers (Basel) 2011; 3:2195-213. [PMID: 24212804 PMCID: PMC3757412 DOI: 10.3390/cancers3022195] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/14/2011] [Accepted: 04/19/2011] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DC) are professional antigen presenting cells that are crucial for the induction of anti-tumor T cell responses. As a consequence, research has focused on the harnessing of DCs for therapeutic interventions. Although current strategies employing ex vivo-generated and tumor-antigen loaded DCs have been proven feasible, there are still many obstacles to overcome in order to improve clinical trial successes and offset the cost and complexity of customized cell therapy. This review focuses on one of these obstacles and a pivotal step for the priming of tumor-specific CD8+ and CD4+ T cells; the in vitro loading of DCs with tumor antigens.
Collapse
Affiliation(s)
- Stefan Nierkens
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Geert Grooteplein 28, Nijmegen 6525 GA, The Netherlands; E-Mail:
| | - Edith M. Janssen
- Division of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
99
|
Wongthida P, Diaz RM, Pulido C, Rommelfanger D, Galivo F, Kaluza K, Kottke T, Thompson J, Melcher A, Vile R. Activating systemic T-cell immunity against self tumor antigens to support oncolytic virotherapy with vesicular stomatitis virus. Hum Gene Ther 2011; 22:1343-53. [PMID: 21366404 DOI: 10.1089/hum.2010.216] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We have shown that the antitumor activity of vesicular stomatitis virus (VSV) against B16ova tumors in C57BL/6 mice is predominantly due to innate antiviral immune effectors. We have also shown that the innate immune-activating properties of VSV can be harnessed to prime adaptive T-cell responses against a tumor-associated antigen (TAA) if the virus is engineered to express the cDNA of the antigen. Here, we show that the combination of VSV expressing OVA as a model tumor antigen, along with adoptive T-cell therapy targeted against the same antigen, is superior to either treatment alone and induces systemic antitumor activity. In addition, we extend our findings with the OVA model to the therapeutic use of VSV expressing hgp100, a self TAA against which tolerance is well established in C57BL/6 mice. In contrast to VSV-ova, T-cell responses raised by VSV-hgp100 were insufficient to improve therapy against B16ova tumors compared with VSV-GFP alone. However, in combination with adoptive transfer of gp100-specific pmel T cells, intratumoral VSV-hgp100 cured significantly more mice than either virus or T cells alone. Even in an aggressive model of metastatic disease, antitumor therapy was generated at levels similar to those observed in the VSV-ova/OT-I model in which a potently immunogenic, nonself TAA was targeted. Therefore, individual poorly effective virotherapies and T-cell therapies that target self TAA of low immunogenicity, which reflects the situation in patients, can be combined to generate very effective antitumor therapy.
Collapse
|
100
|
Archaeosome adjuvant overcomes tolerance to tumor-associated melanoma antigens inducing protective CD8 T cell responses. Clin Dev Immunol 2011; 2010:578432. [PMID: 21318177 PMCID: PMC3034908 DOI: 10.1155/2010/578432] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 12/15/2010] [Accepted: 12/23/2010] [Indexed: 01/16/2023]
Abstract
Vesicles comprised of the ether glycerolipids of the archaeon Methanobrevibacter smithii (archaeosomes) are potent adjuvants for evoking CD8+ T cell responses. We therefore explored the ability of archaeosomes to overcome immunologic tolerance to self-antigens. Priming and boosting of mice with archaeosome-antigen evoked comparable CD8+ T cell response and tumor protection to an alternate boosting strategy utilizing live bacterial vectors for antigen delivery. Vaccination with melanoma antigenic peptides TRP181-189 and Gp10025-33 delivered in archaeosomes resulted in IFN-γ producing antigen-specific CD8+ T cells with strong cytolytic capability and protection against subcutaneous B16 melanoma. Targeting responses against multiple antigens afforded prolonged median survival against melanoma challenge. Entrapment of multiple peptides within the same vesicle or admixed formulations were both effective at evoking CD8+ T cells against each antigen. Melanoma-antigen archaeosome formulations also afforded therapeutic protection against established B16 tumors when combined with depletion of T-regulatory cells. Overall, we demonstrate that archaeosome adjuvants constitute an effective choice for formulating cancer vaccines.
Collapse
|