1
|
Dillman RO, Nistor GI, Keirstead HS. Autologous dendritic cells loaded with antigens from self-renewing autologous tumor cells as patient-specific therapeutic cancer vaccines. Hum Vaccin Immunother 2023:2198467. [PMID: 37133853 DOI: 10.1080/21645515.2023.2198467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
A promising personal immunotherapy is autologous dendritic cells (DC) loaded ex vivo with autologous tumor antigens (ATA) derived from self-renewing autologous cancer cells. DC-ATA are suspended in granulocyte-macrophage colony stimulating factor at the time of each subcutaneous injection. Previously, irradiated autologous tumor cell vaccines have produced encouraging results in 150 cancer patients, but the DC-ATA vaccine demonstrated superiority in single-arm and randomized trials in metastatic melanoma. DC-ATA have been injected into more than 200 patients with melanoma, glioblastoma, and ovarian, hepatocellular, and renal cell cancers. Key observations include: [1] greater than 95% success rates for tumor cell cultures and monocyte collection for dendritic cell production; [2] injections are well-tolerated; [3] the immune response is rapid and includes primarily TH1/TH17 cellular responses; [4] efficacy has been suggested by delayed but durable complete tumor regressions in patients with measurable disease, by progression-free survival in glioblastoma, and by overall survival in melanoma.
Collapse
Affiliation(s)
| | - Gabriel I Nistor
- Research and Development, AIVITA Biomedical Inc, Irvine, CA, USA
| | | |
Collapse
|
2
|
Quixabeira DCA, Cervera-Carrascon V, Santos JM, Clubb JH, Kudling TV, Basnet S, Heiniö C, Grönberg-Vähä-Koskela S, Anttila M, Havunen R, Kanerva A, Hemminki A. Local therapy with an engineered oncolytic adenovirus enables antitumor response in non-injected melanoma tumors in mice treated with aPD-1. Oncoimmunology 2022; 11:2028960. [PMID: 35083096 PMCID: PMC8786329 DOI: 10.1080/2162402x.2022.2028960] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/23/2021] [Accepted: 01/10/2022] [Indexed: 12/22/2022] Open
Abstract
Intratumoral immunotherapies are entering clinical use but concerns remain regarding their effects on non-injected tumors. Here, we studied the impact of local treatment with an adenovirus coding for TNFa and IL-2 on systemic antitumor response in animals receiving aPD-1 (anti-programmed cell death protein 1) therapy. Using bilateral murine melanoma models, we tested systemic tumor response to combined therapy with anti-PD-1 and an adenovirus coding for TNFa and IL-2 ("virus"). Virus was given intratumorally (to one of the two tumors only) and aPD-1 monoclonal antibody systemically. We evaluated both tumors' response to treatment, overall survival, metastasis development, and immunological mechanisms involved with response. Consistent tumor control was observed in both injected and non-injected tumors, including complete response in all treated animals receiving aPD-1+ virus therapy. Mechanistically, virus injections enabled potent effector lymphocyte response locally, with systemic effects in non-injected tumors facilitated by aPD-1 treatment. Moreover, adenovirus therapy demonstrated immunological memory formation. Virus therapy was effective in preventing metastasis development. Local treatment with TNFa and IL-2 coding adenovirus enhanced systemic response to aPD-1 therapy, by re-shaping the microenvironment of both injected and non-injected tumors. Therefore, our pre-clinical data support the rationale for a trial utilizing a combination of aPD-1 plus virus for the treatment of human cancer.
Collapse
Affiliation(s)
- Dafne C. A. Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics, Helsinki, Finland
| | - Joao M. Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics, Helsinki, Finland
| | - James H.A. Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics, Helsinki, Finland
| | - Tatiana V. Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Susanna Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | | | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics, Helsinki, Finland
| | - Anna Kanerva
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
3
|
Koukourakis IM, Koukourakis MI. Combining the past and present to advance immuno-radiotherapy of cancer. Int Rev Immunol 2021; 42:26-42. [PMID: 34511006 DOI: 10.1080/08830185.2021.1974020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since its first clinical application, 120 years ago, radiotherapy evolved into a major anti-cancer treatment modality, offering high cure rates in many human malignancies. During the past ten years, the establishment of immune checkpoint inhibitors (ICIs) in cancer therapeutics has vigorously reintroduced the immune system's role in the outcome of radiotherapy and, conversely, the role of radio-vaccination in the efficacy of immunotherapy. The knowledge and clinical experience that founded the current era of immuno-radiotherapy started alongside with the birth of radiotherapy, and evolved through exhaustive experimental work, clinical trials on active specific immunotherapy, frustrating attempts to validate the importance of cytokine administration with radiotherapy, and, finally, the encouraging ICI-based clinical trials that opened the door to a far more encouraging perspective; radio-vaccination, through its old and new methods, is rising as a research field that promises to cure, previously incurable, disease. In this critical review, we focus on the scientific knowledge gathered through more than a century of research on radiotherapy interactions with the immune system. Understanding the origins of this promising therapeutic approach will substantially contribute to developing new immuno-radiotherapy policies in the fight against cancer.
Collapse
Affiliation(s)
- Ioannis M Koukourakis
- 1st Department of Radiology, Radiotherapy Unit, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael I Koukourakis
- Department of Radiotherapy/Oncology, Medical School, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| |
Collapse
|
4
|
Pawlikowska M, Jędrzejewski T, Slominski AT, Brożyna AA, Wrotek S. Pigmentation Levels Affect Melanoma Responses to Coriolus versicolor Extract and Play a Crucial Role in Melanoma-Mononuclear Cell Crosstalk. Int J Mol Sci 2021; 22:ijms22115735. [PMID: 34072104 PMCID: PMC8198516 DOI: 10.3390/ijms22115735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023] Open
Abstract
Melanoma, the malignancy originating from pigment-producing melanocytes, is the most aggressive form of skin cancer and has a poor prognosis once the disease starts to metastasize. The process of melanin synthesis generates an immunosuppressive and mutagenic environment, and can increase melanoma cell resistance to different treatment modalities, including chemo-, radio- or photodynamic therapy. Recently, we have shown that the presence of melanin pigment inhibits the melanoma cell response to bioactive components of Coriolus versicolor (CV) Chinese fungus. Herein, using the same human melanoma cell line in which the level of pigmentation can be controlled by the L-tyrosine concentration in culture medium, we tested the effect of suppression of melanogenesis on the melanoma cell response to CV extract and investigated the cell death pathway induced by fungus extract in sensitized melanoma cells. Our data showed that susceptibility to CV-induced melanoma cell death is significantly increased after cell depigmentation. To the best of our knowledge, we are the first to demonstrate that CV extract can induce RIPK1/RIPK3/MLKL-mediated necroptosis in depigmented melanoma cells. Moreover, using the co-culture system, we showed that inhibition of the tyrosinase activity in melanoma cells modulates cytokine expression in co-cultured mononuclear cells, indicating that depigmentation of melanoma cells may activate immune cells and thereby influence a host anticancer response.
Collapse
Affiliation(s)
- Małgorzata Pawlikowska
- Department of Immunology, Faculty of Biology and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland; (T.J.); (S.W.)
- Correspondence: ; Tel.: +48-(56)-611-25-15
| | - Tomasz Jędrzejewski
- Department of Immunology, Faculty of Biology and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland; (T.J.); (S.W.)
| | - Andrzej T. Slominski
- Department of Dermatology, Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Laboratory Service of the VA Medical Center, Birmingham, AL 35294, USA
| | - Anna A. Brożyna
- Department of Human Biology, Faculty of Biology and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland;
| | - Sylwia Wrotek
- Department of Immunology, Faculty of Biology and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland; (T.J.); (S.W.)
| |
Collapse
|
5
|
Chen MX, Liu YM, Li Y, Yang X, Wei WB. Elevated VEGF-A & PLGF concentration in aqueous humor of patients with uveal melanoma following Iodine-125 plaque radiotherapy. Int J Ophthalmol 2020; 13:599-605. [PMID: 32399411 DOI: 10.18240/ijo.2020.04.11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
AIM To measure the concentration of vascular endothelial growth factor-A (VEGF-A), and placental growth factor (PLGF) in aqueous humor of uveal melanoma patients before and after Iodine-125 plaque therapy (IPT), determine the postoperative fluctuation and evaluate associated factors in vivo. METHODS Participants were 18 Chinese patients with uveal melanoma who were elected to IPT. Undiluted aqueous humor samples were collected at Iodine plaque implant and removal time, then stored immediately at -80°C until assayed. The concentration of VEGF-A, PLGF and other 7 cytokines comprising interleukin-2 (IL-2), IL-8, IL-10, interferon (IFN)-γ, programmed death (PD)-1, transforming growth factor (TGF)-β1 and insulin-like growth factor (IGF)-1 in aqueous humor was measured using Raybiotech immunoassay kit, a high throughput strategy. The VEGF-A and PLGF levels were compared across preoperation and postoperation subgroups, as well as those of other 7 interleukins. Correlation and grouped analyses were conducted to determine the independent effects of clinical parameters and other cytokines on VEGF-A and PLGF concentration or fluctuation. This study set a self-control design. RESULTS VEGF-A (P=0.038) and PLGF (P=0.026) were the only two increased cytokines after IPT. Preoperative and postoperative level of VEGF-A and PLGF (r=0.575, P=0.013; r=0.987, P<0.001) correlated with each other significantly. Level of VEGF-A (r=0.626, P=0.005; r=0.588, P=0.01) and PLGF (r=0.616, P=0.007; r=0.588, P=0.01) had positive correlation with tumor thickness consistently. Elevated VEGF-A or PLGF level were strong predictive factors of each other (P=0.007, OR=60.0). The elevated VEGF-A group showed a higher postoperative level of IFN-γ (P=0.005), IL-2 (P<0.001) and IL-10 (P=0.004) in aqueous humor. When the elevated PLGF group got similar results that a higher postoperative level of IFN-γ (P=0.007), IL-2 (P<0.001) and IL-10 (P=0.013) in aqueous humor. CONCLUSION This study reveals that VEGF-A and PLGF in aqueous humor significantly increased with tumor thickness and radiation process in uveal melanoma patients. VEGF-A and PLGF may be crucial in uveal melanoma genesis and radiotherapy reactions. Immune mediators comprised IFN-γ, IL-2 and IL-10 could play roles in the link between inflammation and angiogenesis in uveal melanoma when exposed to radiotherapy.
Collapse
Affiliation(s)
- Meng-Xi Chen
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Key Laboratory of Intraocular tumor Diagnosis and Treatment, Beijing 100730, China
| | - Yue-Ming Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Key Laboratory of Intraocular tumor Diagnosis and Treatment, Beijing 100730, China
| | - Yang Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Key Laboratory of Intraocular tumor Diagnosis and Treatment, Beijing 100730, China
| | - Xuan Yang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Key Laboratory of Intraocular tumor Diagnosis and Treatment, Beijing 100730, China
| | - Wen-Bin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Key Laboratory of Intraocular tumor Diagnosis and Treatment, Beijing 100730, China
| |
Collapse
|
6
|
Dillman RO, Cornforth AN, Nistor GI, McClay EF, Amatruda TT, Depriest C. Randomized phase II trial of autologous dendritic cell vaccines versus autologous tumor cell vaccines in metastatic melanoma: 5-year follow up and additional analyses. J Immunother Cancer 2018; 6:19. [PMID: 29510745 PMCID: PMC5840808 DOI: 10.1186/s40425-018-0330-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/26/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Despite improved survival following checkpoint inhibitors, there is still a potential role for anti-cancer therapeutic vaccines. Because of biological heterogeneity and neoantigens resulting from each patient's mutanome, autologous tumor may be the best source of tumor-associated antigens (TAA) for vaccines. Ex vivo loading of autologous dendritic cells with TAA may be associated with superior clinical outcome compared to injecting irradiated autologous tumor cells. We conducted a randomized phase II trial to compare autologous tumor cell vaccines (TCV) and autologous dendritic cell vaccines (DCV) loaded with autologous TAA. METHODS Short-term autologous tumor cell lines were established from metastatic tumor. Vaccines were admixed with 500 micrograms of GM-CSF and injected weekly for 3 weeks, then at weeks 8, 12,16, 20, and 24. The primary endpoint was overall survival. Secondary objectives were identification of adverse events, and results of delayed type hypersensitivity (DTH) reactions to intradermal tumor cell injections. RESULTS Forty-two patients were randomized. All were followed from randomization until death or for five years; none were lost to follow-up. DCV was associated with longer survival: median 43.4 versus 20.5 months (95% CI, 18.6 to > 60 versus 9.3 to 32.3 months) and a 70% reduction in the risk of death (hazard ratio = 0.304, p = 0.0053, 95% CI, 0.131 to 0.702). Tumor DTH reactions were neither prognostic nor predictive. The most common treatment-related adverse events were mild to moderate local injection site reactions and flu-like symptoms; but grade 2 treatment-related adverse events were more frequent with TCV. Serum marker analyses at week-0 and week-4 showed that serum markers were similar at baseline in each arm, but differed after vaccination. CONCLUSIONS This is the only human clinical trial comparing DCV and TCV as platforms for autologous TAA presentation. DCV was associated with minimal toxicity and long-term survival in patients with metastatic melanoma. DTH to autologous tumor cells was neither prognostic for survival nor predictive of benefit for either vaccine. TRIAL REGISTRATION Clinical trials.gov NCT00948480 retrospectively registered 28 July 2009.
Collapse
Affiliation(s)
- Robert O. Dillman
- Hoag Cancer Institute, Newport Beach, CA 92660 USA
- AIVITA Biomedical, Inc., Irvine, CA USA
| | | | | | - Edward F. McClay
- California Cancer Associates for Research and Excellence (cCARE), Institute for Melanoma Research & Education, Encinitas, CA USA
| | | | | |
Collapse
|
7
|
|
8
|
Dillman RO. Long-Term Progression-Free and Overall Survival in Two Melanoma Patients Treated with Patient-Specific Therapeutic Vaccine Eltrapuldencel-T After Resection of a Solitary Liver Metastasis. Cancer Biother Radiopharm 2016; 31:71-4. [PMID: 27093340 DOI: 10.1089/cbr.2016.2003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hepatic metastases from melanoma are usually associated with recurrence and short survival, even in patients with a solitary metastasis. Two patients, one with melanoma of unknown primary and one with ocular melanoma, underwent resection of a solitary liver metastasis followed by treatment with eltrapuldencel-T, a patient-specific therapeutic vaccine consisting of autologous dendritic cells loaded with antigens from irradiated melanoma cells obtained from an autologous tumor cell line. Following surgical resection, the ocular melanoma patient remained progression free for more than 4.5 years and was known to be alive more than 8.5 years later, while the other patient, who previously had experienced lung and small bowel metastases, has remained disease free and is alive more than 12 years later. These two cases illustrate how immunotherapies designed to induce immune responses to tumor-associated antigens (TAA), as opposed to releasing previously existing responses to TAA that have been suppressed, may also enhance long-term disease control and survival.
Collapse
|
9
|
Dillman RO, McClay EF, Barth NM, Amatruda TT, Schwartzberg LS, Mahdavi K, de Leon C, Ellis RE, DePriest C. Dendritic Versus Tumor Cell Presentation of Autologous Tumor Antigens for Active Specific Immunotherapy in Metastatic Melanoma: Impact on Long-Term Survival by Extent of Disease at the Time of Treatment. Cancer Biother Radiopharm 2016; 30:187-94. [PMID: 26083950 PMCID: PMC4492594 DOI: 10.1089/cbr.2015.1843] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In patients with metastatic melanoma, sequential single-arm and randomized phase II trials with a therapeutic vaccine consisting of autologous dendritic cells (DCs) loaded with antigens from self-renewing, proliferating, irradiated autologous tumor cells (DC-TC) showed superior survival compared with similar patients immunized with irradiated tumor cells (TC). We wished to determine whether this difference was evident in cohorts who at the time of treatment had (1) no evidence of disease (NED) or (2) had detectable disease. Eligibility criteria and treatment schedules were the same for all three trials. Pooled data confirmed that overall survival (OS) was longer in 72 patients treated with DC-TC compared with 71 patients treated with TC (median OS 60 versus 22 months; 5-year OS 51% versus 32%, p=0.004). Treatment with DC-TC was associated with longer OS in both cohorts. Among 70 patients who were NED at the time that treatment was started, OS was better for DC-TC: 5-year OS 73% versus 43% (p=0.015). Among 73 patients who had detectable metastases, OS was better for DC-TC: median 38.8 months versus 14.7 months, 5-year OS 33% versus 20% (p=0.025). This approach is promising as an adjunct to other therapies in patients who have had metastatic melanoma.
Collapse
Affiliation(s)
| | - Edward F McClay
- 2 California Cancer Associates for Research and Excellence (cCARE) , Institute for Melanoma Research & Education , Encinitas California
| | - Neil M Barth
- 3 Genomics Institute Inc. , Laguna Beach, California
| | | | | | | | - Cristina de Leon
- 7 Hoag Institute for Research and Education , Newport Beach, California
| | - Robin E Ellis
- 7 Hoag Institute for Research and Education , Newport Beach, California
| | - Carol DePriest
- 8 Cancer Biotherapy Research Group , Franklin, Tennessee
| |
Collapse
|
10
|
Sarkar D, Leung EY, Baguley BC, Finlay GJ, Askarian-Amiri ME. Epigenetic regulation in human melanoma: past and future. Epigenetics 2015; 10:103-21. [PMID: 25587943 PMCID: PMC4622872 DOI: 10.1080/15592294.2014.1003746] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The development and progression of melanoma have been attributed to independent or combined genetic and epigenetic events. There has been remarkable progress in understanding melanoma pathogenesis in terms of genetic alterations. However, recent studies have revealed a complex involvement of epigenetic mechanisms in the regulation of gene expression, including methylation, chromatin modification and remodeling, and the diverse activities of non-coding RNAs. The roles of gene methylation and miRNAs have been relatively well studied in melanoma, but other studies have shown that changes in chromatin status and in the differential expression of long non-coding RNAs can lead to altered regulation of key genes. Taken together, they affect the functioning of signaling pathways that influence each other, intersect, and form networks in which local perturbations disturb the activity of the whole system. Here, we focus on how epigenetic events intertwine with these pathways and contribute to the molecular pathogenesis of melanoma.
Collapse
Key Words
- 5hmC, 5-hydroxymethylcytosine
- 5mC, 5-methylcytosine
- ACE, angiotensin converting enzyme
- ANCR, anti-differentiation non-coding RNA
- ANRIL, antisense noncoding RNA in INK4 locus
- ASK1, apoptosis signal-regulating kinase 1
- ATRA, all-trans retinoic acid
- BANCR, BRAF-activated non-coding RNA
- BCL-2, B-cell lymphoma 2
- BRAF, B-Raf proto-oncogene, serine/threonine kinase
- BRG1, ATP-dependent helicase SMARCA4
- CAF-1, chromatin assembly factor-1
- CBX7, chromobox homolog 7
- CCND1, cyclin D1
- CD28, cluster of differentiation 28
- CDK, cyclin-dependent kinase
- CDKN2A/B, cyclin-dependent kinase inhibitor 2A/B
- CHD8, chromodomain-helicase DNA-binding protein 8
- CREB, cAMP response element-binding protein
- CUDR, cancer upregulated drug resistant
- Cdc6, cell division cycle 6
- DNA methylation/demethylation
- DNMT, DNA methyltransferase
- EMT, epithelial-mesenchymal transition
- ERK, extracellular signal-regulated kinase
- EZH2, enhancer of zeste homolog 2
- GPCRs, G-protein coupled receptors
- GSK3a, glycogen synthase kinase 3 α
- GWAS, genome-wide association study
- HDAC, histone deacetylase
- HOTAIR, HOX antisense intergenic RNA
- IAP, inhibitor of apoptosis
- IDH2, isocitrate dehydrogenase
- IFN, interferon, interleukin 23
- JNK, Jun N-terminal kinase
- Jak/STAT, Janus kinase/signal transducer and activator of transcription
- MAFG, v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog G
- MALAT1, metastasis-associated lung adenocarcinoma transcript 1
- MAPK, mitogen-activated protein kinase
- MC1R, melanocortin-1 receptor
- MGMT, O6-methylguanine-DNA methyltransferase
- MIF, macrophage migration inhibitory factor
- MITF, microphthalmia-associated transcription factor
- MRE, miRNA recognition element
- MeCP2, methyl CpG binding protein 2
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NOD, nucleotide-binding and oligomerization domain
- PBX, pre-B-cell leukemia homeobox
- PEDF, pigment epithelium derived factor
- PI3K, phosphatidylinositol-4, 5-bisphosphate 3-kinase
- PIB5PA, phosphatidylinositol-4, 5-biphosphate 5-phosphatase A
- PKA, protein kinase A
- PRC, polycomb repressor complex
- PSF, PTB associated splicing factor
- PTB, polypyrimidine tract-binding
- PTEN, phosphatase and tensin homolog
- RARB, retinoic acid receptor-β2
- RASSF1A, Ras association domain family 1A
- SETDB1, SET Domain, bifurcated 1
- SPRY4, Sprouty 4
- STAU1, Staufen1
- SWI/SNF, SWItch/Sucrose Non-Fermentable
- TCR, T-cell receptor
- TET, ten eleven translocase
- TGF β, transforming growth factor β
- TINCR, tissue differentiation-inducing non-protein coding RNA
- TOR, target of rapamycin
- TP53, tumor protein 53
- TRAF6, TNF receptor-associated factor 6
- UCA1, urothelial carcinoma-associated 1
- ceRNA, competitive endogenous RNAs
- chromatin modification
- chromatin remodeling
- epigenetics
- gene regulation
- lncRNA, long ncRNA
- melanoma
- miRNA, micro RNA
- ncRNA, non-coding RNA
- ncRNAs
- p14ARF, p14 alternative reading frame
- p16INK4a, p16 inhibitor of CDK4
- pRB, retinoblastoma protein
- snoRNA, small nucleolar RNA
- α-MSHm, α-melanocyte stimulating hormone
Collapse
Affiliation(s)
- Debina Sarkar
- a Auckland Cancer Society Research Center ; University of Auckland ; Auckland , New Zealand
| | | | | | | | | |
Collapse
|
11
|
Whiteside TL, Ferris RL, Szczepanski M, Tublin M, Kiss J, Johnson R, Johnson JT. Dendritic cell-based autologous tumor vaccines for head and neck squamous cell carcinoma. Head Neck 2015; 38 Suppl 1:E494-501. [PMID: 25735641 DOI: 10.1002/hed.24025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2015] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND An autologous vaccine of apoptotic tumor cells (ATCs) and dendritic cells (DCs) was administered to patients with stage III/IV head and neck squamous cell carcinoma (HNSCC) to study safety and feasibility. METHODS Autologous DCs were generated from monocytes, loaded with ATCs, and delivered intranodally. Delayed-type hypersensitivity (DTH) and immunological endpoints were measured prevaccination and postvaccination. Clinical follow-up was required. RESULTS Tumors obtained from 30 patients yielded 2 × 10(6) to 2 × 10(8) tumor cells. Only 19 of 30 (63%) were sterile. Ten of 30 patients (33%) had ≥1 × 10(7) sterile tumor cells required for vaccine production. Eight of 10 patients had positive recall DTH. Five of 10 patients were leukapheresed to generate DCs. Four of 5 patients were vaccinated. ATC-reactive T cells were detected in 3 of 4 patients. All 4 patients survived >5 years. The trial failed to enroll the projected 12 patients and was terminated. CONCLUSION This vaccine was safe and immunogenic but feasible only in patients with HNSCC with positive prevaccine DTH and ≥1 × 10(7) sterile tumor cells. All vaccinated patients were long-term disease-free survivors. © 2015 Wiley Periodicals, Inc. Head Neck 38: E494-E501, 2016.
Collapse
Affiliation(s)
- Theresa L Whiteside
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Miroslaw Szczepanski
- University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Mitchell Tublin
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joseph Kiss
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Rita Johnson
- University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Jonas T Johnson
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Matikas A, Mavroudis D. Beyond CTLA-4: novel immunotherapy strategies for metastatic melanoma. Future Oncol 2015; 11:997-1009. [PMID: 25760979 DOI: 10.2217/fon.14.287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The treatment of metastatic melanoma is rapidly evolving. The discovery of BRAF and MEK inhibitors was an important milestone. Unfortunately, although response rates are high, disease progression is universal. Despite the success of IL-2 and adjuvant IFL-α2b, these two agents remained the only approved immunotherapy approaches. In recent years, the use of immunotherapy has drawn attention with the recognition of the mechanisms of tumor immune evasion. Blockade of these mechanisms may improve outcomes. However, the potential adverse events, the optimal use of these modalities, the high cost and the absence of predictive markers remain unmet challenges. Herein, we review the immunotherapy strategies in melanoma, either approved or under evaluation, and present the relevant data concerning their efficacy and safety.
Collapse
Affiliation(s)
- Alexios Matikas
- Department of Medical Oncology, University Hospital of Heraklion, Heraklion, Crete, Greece
| | | |
Collapse
|
13
|
dePillis LG, Eladdadi A, Radunskaya AE. Modeling cancer-immune responses to therapy. J Pharmacokinet Pharmacodyn 2014; 41:461-78. [DOI: 10.1007/s10928-014-9386-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/17/2014] [Indexed: 12/26/2022]
|