Skip to main content
Erschienen in: Molecular Cancer 1/2021

Open Access 01.12.2021 | Letter to the Editor

Tumor-antigens and immune landscapes identification for prostate adenocarcinoma mRNA vaccine

verfasst von: Xiaonan Zheng, Hang Xu, Xianyanling Yi, Tianyi Zhang, Qiang Wei, Hong Li, Jianzhong Ai

Erschienen in: Molecular Cancer | Ausgabe 1/2021

Abstract

Prostate adenocarcinoma (PRAD) is a leading cause of death among men. Messenger ribonucleic acid (mRNA) vaccine presents an attractive approach to achieve satisfactory outcomes; however, tumor antigen screening and vaccination candidates show a bottleneck in this field. We aimed to investigate the tumor antigens for mRNA vaccine development and immune subtypes for choosing appropriate patients for vaccination. We identified eight overexpressed and mutated tumor antigens with poor prognostic value of PRAD, including KLHL17, CPT1B, IQGAP3, LIME1, YJEFN3, KIAA1529, MSH5 and CELSR3. The correlation of those genes with antigen-presenting immune cells were assessed. We further identified three immune subtypes of PRAD (PRAD immune subtype [PIS] 1–3) with distinct clinical, molecular, and cellular characteristics. PIS1 showed better survival and immune cell infiltration, nevertheless, PIS2 and PIS3 showed cold tumor features with poorer prognosis and higher tumor genomic instability. Moreover, these immune subtypes presented distinguished association with immune checkpoints, immunogenic cell death modulators, and prognostic factors of PRAD. Furthermore, immune landscape characterization unraveled the immune heterogeneity among patients with PRAD. To summarize, our study suggests KLHL17, CPT1B, IQGAP3, LIME1, YJEFN3, KIAA1529, MSH5 and CELSR3 are potential antigens for PRAD mRNA vaccine development, and patients in the PIS2 and PIS3 groups are more suitable for vaccination.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12943-021-01452-1.
Xiaonan Zheng, Hang Xu and Xianyanling Yi contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
PRAD
Prostate adenocarcinoma
mRNA
Messenger ribonucleic acid
TCGA
The Cancer Genome Atlas
GEO
Gene Expression Omnibus
KLHL17
Kelch Like Family Member 17
CPT1B
Carnitine Palmitoyltransferase 1B
IQGAP3
IQ Motif Containing GTPase Activating Protein 3
LIME1
Lck Interacting Transmembrane Adaptor 1
YJEFN3
YjeF N-Terminal Domain Containing 3
MSH5
MutS Homolog 5
CELSR3
Cadherin EGF LAG seven-pass G-type receptor 3
PIS
PRAD immune subtype
PD-1
Programmed cell death protein 1
PD-L
Programmed cell death protein ligand 1
CTLA4
Cytotoxic T lymphocyte antigen 4
CRPC
Castration-resistant prostate cancer
OS
Overall survival
APCs
Antigen-presenting cells
DFI
Disease-free interval
ICI
Immune-checkpoint inhibitors
TIP
Tracking Tumor Immunophenotype
TIMER
Tumor IMmune Estimation Resource
PAM
Partition around medoids
MSigDB
The Molecular Signatures Database
TMB
Tumor mutation burden
CNV
Copy number variation
HRD
Homologous recombination deficiency
NtAI
Allelic imbalance
LST
Large scale transition
LOH
Loss of heterozygosity
mRNAsi
mRNA stemness index
ICP
Immune-checkpoint
ICD
Immunogenic cell death
WGCNA
Weighted correlation network analysis
DEG
Differential expression gene
DCs
Dendritic cells
HOXC6
Homeobox C6
PDK4
Pyruvate Dehydrogenase Kinase 4
STAT3
Signal Transducer and Activator of Transcription 3
HR
Hazard ratio
CDC20
Cell Division Cycle 20
ESPL1
Extra Spindle Pole Bodies Like 1
MAPK8IP3
Mitogen-Activated Protein Kinase 8 Interacting Protein 3
AUC
Area under the curve
ICB
Immune-checkpoint blockade

Background

Prostate adenocarcinoma (PRAD) is the second diagnosed and the fifth death-related malignancy among men worldwide [1]. Positive responses in patients with PRAD were rarely observed after immunotherapies including programmed cell death protein 1 (PD-1), PD-Ligand 1 (PD-L1), or cytotoxic T lymphocyte antigen 4 (CTLA4). Previously, Sipuleucel-T brought prostate cancer immunotherapy into a sharper focus whereas no significant effect was reported regarding progression-free survival [2, 3]. Therefore, novel therapeutics should be developed for effective PRAD treatment. In the past 2 years, under the background of coronavirus disease-2019 pandemic, the enthusiasm for mRNA vaccine development, showing advantages of flexibility, productivity, non-genomic integration, and low immunogenicity [4], was also brought into the field of cancer therapy [5].
Previous phase I/II clinical trial showed good tolerability and favorable immune activation of mRNA vaccines CV9103 for PRAD; however, the subsequent clinical trials of CV9104 containing two more antigens (prostatic acid phosphatase [PAP] and Mucin-1) were terminated due to failure of improving the overall survival (OS) [6]. These findings indicated that antigen selection is critical for activating antigen-presenting cells (APCs) and immune response. Moreover, the identification of immune subtypes of patients with PRAD for mRNA vaccination is another crucial factor for the curative effect [7]. Hence, this study, exploring novel candidate tumor antigens for PRAD mRNA vaccine and identifying suitable patients for vaccination, aims to pave an avenue for the application of mRNA vaccine in PRAD population.

Results and discussion

Identification of potential tumor antigens of PRAD

A total of 733 overexpressed genes in TCGA-PRAD samples were identified (Fig. S1A, B) and their distribution in chromosomes was shown in Fig. 1A. We then identified 10881 genes that potentially encode tumor-specific antigens through calculating the fraction of altered genome and tumor mutational counts (Fig. 1B, C). Ten genes with the highest altered genome fractions and mutation counts were displayed in Fig. S1C and D. The 733 overexpressed genes were then intersected with the 10881 mutated tumor antigen-encoding genes, and 311 genes were identified afterwards (Fig. S1E). Cox regression revealed that 13 genes were significantly associated with OS (Fig. 1D) and 70 genes were significantly associated with disease free interval (DFI) (Fig. 1E). Further intersection analysis indicated eight genes, including KLHL17, CPT1B, IQGAP3, LIME1, YJEFN3, MSH5, CELSR3 and KIAA1529, were correlated with both OS and DFI (Fig. 1F). The Kaplan–Meier survival curves of OS for those eight genes were shown in Fig. 1G, and the higher expression of them was indicative of worse survival. The correlation between them and the infiltration of major APCs, including B cells, macrophages as well as dendritic cells (DCs), was also analyzed. Figure 1H showed a significantly positive correlation of IQGAP3, CELSR3, and KIAA1529 with APCs, whereas negative for CPT1B (Fig. S1F). Taken together, our evidence identified tumor-specific antigens owning potentiality of mRNA vaccine development for PRAD.
To the best of our knowledge, our study is the first to systematically screen suitable tumor antigens for mRNA vaccine development in PRAD. Despite the non-application of these antigens in mRNA vaccine development until the present, some of these antigens have been functionally explored in previous studies. For instance, CPT1B silencing could reduce cell proliferation and invasion in PRAD cell lines and its expression level might be regulated by androgen receptors [8]. IQGAP3 was upregulated in most cancer types and predicted a poor prognosis. It might also participate in the Paris forrestii antitumor effect [9]. Besides, CELSR3 downregulation significantly suppresses PRAD cell proliferation and migration [10]. MSH5 has been reported as a pleiotropic susceptibility locus for several cancers and was identified as a novel candidate gene warranting additional follow-up as a prospective PRAD risk locus [11]. However, KLHL17, KIAA1529, LIME1, and YJEFN3 were not fully elucidated in PRAD or other cancers. Their function in cancers especially PRAD warrants further exploration.

Identification and validation of the PRAD immune subtypes

A total of 13426 immunogenic genes were obtained from the MSigDB c7 datasets, 23 of which were associated with predictive survival outcomes through Lasso regression. The PAM algorithm accordingly identified the optimal number of clusters as three based on the training cohort (Fig. 2A). The accumulative curve and delta area of clustering were displayed in Fig. S2A and B. Principal component analysis showed the distribution of TCGA-PRAD individuals in each cluster (Fig. S2C), and heatmap revealed the differential expression of partial immunogenic genes across the three clusters (Fig. S2D). Importantly, the PRAD immune subtype 1 (PIS1) consistently had better survival outcomes compared to the PIS2 and PIS3 in both training cohort (P < 0.0001) and validation cohort (P = 0.041) (Fig. 2B and S2C).

Clinical, mutational and immunological features of the PRAD immune subtypes

Clinical features of the PRAD immune subtypes were assessed. The predicted response to immunotherapy of the subtypes indicated that PIS2 and PIS3 were more likely to respond to anti-PD-L1 treatment (Fig. 2D). PIS3 had a higher frequency of biochemical recurrence (Fig. 2E) and pathological N1 stage and higher pathological T stage (Fig. S2E, F). Moreover, patients in PIS3 also had a higher risk of receiving radiation therapy (Fig. S2G). This evidence implies that PIS2 and PIS3 are associated with more aggressive clinical features and more suitable for immunotherapy.
The correlation of existing PRAD biomarkers with PRAD immune subtypes was evaluated. The expression of HOXC6, whose higher expression indicated short survival and higher recurrence rate of PRAD [12], was significantly higher in PIS2 and PIS3 than in PIS1 (Fig. S3A). Nevertheless, PDK4 and STAT3, two classical genes whose low expression was associated with worse survival of PRAD [13, 14], were significantly less expressed in PIS2 and PIS3 (Fig. S3B and C). Notably, our PRAD immune subtypes were also compared with previously published pan-cancer immune subtypes. Figure 2F demonstrated that the C3 subtype had a decreasing distribution, and C1 and C2 showed an increasing tendency across PIS1 to PIS3. Interestingly, C3 was claimed to be a positive marker of prognosis but C1 and C2 were negatively associated with survival [15], which was consistent with our findings.
In terms of mutation status, PIS3 and PIS2 presented more frequent CNV (either gain or loss) across the chromosomes (Figs. 2G and S3D). Similarly, it could be seen from the mutation landscape of the immune subtypes that PIS3 had a more frequent mutation of the top 20 most mutated genes (Fig. S3E), and PIS2 and PIS3 had significantly higher tumor mutation counts (Fig. 2H). In addition, tumor mutation burden (TMB) was also found to be significantly heavier in PIS2 (P = 0.0053) and PIS3 (P < 0.0001) than in PIS1 (Fig. 2I). Our results also demonstrated that PIS2 and PIS3 had higher telomeric allelic imbalance (HRD-NtAI), large scale transition (HRD-LST) and loss of heterozygosity (HRD-LOH) and combined homologous recombination deficiency (HRD scores) (Fig. 2J, S3F-H). Moreover, mRNA stemness index (mRNAsi) was also higher in PIS2 and PIS3 compared to that of PIS1 (Fig. 2K).
As shown in Fig. S4A and D, the tendency of stromal score, immune score, and tumor purity was variable across the subtypes in both training and validation cohorts. In the training cohort, PIS3 had richer infiltration of M2 macrophages and memory B cells, but PIS1 had more infiltration of naïve B cells and memory-resting CD4+ T cells (Fig. S4B, C). Consistently, PIS3 still had a higher degree of memory B cell infiltration compared to PIS1 and PIS2 in the validation cohort (Fig. S4E, F). Hence, PIS3 implies having a better performance of presenting tumor antigen during the immune response.
The anticancer immune activity of the three immune subtypes was calculated with the TIP analysis. PIS1 performed better at recruiting CD4+ T cells, Th22 cells, and monocytes, whereas PIS2 and PIS3 were still proved to be better at recruiting B cells (Fig. S5A). These outcomes may explain the better survival of PIS1 and also indicate the suitability of PIS2 and PIS3 for receiving tumor vaccines. As for the immune modulators, a total of 37 ICP genes and 25 ICD genes were analyzed, which revealed that 31 ICP genes and 18 ICD genes were differentially expressed across the immune subtypes in the training cohort from TCGA datasets (Fig. S5B, C). Interestingly, fewer ICPs and ICDs were significantly differentially expressed among three clusters in the GEO cohorts, and the PIS3 cluster showed markedly lower ICP and ICD expression (Fig. S5D, E). These findings indicated that the immunotyping showed a distinct expression pattern of ICPs and ICDs, and these modulators could be utilized as potential markers for treatment with mRNA vaccines.

Immune subtype-based landscape of PRAD

The gene expression value of each patient across the three PRAD immune subtypes was used to build the immune landscape of PRAD (Fig. 2L), with PIS1, PIS2 and PIS3 were generally distributed in different branches of the tree. Principal component 1 (horizontal axis) was positively correlated with plasma cells and M2 macrophages, but negatively correlated with naïve B cells, resting DCs and M1macrophages. Interestingly, principal component 2 (vertical axis) had a positive correlation with DCs and naïve B cells, but a negative correlation with M0 and M1 macrophages (Fig. 2M). The general distribution of PIS1 can be observed to contrast PIS3. Also, individuals within the same immune subtype of PIS1 and PIS3 showed opposing distribution. Therefore, PIS1 and PIS3 were further divided (Fig. 2N), which turned out that PIS1A had a generally higher enrichment score regarding activated DCs and memory B cells compared to PIS1B (Fig. S6A). Similarly, PIS3A had a higher enrichment score of activated DCs, activated B cells and memory B cells than PIS3B (Fig. S6B). Therefore, tumor antigen may be more effective in PIS1A and PIS3A compared to PIS1B and PIS3B, respectively. Besides, individuals with extreme distribution in the immune landscape (Fig. 2O) were taken into further survival analysis. Group N1 was associated with the worst survival and group N3 had the best survival outcomes (P = 0.0011) (Fig. 2P). The immune subtype-based landscape can potentially designate the precise mRNA vaccine therapeutics for patients with PRAD by identifying immune components of patients with PRAD and predict survival.

Weighted immunogenic gene co-expression network of PRAD

WGCNA with a fixed soft threshold of nine was used to construct the immunogenic gene co-expression network of PRAD (Fig. S7A-C). Eventually, 9 co-expression modules were obtained (Fig. S7D, E). Distribution analysis showed that PRAD immune subtypes were differentially distributed in most of the modules (Fig. S7F). The negative prognostic value of the pink (Hazard ratio (HR) 2.30, 95% CI 1.59–3.32), magenta (HR 2.19, 95% CI 1.56–3.08), and purple modules (HR 1.61, 95% CI 1.16–2.24) were presented in Fig. S8A. The biological function of the prognostic modules, including B cell activation, and regulation of adaptive immune response were also displayed in Fig. S8B–D. 62 hub genes in the prognostic modules were identified, and three of them (CDC20, ESPL1, MAPK8IP3) were eventually selected after multivariate Cox regression (Fig. S8E, F). Patients were divided into the high-risk and low-risk groups. KM curve demonstrated that the high-risk group had worse survival (P = 0.0011) and the area under the receiver operating curve (AUC) was 0.852, indicating a good accuracy of the model (Fig. S8G, H). Thus, this risk model based on immunogenic genes co-expression network may work as a novel biomarker for predicting the prognosis.

DEG-based risk model construction

391 DEGs across PRAD immune subtypes were found and displayed (Fig. S9A, B). The prognostic value of these DEGs was calculated and 93 DEGs were prognostic. Lasso regression reduced the dimension of these DEGs and 21 genes were finally used to construct the risk model (Fig. S9C–F). The risk of each patient was calculated based on the expression value of the 21 genes and their coefficients in Lasso regression (Fig. S9G, H). Patients were categorized into high-risk or low-risk groups, and the high-risk group had worse survival with an AUC of 0.892 (Fig. S10A, B). Consistently, Fig. S10C and D summarized that PIS2 and PIS3 had higher risk scores with more PIS1 were distributed in the low-risk group, which in reverse proved the accuracy of PRAD immune subtype in predicting PRAD prognosis.

Conclusions

In this study, KLHL17, CPT1B, IQGAP3, LIME1, YJEFN3, KIAA1529, MSH5 and CELSR3 were identified as potential tumor-specific antigens for PRAD mRNA vaccine development. PRAD patients of PIS2 and PIS3 might be suitable candidates of vaccination. These findings provided new sights in selecting antigens and populations for future PRAD mRNA vaccine development and application.

Methods and availability of supporting data

Methods and materials used in our study are attached as Supplementary information. All data are freely available from the public databases and the other necessary and reasonable information could be obtained from the corresponding author.

Acknowledgements

We would like to thank TCGA and GEO datasets for data mining.

Declarations

Not applicable
Not applicable

Competing interests

None
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.CrossRef Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.CrossRef
2.
Zurück zum Zitat Sinha M, Zhang L, Subudhi S, Chen B, Marquez J, Liu EV, et al. Pre-existing immune status associated with response to combination of sipuleucel-T and ipilimumab in patients with metastatic castration-resistant prostate cancer. J Immunother Cancer. 2021;9:e002254.CrossRef Sinha M, Zhang L, Subudhi S, Chen B, Marquez J, Liu EV, et al. Pre-existing immune status associated with response to combination of sipuleucel-T and ipilimumab in patients with metastatic castration-resistant prostate cancer. J Immunother Cancer. 2021;9:e002254.CrossRef
3.
Zurück zum Zitat Sartor O, Armstrong AJ, Ahaghotu C, McLeod DG, Cooperberg MR, Penson DF, et al. Survival of African-American and Caucasian men after sipuleucel-T immunotherapy: outcomes from the PROCEED registry. Prostate Cancer Prostatic Dis. 2020;23:517–26.CrossRef Sartor O, Armstrong AJ, Ahaghotu C, McLeod DG, Cooperberg MR, Penson DF, et al. Survival of African-American and Caucasian men after sipuleucel-T immunotherapy: outcomes from the PROCEED registry. Prostate Cancer Prostatic Dis. 2020;23:517–26.CrossRef
4.
Zurück zum Zitat Wang Y, Zhang Z, Luo J, Han X, Wei Y, Wei X. mRNA vaccine: a potential therapeutic strategy. Mol Cancer. 2021;20:33.CrossRef Wang Y, Zhang Z, Luo J, Han X, Wei Y, Wei X. mRNA vaccine: a potential therapeutic strategy. Mol Cancer. 2021;20:33.CrossRef
5.
Zurück zum Zitat Heine A, Juranek S, Brossart P. Clinical and immunological effects of mRNA vaccines in malignant diseases. Mol Cancer. 2021;20:52.CrossRef Heine A, Juranek S, Brossart P. Clinical and immunological effects of mRNA vaccines in malignant diseases. Mol Cancer. 2021;20:52.CrossRef
6.
Zurück zum Zitat Stenzl A, Feyerabend S, Syndikus I, Sarosiek T, Kübler H, Heidenreich A, et al. Results of the randomized, placebo-controlled phase I/IIB trial of CV9104, an mRNA based cancer immunotherapy, in patients with metastatic castration-resistant prostate cancer (mCRPC). Annals of Oncology. 2017;28:408–9.CrossRef Stenzl A, Feyerabend S, Syndikus I, Sarosiek T, Kübler H, Heidenreich A, et al. Results of the randomized, placebo-controlled phase I/IIB trial of CV9104, an mRNA based cancer immunotherapy, in patients with metastatic castration-resistant prostate cancer (mCRPC). Annals of Oncology. 2017;28:408–9.CrossRef
7.
Zurück zum Zitat Amin A, Dudek AZ, Logan TF, Lance RS, Holzbeierlein JM, Knox JJ, et al. Survival with AGS-003, an autologous dendritic cell-based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): Phase 2 study results. J Immunother Cancer. 2015;3:14.CrossRef Amin A, Dudek AZ, Logan TF, Lance RS, Holzbeierlein JM, Knox JJ, et al. Survival with AGS-003, an autologous dendritic cell-based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): Phase 2 study results. J Immunother Cancer. 2015;3:14.CrossRef
8.
Zurück zum Zitat Abudurexiti M, Zhu W, Wang Y, Wang J. Targeting CPT1B as a potential therapeutic strategy in castration-resistant and enzalutamide-resistant prostate cancer. Prostate. 2020;80:950–61.CrossRef Abudurexiti M, Zhu W, Wang Y, Wang J. Targeting CPT1B as a potential therapeutic strategy in castration-resistant and enzalutamide-resistant prostate cancer. Prostate. 2020;80:950–61.CrossRef
9.
Zurück zum Zitat Xia C, Chen L, Sun W, Yan R, Xia M, Wang Y, et al. Total saponins from Paris forrestii (Takht) H. Li. show the anticancer and RNA expression regulating effects on prostate cancer cells. Biomed Pharmacother. 2020;121:109674.CrossRef Xia C, Chen L, Sun W, Yan R, Xia M, Wang Y, et al. Total saponins from Paris forrestii (Takht) H. Li. show the anticancer and RNA expression regulating effects on prostate cancer cells. Biomed Pharmacother. 2020;121:109674.CrossRef
10.
Zurück zum Zitat Chen X, Ma Q, Liu Y, Li H, Liu Z, Zhang Z, et al. Increased expression of CELSR3 indicates a poor prognostic factor for prostate cancer. J Cancer. 2021;12:1115–24.CrossRef Chen X, Ma Q, Liu Y, Li H, Liu Z, Zhang Z, et al. Increased expression of CELSR3 indicates a poor prognostic factor for prostate cancer. J Cancer. 2021;12:1115–24.CrossRef
11.
Zurück zum Zitat Saunders EJ, Dadaev T, Leongamornlert DA, Al Olama AA, Benlloch S, Giles GG, et al. Gene and pathway level analyses of germline DNA-repair gene variants and prostate cancer susceptibility using the iCOGS-genotyping array. Br J Cancer. 2018;118:e9.CrossRef Saunders EJ, Dadaev T, Leongamornlert DA, Al Olama AA, Benlloch S, Giles GG, et al. Gene and pathway level analyses of germline DNA-repair gene variants and prostate cancer susceptibility using the iCOGS-genotyping array. Br J Cancer. 2018;118:e9.CrossRef
12.
Zurück zum Zitat Zhou J, Yang X, Song P, Wang H, Wang X. HOXC6 in the prognosis of prostate cancer. Artif Cells Nanomed Biotechnol. 2019;47:2715–20.CrossRef Zhou J, Yang X, Song P, Wang H, Wang X. HOXC6 in the prognosis of prostate cancer. Artif Cells Nanomed Biotechnol. 2019;47:2715–20.CrossRef
13.
Zurück zum Zitat Oberhuber M, Pecoraro M, Rusz M. STAT3-dependent analysis reveals PDK4 as independent predictor of recurrence in prostate cancer. Mol Syst Biol. 2020;16:e9247.CrossRef Oberhuber M, Pecoraro M, Rusz M. STAT3-dependent analysis reveals PDK4 as independent predictor of recurrence in prostate cancer. Mol Syst Biol. 2020;16:e9247.CrossRef
14.
Zurück zum Zitat Atas E, Oberhuber M, Kenner L. The implications of PDK1-4 on tumor energy metabolism, aggressiveness and therapy resistance. Front Oncol. 2020;10:583217.CrossRef Atas E, Oberhuber M, Kenner L. The implications of PDK1-4 on tumor energy metabolism, aggressiveness and therapy resistance. Front Oncol. 2020;10:583217.CrossRef
15.
Zurück zum Zitat Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2019;51:411–2.CrossRef Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2019;51:411–2.CrossRef
Metadaten
Titel
Tumor-antigens and immune landscapes identification for prostate adenocarcinoma mRNA vaccine
verfasst von
Xiaonan Zheng
Hang Xu
Xianyanling Yi
Tianyi Zhang
Qiang Wei
Hong Li
Jianzhong Ai
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2021
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-021-01452-1

Weitere Artikel der Ausgabe 1/2021

Molecular Cancer 1/2021 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.