You are on page 1of 9

Dendritic Cells as Adjuvant Therapy in Increasing Survival Rate

for Glioblastoma Patients: Meta-Analysis

ABSTRACT

Introduction: Glioblastoma Multiforme (GBM) is a primary neoplasm of the central nervous system with a low
survival rate, requiring more effective treatment to improve long-term survival. Dendritic Cell (DC) therapy is
expected to reduce tumor progressivity. Objective: This study aimed to analyze the administration of DC on the
overall survival (OS) value of GBM patients. Methods: A systematic literature search using the PRISMA
method through the Embase database, PubMed, and the Cochrane Controlled Trial Register for relevant studies
between the administration of DC in GBM patients with conventional therapy on the value of OS compared to
controls. Article quality was assessed using the Newcastle Ottawa Scale and statistically analyzed using RevMan
5.4. Results: From 14 articles, there was an increase in the value of OS from year to year for DC vaccine until
year 3 [1st-y HR: 0.74(0.57-0.95), I2: 15%, p=0.02; 2nd-y HR: 0.64(0.51-0.81), I2: 14%, p=0.0002; 3rd-y HR:
0.62(0.48-0.81), I2: 0%, p=0.0004]. However, for a period of 5 years, there was no difference [HR 0.81(0.62-
1.06), I2: 0%, p=0.13]. . Conclusion: DC vaccine improves survival in the early years of therapy but has not been
proven for long-term therapy DCs that act as antigen-presenting cells are tasked with inducing a specific
cytotoxic T cell response to eliminate tumor cells and B cells associated with long-term anti-tumor protection.
However, several factors need to be considered such as age, severity, volume of tumor removal.

Keywords : dendritic cell therapy, glioblastoma, overall survival, survival rate, vaccine

Article Highlight:
1. Dendritic cells (DC) are one type of immune therapy that is being explored to improve treatment
effectiveness in glioblastoma multiforme (GBM)
2. DC was predicted to improve survival rates in GBM patients within 3 years
3. Effect of DC in fifth year need to be explored to prove its effectiveness in increasing GBM survival rate

INTRODUCTION So new treatment strategies are needed, one of


Glioblastoma multiforme (GBM) is the most which is immunotherapy, which is currently being
malignant primary brain tumor of the central studied. An immunotherapy strategy is now in
nervous system (CNS) and one of the deadliest adult clinical trials, namely a dendritic cell (DC)-based
cancers worldwide.1 GBM accounted for 45.6% of vaccine.3 DCs act as regulators of the initial immune
all primary malignant brain tumors, with a mean response by releasing cytokines that activate
incidence of 3.1 cases per 100,000 where the cytotoxic lymphocytes and NK cells and then
highest peak incidence occurred at >75 years of age represent antigens to a subset of B and T
reaching 15.03/100,000.2 lymphocytes.6 Thus, DCs can modulate the patient's
Currently, the standard first-line treatment for immune system against tumors, thereby inducing
GBM is maximum surgical resection followed by immunological memory related to long-term anti-
chemotherapy with temozolomide (TMZ) and tumor protection,7 so this therapy is expected to
radiotherapy.3 The median OS of glioblastoma provide the prospect of high OS values in GBM
patients after complete resection was reported to patients. Several studies of DC administration into
have increased to 15.5 months compared with 11.7 the scope of GBM therapy in the last decade have
months for those who underwent subtotal resection been carried out quite a lot. However, the research
and 5.9 months for those without resection. 4 Despite that has been done has varying parameters and
using multimodal treatment, the prognosis of newly results. Thus, a study is needed to analyze the
diagnosed GBM patients remains low with a mean overall effect of giving DC on the survival rate of
OS time of around 15-17 months and a 5-year GBM patients.
survival rate of even <2%. 1,4 This is because tumor
cells, especially brain tumors, can evade immune
cells through different mechanisms, such as OBJECTIVE
antigenic modulation, decreased immunogenicity, This meta-analysis study aims to see whether
and immune suppression.5 this DC has a positive effect, especially on the
survival rate of GBM patients.
METHODS the treatment, the control group was given
a. Selection and Screening conventional therapy through operative methods
The authors searched the experimental and/or chemotherapy. All of these characteristics
literature systematically using the PRISMA method are reported in detail in Table 1.
in PubMed, EMBASE, and Cochrane Controlled Then we tested the quality of the articles using
Trial Register with categories of Dendritic cell the Newcastle-Ottawa scale with a result range of 7-
therapy, Glioblastoma, overall survival, and survival 9, which indicates the article has high publication
rate from the beginning of the database until July quality. These results are shown in Table 2.
2022. Downloaded articles were identified by Of all 14 data studies describing patients' OS
duplicate publications and then reviewed. scores, we analyzed survival rates in the first,
second, third, and fifth years. In the first year, we
b. Eligibility Criteria included data from 12 articles on the number of
The inclusion criteria for this study were as living GBM patients, 173/260 in the DC group and
follows: (a) published articles describing 234/414 in the control group. These results showed
quantifiable survival rates in patients with GBM that DC administration significantly increased the
with DC administration; (b) the DC intervention survival rate of GBM patients with [HR 0.74(0.57-
performed will be compared with the 0.95), I2: 15%, p=0.02] (Figure 1). In the second
control/placebo group, namely the group of GBM year, there were 12 studies stating the number of
patients who were given conventional therapy, living patients with DC administration was 85/190,
either: surgery and/or chemotherapy and/or while conventional therapy was 62/389. The
radiotherapy; (c) clinical trials; (d) the article has a comparison value increased significantly [HR
complete publication format; and (e) literature 0.64(0.51-0.81), I2: 14%, p=0.0002] (Figure 2).
published in English. However, we excluded when Even in the third year of evaluation of the number
the study experienced any of the following of surviving patients of 8 articles, DC showed an
conditions: (a) data could not be assessed; (b) increased protective ability with HR 0.62(0.48-
duplication; (c) not the original experimental study 0.81), I2: 0%, p=0.0004. However, in the fifth year,
administration of DC compared to conventional did
c. Data Quality Analysis not show a significant difference [HR 0.81(0.62-
The study quality of included articles was 1.06), I2: 0%, p=0.13]. These results are shown in
assessed using the Newcastle-Ottawa Scale (NOS) the forest plot shown in Figures 3-4.
for the cohort study. An article is of high quality if it
gets 3 or 4 points on choice ratings, 1 or 2 points for
comparability, and 2 or 3 points for results.8 DISCUSSION
Pathologically, in the case of brain tumors,
d. Data analysis there is an increase in the reactivity of CD8+ T cells
Data were analyzed using Review Manager 5.4 systemically to eliminate malignant cells. 9,10 Most
software. Calculate the Hazard Ratio (HR) with the clinicians use standard therapy modalities carried
inverse variance method for the random effect out continuously and intersecting, such as subtotal
model. We use generic inverse variance data with a and total tumor resection, radiotherapy, and
95% confidence interval (CI). The heterogeneity of chemotherapy.11 Currently, immune therapy is being
clinical trial data will be tested using I2. If p<0.05, developed to increase the effectiveness of treatment,
then the results are considered significant. one of which is dendritic cells (DC).
Therapeutic collaboration between
radiotherapy, chemotherapy, and DC has a positive
RESULTS interaction, where radiotherapy and chemotherapy
From a systematic search through PubMed, are known to decrease CD4+ activity globally,
EMBASE, and the Cochrane Controlled Trial which affects increasing other immune responses. 12
Register, 256 articles were obtained, which were Local radiotherapy also reduces suppressor T cells
then entered into the duplication screening process, that enhance effective T cell stimulation. DC
reading titles and abstracts, and the suitability of OS becomes the most potent antigen-presenting cell
values as an outcome for giving DC to GBM (APC) in the immune system to activate the immune
patients, and completeness of data so that 14 studies response against tumors.7,13 Where DCs are bound to
were included in this meta-analysis study. The flow pathogenic molecules such as lipopolysaccharide
is depicted in Figure 1. (LPS), they then become active as strong immune
All the studies that have been selected have stimulators by releasing interleukin (IL)-12, which
several assessments and parameters that vary; with a triggers the response of type 1 T-helper (Th1)
total sample of 826, the mean age is between 14-70 lymphocytes and tumor antigen cytotoxic T cells to
years. The types of DCV used vary with the method be eliminated.9,10,14 The mechanism of this protective
of administration via intracutaneous, subcutaneous, effect leads to an increase in the sensitivity of
or intratumor. The time of administration and dose chemotherapy in the remaining tumor cells to
of each study has a different benchmark. Then for cytotoxic activity.15 In a study by Jie et al., 2011,
there was a significant increase in CD3+, In addition, the initiation of therapy is expected
CD3+/CD4+, CD4+/CD8+ ratio, NK cell to be as early as possible.11 In most relapse cases,
percentage in the DC group, and serum levels of IL- DC will be given earlier because no re-operation or
2, IL-12, and IFN- indicating that DCs can induce radiotherapy is performed.13 Newly diagnosed GBM
Th1 immune responses as antitumor. 16,17 Plus, the patients often experience recurrence at the start of
ability to induce B and T cells theoretically can immunotherapy due to the formation of clones of
increase long-term antitumor protection.7 mutant GBM cells by radiotherapy induction with
In the results of our meta-analysis, the survival changes in the antigenicity of the original GBM
rates of GBM patients who received DC were cells used for vaccine preparation. 10 It is necessary
significantly better than controls in the first year, to conduct a study to see the feasibility of starting
increased considerably in the third year, and rose to DC therapy as soon as possible after surgery and
a peak in the third year. This indicates a delaying without waiting for radiotherapy.23
effect on the effectiveness of DC therapy. A meta- Another factor is the patient's age, where
analysis that examined the short-term impact of DC patients with ages <50 years have a higher survival
on the survival rate of GBM patients showed a non- rate.24 Then the volume of the resected tumor. 13
significant result at six months. 6 This delaying Surgical tumor removal is carried out as safely as
effect of therapeutic activity was due to inhibition of possible with minimal residual.25 It has been proven
DC transfer to lymph nodes, thereby reducing that postoperative chemotherapy has a large positive
efficacy.18 On the other hand, the generation of effect on GBM patient.24 However, Ardon et al.,
immune response after DC administration may 2012 stated that there was no significant difference
undergo sensitization wherein the proliferative between total and subtotal resection. This is because
intensification of mononuclear cells and a some samples in subtotal resection have lower RPA
significant increase in the leukocyte migration index values and have the promoter of the O6-
after the third to sixth injection. 1 In addition, several methylguanine-DNA methyltransferase (MGMT)
factors influence disease progression and pre- gene in tumor cells which are generally more
vaccination immune status.19 A study examining sensitive to chemotherapy reagents.15 Patients
pre-vaccination immune status with Th1 indicators, whose methylated promoter of the MGMT gene, it
IFNγ, CD8+ cells, and monocytes showed that the improves the prognosis and enhances the survival
category of patients with high immunity showed rate.24 However, a study stated that both groups with
higher OS values.3,20 The lower the tumor grade, the methylated and unmethylated MGMT showed an
better the prognosis.16 increase in OS values.26
In the fifth year, the DC administration showed Then the different methods of producing DC
no significant results compared to the control. This therapy (such as induction methods with LPS, IFN,
is thought to be due to limited studies assessing etc.) result in the procedure having different results. 9
long-term survival effects. In addition, antigenic This has made it difficult for several studies to
proteins introduced, processed, and presented by apply the standardization in antitumor immune
DCs are likely to be mutated so that they are monitoring to date, which is necessary to prevent
recognized as non-malignant.9 GBM tumor cells bias in various immune therapies, especially when
have a strong ability to inhibit the immune system using whole tumor cell lysates as antigens. 15 In
so that it affects the proliferation and immune 2012, Cho et al. reported using DC-induced intact
function, and necrosis around the lesion reduces the tumor cells that died with gamma radiotherapy to
ability to circulate T cells to reach the site. 21 In provide a more heterogeneous protein antigen
addition, the hematotoxic effect of TMZ matrix to minimize the potential for tumor immune
chemotherapy inhibits the multiplication of effector escape than lysates, peptides, DNA, and mRNA. 13
T cells that function to reduce tumor immune Moreover, tumor lysates have advantages:
tolerance.9 producing a more specific antigen immune
Several factors that need to be considered in response, minimal HLA restriction, and are easy to
the success of DC as adjuvant therapy include dose personalize for each patient. 20,27 A study showed,
and administration interval.13 However, a study by induction with viral antigens that have a DNA
Chang et al., 2011 using relatively higher DC doses matrix with GBM, such as cytomegalovirus also
with short administration intervals gave a non- showed increased survival rates.28,29
significant survival rate in the first year. 10 In Several studies have not been able to explain
subsequent years consistently provided good the immunological pathways involved, so future
protective results. Dendritic cell vaccination of research is expected to explore more enhancement
patients with glioma seems to be safe and not of anti-tumor immune reactions, for example, by
associated with autoimmunity.22 However, it is combining DC vaccination with other
necessary to pay attention to doses that can give immunotherapy (checkpoint inhibitors, anti-PDI).28
dangerous side effects, such as liver failure or This treatment cannot replace chemotherapy but
delayed hypersensitivity, fever, and gastrointestinal contributes a unique benefit to patients that is not
complaints.10 proven either by vaccination in the absence of
subsequent chemotherapy or chemotherapy alone.
However, with increased survival in the population, 4. Luo C, Song K, Wu S, Hameed NU, Kudulaiti N, Xu H, et
this therapy can be combined with other therapies. 30 al. The prognosis of glioblastoma: A large, multifactorial
study. British Journal of Neurosurgery. 2021;35(5):555–61. 
In addition, the relatively small number of GBM 5. Inogés S, Tejada S, de Cerio AL-D, Gállego Pérez-Larraya
patients with a relatively poor prognosis makes the J, Espinós J, Idoate MA, et al. A phase II trial of autologous
clinical trials into studies that have not represented dendritic cell vaccination and radiochemotherapy following
mass efficacy. fluorescence-guided surgery in newly diagnosed
glioblastoma patients. Journal of Translational Medicine.
2017;15(1). 
6. Cozzi S, Najafi M, Gomar M, Ciammella P, Iotti C,
CONCLUSION Iaccarino C, et al. Delayed effect of dendritic cells
Administration of the DC vaccine has been vaccination on survival in glioblastoma: A systematic
shown to improve survival in the early years of review and meta-analysis. Current Oncology.
2022;29(2):881–91. 
therapy but is not significant for long-term 7. Antonopoulos Markos, Van Gool Stefaanw, Dionysiou
treatment. Thus, further research is required Dimitra, Graf Norbert, Stamatakos Georgios. Immune
regarding the standardization of DC administration phenotype correlates with survival in patients with GBM
in GBM patients with a larger sample size to assess treated with standard temozolomide-based therapy and
immunotherapy. Anticancer Research. 2019;39(4):2043–
the effectiveness of therapy. However, several
51. 
factors need to be considered, such as age, severity, 8. Sharmin S, Kypri K, Khanam M, Wadolowski M, Bruno R,
the volume of tumor resection, dose, and timing of Mattick R. Parental Supply of Alcohol in Childhood and
administration. Risky Drinking in Adolescence: Systematic Review and
Meta-Analysis. International Journal of Environmental
Research and Public Health. 2017;14(3):287.
9. Buchroithner J, Erhart F, Pichler J, Widhalm G, Preusser M,
ACKNOWLEDGMENT Stockhammer G, et al. Audencel immunotherapy based on
Thank you to all committees and assessors of dendritic cells has no effect on overall and progression-free
the 21st Continuing Neurological Education (CNE) survival in newly diagnosed glioblastoma: A phase II
randomized trial. Cancers. 2018;10(10):372. 
scientific meeting, Surabaya, Indonesia.
10. Chang C-N, Huang Y-C, Yang D-M, Kikuta K, Wei K-J,
Kubota T, et al. A phase I/II clinical trial investigating the
adverse and therapeutic effects of a postoperative
CONFLICT OF INTEREST autologous dendritic cell tumor vaccine in patients with
There is no conflict of interest regarding the malignant glioma. Journal of Clinical Neuroscience.
2011;18(8):1048–54. 
publication of this article. 11. Mitsuya Koichi, Akiyama Yasuto, Iizuka Akira, Miyata
Haruo, Deguchi Shoichi, Hayashi Nakamasa, Et Al. Alpha-
Type-1 Polarized Dendritic Cell-Based Vaccination In
FUNDING Newly diagnosed high-grade glioma: A phase II clinical
trial. Anticancer Research. 2020;40(11):6473–84. 
This research did not receive a specific grant
12. Fadul CE, Fisher JL, Hampton TH, Lallana EC, Li Z, Gui J,
from any funding agency in the public, commercial, et al. Immune response in patients with newly diagnosed
or not-for-profit sector. glioblastoma multiforme treated with intranodal autologous
tumor lysate-dendritic cell vaccination after radiation
chemotherapy. Journal of Immunotherapy. 2011;34(4):382–
9. 
AUTHOR CONTRIBUTION 13. Cho D-Y, Yang W-K, Lee H-C, Hsu D-M, Lin H-L, Lin S-
AR contributes to conceptualization, drafting, Z, et al. Adjuvant immunotherapy with whole-cell lysate
data extraction, and editing, administration. EM dendritic cells vaccine for glioblastoma multiforme: A
performs all data processing, editing and review, phase II clinical trial. World Neurosurgery. 2012;77(5-
6):736–44. 
monitoring. All authors read and approved the final
14. Buchroithner J, Pichler J, Marosi C, Widhalm G, Seiz-
draft. Rosenhagen M, Novosielski M, et al. Vascular Endothelia
growth factor targeted therapy may improve the effect of
dendritic cell-based cancer immune therapy. Int Journal of
REFERENCES Clinical Pharmacology and Therapeutics. 2014;52(01):76–
7. 
15. Ardon H, Van Gool SW, Verschuere T, Maes W, Fieuws S,
1. Liau LM, Prins RM, Kiertscher SM, Odesa SK, Kremen TJ, Sciot R, et al. Integration of autologous dendritic cell-based
Giovannone AJ, et al. Dendritic cell vaccination in immunotherapy in the standard of care treatment for
glioblastoma patients induces systemic and intracranial T- patients with newly diagnosed glioblastoma: Results of the
cell responses modulated by the local central nervous HGG-2006 phase I/II trial. Cancer Immunology,
system tumor microenvironment. Clinical Cancer Research. Immunotherapy. 2012;61(11):2033–44. 
2005;11(15):5515–25. 16. Jie X, Hua L, Jiang W, Feng F, Feng G, Hua Z. Clinical
2. Wirsching H-G, Galanis E, Weller M. Glioblastoma. application of a dendritic cell vaccine raised against heat-
Handbook of Clinical Neurology. 2016;:381–97.  shocked glioblastoma. Cell Biochemistry and Biophysics.
2011;62(1):91–9. 
3. Erhart F, Buchroithner J, Reitermaier R, Fischhuber K, 17. Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-
Klingenbrunner S, Sloma I, et al. Immunological analysis of Tønnesen P, Suso EM, et al. Therapeutic vaccination
phase II glioblastoma dendritic cell vaccine (Audencel) against autologous cancer stem cells with mrna-transfected
trial: Immune system characteristics influence outcome and dendritic cells in patients with glioblastoma. Cancer
Audencel up-regulates th1-related immunovariables. Acta Immunology, Immunotherapy. 2013;62(9):1499–509.
Neuropathologica Communications. 2018;6(1). 
18. Rangel-Reyes, J.C.; Chimal-Eguia, J.C.; Castillo-Montiel, newly diagnosed glioblastoma. Journal of Translational
E. Dendritic Immunotherapy Improvement for an Optimal Medicine. 2018;16(1). 
Control Murine Model. Comput. Math. Methods Med. 2017, 25. Leplina OY, Stupak VV, Kozlov YP, Pendyurin IV,
2017, 1–9 Nikonov SD, Tikhonova MA, et al. Use of interferon-α-
19. Yao Y, Luo F, Tang C, Chen D, Qin Z, Hua W, et al. induced dendritic cells in the therapy of patients with
Molecular subgroups and B7-H4 expression levels predict malignant brain gliomas. Bulletin of Experimental Biology
responses to dendritic cell vaccines in glioblastoma: An and Medicine. 2007;143(4):528–34. 
exploratory randomized phase II clinical trial. Cancer
Immunology, Immunotherapy. 2018;67(11):1777–88.  26. Wen PY, Reardon DA, Armstrong TS, Phuphanich S,
Aiken RD, Landolfi JC, et al. A randomized double-blind
20. Yu JS, Liu G, Ying H, Yong WH, Black KL, Wheeler CJ. placebo-controlled phase II trial of dendritic cell vaccine
Vaccination with tumor lysate-pulsed dendritic cells elicits ICT-107 in newly diagnosed patients with glioblastoma.
antigen-specific, cytotoxic T-cells in patients with Clinical Cancer
malignant glioma. Cancer Research. 2004;64(14):4973–9.  27. Akiyama Y, Oshita C, Kume A, Iizuka A, Miyata H,
21. Jan C-I, Tsai W-C, Harn H-J, Shyu W-C, Liu M-C, Lu H- Komiyama M, et al. Α-type-1 polarized dendritic cell-based
M, et al. Predictors of response to autologous dendritic cell vaccination in recurrent high-grade glioma: A phase I
therapy in glioblastoma multiforme. Frontiers in clinical trial. BMC Cancer. 2012;12(1).
Immunology. 2018;9.  28. Batich KA, Reap EA, Archer GE, Sanchez-Perez L, Nair
22. Yamanaka R, Homma J, Yajima N, Sano M, Takahashi M. SK, Schmittling RJ, et al. Long-term survival in
277. clinical evaluation of dendritic cell vaccination for glioblastoma with cytomegalovirus PP65-targeted
patients with recurrent glioma: Results of a clinical phase vaccination. Clinical Cancer Research. 2017;23(8):1898–
I/II trial. Molecular Therapy. 2006;13.  909. 
23. De Vleeschouwer S, Fieuws S, Rutkowski S, Van 29. Batich KA, Mitchell DA, Healy P, Herndon JE, Sampson
Calenbergh F, Van Loon J, Goffin J, et al. Postoperative JH. Once, twice, three times a finding: Reproducibility of
adjuvant dendritic cell–based immunotherapy in patients dendritic cell vaccine trials targeting cytomegalovirus in
with relapse glioblastoma multiforme. Clinical Cancer glioblastoma. Clinical Cancer Research. 2020;26(20):5297–
Research. 2008;14(10):3098–104.  303. 
30. Wheeler CJ, Das A, Liu G, Yu JS, Black KL. Clinical
24. Liau LM, Ashkan K, Tran DD, Campian JL, Trusheim JE, responsiveness of glioblastoma multiforme to
Cobbs CS, et al. First results on survival from a large phase chemotherapy after vaccination. Clinical Cancer Research.
3 clinical trial of an autologous dendritic cell vaccine in 2004;10(16):5316–26. 

ATTACHMENT

Figure 1. Flow Diagram of Study Selection with PRISMA Method


Abstract Identification (n :
256) By title, abstracts and duplicates
are eliminated (n : 203)

Articles Screened (n : 53)


There is no correlation between
OS values and DC
administration (n :22)
Assessed for eligibility (n : 31)
Lack of data (n : 17)
Articles included in this
study (n : 14)
Table 1. Study Characteristics
Author, year Sample Age Outcome DCV Type Method Time Dose
Batich 2017 34 55 PFS, OS, IFN-𝜸, pp65 lysosome- IC During Early: 2x107.
MRI Changes, Treg associated CX-RT follow up:
respond, CD8 membrane 107/month until
CD8+ glycoprotein 10x
mRNA-pulsed DCs
Batich 2020 34 56-57 OS, migration rate pp65 RNA-pulsed IC NA  NA
DCs
Buchroithner 2013 40 NA PFS, OS Tumor lysate- NA After 10x
charged autologous RT-CX administration
DCs
Buchroithner 2018 76 54 PFS, OS, AE, MRI Tumor lysate- NA After 1–5 × 106
changes charged autologous RT-CX
DCs
Chang 2010 76 44.7 AE; median Phagocytic DCs SC After 1–6 x 107
survival, survival surgical
rate, CD8(+) RT
Cho 2012 52 14-70 OS, PFS; 1-, 2-, and Whole-tumor lysate SC During 2–5 × 107
3-year survival pulsed DCs CX-RT
rates, QoL
Jan 2018 47 51.8 CCRT, PD-1+, OS, autologous NA Before 2–5 × 107 total
PFS, CD45+, dendritic cell/tumor CX-RT 14-16x
CD4+, CD8+, PD- antigen vaccine
L1
Jie 2012 25 43.1 cd3+; cd3+ cd4+; Autologous SC Before 106 total 6x
CD3+ CD8+; CD4+ glioblastoma-DCs CX-RT
CD8+, NK,
IL2,IL12, IFN, CR,
NC, PD, PR partial
response
Leplina 2006 119 42.6 IFN-𝜸, TNF-α, IL- Interferon-induced SC After 10x106 total 6x
13, IL-10, Antigen- DCs surgery
Specific Response,
Patient Survival
Vik-mo 2013 84 NA TSL, hTERT, size autolog DC based IC After 107 total 9-18x
tumor, AE, OS, PFS cancer stem cell- surgery-
mRNA RT until
CX
Wheeler 2004 36 55 CTL, volume tumor autologous tumor SC After 10 – 40x 106
by MRI, OS freeze-thaw lysate surgery
Wen 2019 124 59.2 PFS, OS, AE,HLA- DCs pulsed with six IC During 1.1 × 107
A2 antigens, HLA- synthetic peptide CX-RT 1x/month during
A1 antigens, epitopes targeting CX
immune response GBM tumor
Yamanka 2005 45 48.9 hypersensitivity, Peripheral blood IC / Before 1x107
OS, IFN-g DCs pulsed with intratumor CX-RT
ELISPOT assay, autologous tumor
radiological lysate
findings
Yu 2004 34 44.7 OS, AE, IFN-𝜸, autologous DCs IC Before 2 x 105.
pulsed with tumor
HER-2 CTLs, lysate surgery
gp100-specific
CTLs, MAGE-1-
specific CTLs
Notes : PFS= Progression Free Survival; IFN= Interferon; CX= Chemotherapy; RT= Radiotherapy; RNA= Ribonucleic Acid;
SC= Subcutan; IC= Intracutan; AE= Adverse Effect; NA= Not Available; QoL= Quality of Life; CR= Complete Response;
DC= Dendritic Cells; NC= No Change; PD= Progressive Disease; PR= Partial Response; IL= Interleukin; TSL= Tumorsphere
Lysate; GBM= Glioblastoma Multiforme; CTL= Cytotoxic T Lymphocyte

Table 2. Data Quality Assessment

Article Selection Comparability Outcome Total Score

Batich 2017 *** ** *** 8

Batich 2020 *** ** *** 8

Buchroithner 2013 *** ** ** 7

Buchroithner 2018 *** ** *** 8

Chang 2011 **** ** *** 9

Cho 2012 **** ** *** 9

Jan 2018 **** ** *** 9

Jie 2012 **** ** *** 9

Leplina 2006 **** ** *** 9

Muller 2015 **** ** *** 9

Vik-Mo 2013 **** ** *** 9

Wen 2019 **** ** *** 9

Wheeler 2004 **** ** *** 9

Yamanka 2005 **** ** *** 9

Yu 2004 **** ** *** 9


Figure 2. Forest Plot of Survival Rate in the First Year

Figure 3. Forest Plot of Survival Rate in the Second Year

Figure 4. Forest Plot of Survival Rate in the Third Year


Figure 5. Forest Plot of Survival Rate in the Fifth Year

You might also like