Next Article in Journal
The Role of Native T1 and T2 Mapping Times in Identifying PD-L1 Expression and the Histological Subtype of NSCLCs
Previous Article in Journal
Genomic Mapping of Epidermal Growth Factor Receptor and Mesenchymal–Epithelial Transition-Up-Regulated Tumors Identifies Novel Therapeutic Opportunities
Previous Article in Special Issue
Diagnosis and Treatment of Peripheral and Cranial Nerve Tumors with Expert Recommendations: An EUropean Network for RAre CANcers (EURACAN) Initiative
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

DCVax-L Vaccination in Patients with Glioblastoma: Real Promise or Negative Trial? The Debate Is Open

1
Department of Oncology, Azienda Unità Sanitaria Locale (AUSL) Bologna, 40139 Bologna, Italy
2
Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(12), 3251; https://doi.org/10.3390/cancers15123251
Submission received: 24 May 2023 / Revised: 9 June 2023 / Accepted: 16 June 2023 / Published: 20 June 2023
(This article belongs to the Special Issue Rare Primary Brain Tumors in Adults)

Abstract

:

Simple Summary

Glioblastoma is the most common of primary brain tumors, accounting for approximately 50% of intracranial malignancies. It is an aggressive neoplasm with a poor prognosis. To date, the standard of care is a treatment involving maximal surgery, radiotherapy concurrent with and followed by maintenance chemotherapy with temozolomide. Despite this multimodal approach and the continuous advances in molecular biology, median survival is 13–14 months and 5-year survival does not exceed 10% of patients. Therefore, the need to develop new treatments that can impact the survival of glioblastoma patients is urgent. After decades of research failures, immunotherapy timidly begins to give the first results in the treatment of this tumor. The publication of the phase III study on the use of the dendritic cell vaccine DCVax-L in glioblastoma has aroused much interest in neuro-oncology. We report the promising results of this trial, which, however, is worthy of a critical debate regarding both the special study design and the authors’ conclusions.

Abstract

The lack of significant improvement in the prognosis of patients with GB over the last decades highlights the need for innovative treatments aimed at fighting this malignancy and increasing survival outcomes. The results of the phase III clinical trial of DCVax-L (autologous tumor lysate-loaded dendritic cell vaccination), which has been shown to increase both median survival and long-term survival in newly diagnosed and relapsed glioblastoma, have been enthusiastically received by the scientific community. However, this study deserves some reflections regarding methodological issues related to the primary endpoint change, the long accrual period, and the suboptimal validity of the external control population used as the comparison arm.

1. Introduction

Glioblastoma (GB) is the most aggressive CNS tumor, with an incidence rate of about 8 per 100,000 people and an average survival of approximately 13–14 months [1]. The 5-year survival rate is less than 5% [2]. GB recurrence is near-universal despite surgical removal and multimodal treatments [3]. Although many trials have been concluded in the last decades, almost all therapeutic agents have proved ineffective in increasing survival and maximal surgical resection, followed by concomitant radio-chemotherapy and adjuvant temozolomide, remains the established therapeutic standard of care [4]. With the introduction of loco-regional tumor-treating fields (TTFields) therapy, an innovative strategy consisting of low-intensity alternating electric fields approved for the treatment of recurrent and newly diagnosed GB, the hope to increase PFS and OS while improving quality of life, fired up [5,6].
Immunotherapy has revolutionized the outcome of various solid tumors [7,8,9,10,11,12], but it has proven ineffective in the treatment of GB [13,14,15]. GB is an extremely heterogeneous cancer and poses several difficult challenges to the success of immunotherapy. First of all, it is an immunologically “cold” tumor, characterized by a tumor microenvironment (TME) enriched with immunosuppressive cytokines including transforming growth factor-beta (TGF-B), interleukine-6 (IL-6), IL-10, and immune regulatory cells (T-regulatory lymphocytes, ‘protumoral’ M2 macrophages, myeloid-derived suppressor cells, and tumor-associated macrophages) that turn off inflammation and disable an effective immune response by T CD8+ lymphocytes and natural killer (NK) cells [16,17,18,19,20,21]. Another crucial issue is the presence of both intra-tumor and inter-tumor heterogeneity in GB cells, resulting in the production of “poor quality” neoantigens, named “sub-clonal” neoantigens (present only in a subset of tumor cells and not in all tumor cells), that are much less immunogenic than clonal neoantigens and inadequate to elicit an effective immune response [22]. The secretion of immunosuppressive factors by glioma cells and the chronic exposure to “sub-threshold” antigenic stimulation lead T lymphocytes to a state of metabolic “exhaustion”, which renders them inactive [23,24]. Furthermore, GB cells overexpress the checkpoint protein PD-L1, which binds its ligand PD-1 in microglia, resulting in down-regulation of T-lymphocyte proliferation and cytotoxic activity [25]. The big bet of immunotherapy in neuro-oncology is controlling the enormous heterogeneity of GB and finding a way to modify the TME, “manipulating” it to obtain an effective antigen-specific immune response without triggering a severe intracranial local inflammatory reaction.
Dendritic cell vaccination is a type of immunotherapy consisting of autologous innate immune cells, dendritic cells, pulsed with autologous tumor lysate that has emerged as a promising novel treatment because it is the paradigm of tailored therapy, producing active immune products manufactured on the specific tumor antigens of each patient [26,27]. Promising results have been reported for sipuleucel-T [28], approved by the FDA in 2010 for metastatic castration-resistant prostate cancer (not in Europe due to the modest advantage offered compared to other therapies), and for other dendritic vaccines applied to various types of solid and haematologic malignancies [29,30,31,32]. To date, many dendritic cell vaccinations are in the experimental stage for melanoma (NCT00390338), breast cancer (NCT04348747), hepatocellular cancer (NCT04912765), and colorectal cancer (NCT03827967). Active immunotherapy with dendritic cell vaccination in GB has been observed since 1999, demonstrating reduced tumor growth, prolonged survival, and antigen-specific cytotoxic T-CD8+ lymphocyte responses in murine models and early-stage clinical trials [33,34,35,36,37].
Liau et al. [38,39] reported the results of a phase III clinical trial (NCT00045968) testing DCVax-L vaccine on patients with either newly diagnosed or recurrent GB to assess whether autologous tumor lysate-loaded dendritic cell vaccine administered in addition to standard of care (SOC) may improve survival outcomes.
DCVax-L is a highly personalized vaccination that uses tumor lysate as a source of antigens and uses the patient’s autologous dendritic cells harvested by leukapheresis and then expanded in vitro.

2. Study Design

The study was a prospective multicentric randomized double-blind phase III trial, involving 331 patients (232 patients in the DCVax-L arm and 99 patients randomized to the placebo group) among 94 centers in 4 countries (US, Canada, UK, and Germany) [39]. The trial began in 2007, was suspended from 2008 to 2011 for economic reasons, and ended in 2015. Approximately 90% of patients were randomized from 2012 to 2015.
Patients with newly diagnosed GB were to be randomized 2:1 to standard radio-chemotherapy with either placebo or DCVax-L. Progression-free survival (PFS) was selected as the primary endpoint. The study design included the cross-over, so all patients could receive DCVax-L following tumor recurrence. With cross-over at progression, overall, 90% of patients from the two arms, experimental and control, received DCVax-L.

3. Results

A first report of the interim data of the ITT (intention to treat) population was published in 2018; investigators did not report the results for PFS, the primary endpoint, explaining that PFS could not be assessed for that publication and that it would be analyzed later to allow for central, multi-factorial assessment by an expert panel [39]. The authors only reported survival data, underscoring the high percentage of long-term survivors. In particular, the median OS (mOS) in the ITT population was 23.1 months (95% CI 21.2–25.4). mOS in the subgroup of patients with methylated O-6-methylguanine-DNA methyltransferase (MGMT) was 34.7 months (95% CI 27.0–40.7). The three-year survival percentage was 25.4%. In addition, a subpopulation of extended survivors (n = 100) with mOS of 40.5 months, only partially justified by favorable prognostic factors, was reported (in this subgroup, about 30% of patients were <50 years old, 70% underwent complete surgical resection, and 65% had MGMT-methylated tumors) [39].
Although it was a double-arm phase III trial, a single survival curve was presented, an anomalous fact for a randomized study.
In 2023, the final results of the study will be published [38]. Investigators stated that PFS was not an appropriate endpoint for two reasons: first, the placebo group was excessively depleted by the cross-over (overall, 90% of patients from the two arms received DCVax-L); second, the interpretation of PFS data was difficult due to the phenomenon of pseudoprogression. Therefore, the study design was changed and adapted at later stages of the research project, and the primary endpoint of the study was changed from PFS to OS.
An external control group was created as a comparator group using several selected randomized clinical trials, both in the adjuvant and recurrence settings.
A systematic literature review was conducted to identify relevant studies in newly diagnosed GB and recurrent GB, respectively, to provide comparator control populations. Five phase III studies [6,40,41,42,43], each with a control arm treated with the standard radiotherapy and temozolomide regimen, were selected as the comparator control group for the newly diagnosed setting (for a total of 1366 patients). As a consequence of the cross-over design, approximately 90% of patients were treated with DCVax-L thus, the two arms (DCVax-L and placebo) from the original study were merged together in the OS analyses.
A separate survival analysis of patients receiving DCVax-L at progression (recurrent GB) was performed; for this purpose, ten phase III comparator studies (for a total of 640 patients treated with SOC, such as lomustine or bevacizumab) [44,45,46,47,48,49,50,51,52,53] were selected as the control group. Practically, they created a new study population (recurrent GB), on which they conducted analyses that were not initially planned.
Thus, they finally compared OS in patients with newly diagnosed GB and recurrent GB treated with DCVax-L plus SOC vs. external control patients treated with SOC. In order to minimize biases due to confounding factors in this adapted study design and imbalances in patients’ characteristics, the authors performed a matching-adjusted indirect comparison.
Therefore, this study is configured, after modification of the study design and of the primary endpoint, as a non-randomized single-arm trial with an external control group.
In this second report recently published, finally, PFS data were reported and were not encouraging since DCVax-L + SOC performed worse than SOC alone. The median PFS was 6.2 months for the DCVax-L arm and 7.6 months for the placebo group (p = 0.47). Nevertheless, significant OS improvement was registered in both newly diagnosed and relapsed GB patients treated with DCVax-L + SOC compared with the external control group that received SOC alone.
Patients with newly diagnosed GB treated with DCVax-L + SOC survived 19.3 months (95% CI, 17.5–21.3) compared to 16.5 months (95% CI, 16.0–17.5) for the control group (p = 0.002). Patients with recurrent GB treated with the vaccine survived 13.2 (95% CI, 9.7–16.8) months versus 7.8 (95% CI, 7.2–8.2) months for the control group (p < 0.001).
OS was improved in patients with newly diagnosed MGMT-methylated GBs receiving DCVax-L (30.2 months) compared with external control patients (21.3 months) (HR, 0.74; 98% CI, 0.55–1.00; p = 0.03).
Survival at 48 months was 15.7% in the experimental group vs. 9.9% in the control group, with the longest survivor still alive 8 years after randomization. Survival at 60 months was 13% in the investigational arm and 5.7% in the external control group.
The survival advantage of DCVax-L was better in poor prognosis subpopulations, including older patients, patients with suboptimal surgical resection, and patients with relapsed disease.
The investigational treatment was well tolerated, and most patients did not experience serious side effects from the immunotherapy vaccine. Only 5 serious adverse events possibly related to the vaccine were reported: 3 cases of grade 2/3 intracranial edema, 1 case of grade 3 nausea, and 1 case of grade 3 infection [38].

4. Discussion

At first glance, the results of this study may seem surprising: in the last 18 years, it has been one of the first, if not the first, phase III studies to show significant increases in long-term survival for both newly diagnosed and relapsed GB, with an even greater benefit in the relapsed population. Specific poor prognosis subpopulations in this study showed unexpected benefits, including older patients and patients with significant residual disease where radical surgery was not possible.
However, some reflections regarding the methodology are required [54,55].
When considering PFS data, the study is negative, and the trial did not reach its prospectively defined primary endpoint. Therefore, from a purely formal point of view, the study should be declared negative.
The comparison of the investigational arm with an external control group should be considered a post-hoc retrospective analysis, suitable for generating hypotheses but not providing high-quality evidence. Non-randomized externally controlled studies are gradually becoming attractive because they are faster, cheaper, and limit the number of patients exposed to substandard or ineffective interventions, but they are inadequate for a phase III trial and require a pre-specified detailed protocol and robust statistical methods to minimize the risk of bias [56,57]. In this trial, the comparison of the active treatment arm with the external control population was not based on individual datasets from the selected randomized clinical trials; however, an indirect analysis was performed at the trial level with survival data reconstructed by an algorithm. The lack of individual patient data analysis represents a limitation: this trial does not provide a comparison on patient-level data, which compromises the quality of the evidence and the reliability of the results.
Furthermore, the artificial generation of the external control group resulted in impressive differences in the control population from the vaccine arm. This is a further major methodological limitation: the validity of external controls was compromised by the demographic characteristics of the comparison studies. The studies selected as an external control group had different patient characteristics, and this represents an important confounding factor.
Randomized controlled trials, even if difficult to conduct, are the gold standard for producing high-quality scientific evidence in phase III clinical trials and should always be pursued. The validity of external controls depends on the availability of high-quality patient-level data, methodological accuracy, and validation analyses to reduce the risk of distortions [58].
The DCVax-L trial included only patients who received gross or near total resection of the tumor mass, patients with disease confined to one hemisphere, and patients who had been off glucocorticoids for at least three weeks. All these criteria inevitably represent factors capable of favorably impacting survival; however, these inclusion criteria were not present in the studies used for comparison.
Moreover, patients with disease progression after completion of radiotherapy (which presumably have a poorer prognosis) were excluded from randomization in the DCVax-L trial, but this criterion was not included in all the comparison trials of the external control group; therefore, the vaccine trial selected patients with a more favorable prognosis, which could justify the long mOS observed. Similarly, other important patient characteristics, known as established prognostic factors, such as age, steroid use, performance status, and extent of resection, were not easily comparable between the two groups or were even missing in almost all selected trials. In several studies, the evaluation of MGMT methylation status was absent, and isocitrate dehydrogenase (IDH) mutational status has never been analyzed in any study.
The long randomization period implies that the criteria adopted for recruiting GB patients do not consider the 2016/2021 WHO classification. Consequently, the patients were not molecularly stratified, and this also creates a bias. In fact, it cannot be excluded that long-survivors might have had less aggressive tumors, for example, if IDH 1/2 mutated.
Certainly, we can agree with the investigators that PFS is not an adequate endpoint for immunotherapy studies because of the phenomenon of pseudoprogression, which is observed in approximately 40% of cases in vaccination trials [59]. Moreover, pseudoprogression is a frequent problem, especially in newly diagnosed MGMT-methylated GB; thus, PFS is a suboptimal endpoint for phase III trials, especially in this malignancy.
Despite the many doubts and perplexities that the methodology of this study raises, some cornerstones of the worth of its results remain unchanged: in a disease that is basically orphaned of treatments, such as GB, a therapy that might increase survival at the cost of low toxicities should not only arouse skepticism but also efforts and commitment to verify its efficacy with further appropriate clinical trials. Considering the extremely personalized nature of this treatment, when a GB patient treated with DCVax-L undergoes disease recurrence, a new, more specific vaccine batch can be prepared in order to restore disease control and effectively counteract any resistant clones.
The significant percentage of long-term survivors that is reported in this study could be consistent with an effect on immune memory by T lymphocytes; therefore, this new type of immunotherapy deserves further investigation.
Dendritic cell vaccines are well suited to be used in combination therapeutic regimens, for example, in association with immune check-point blockade, oncolytic viruses, CAR-T therapy, Optune, other vaccines, etc. This is essential because all the latest studies conducted in neuro-oncology show that the most effective strategies to fight GB seem to be combination treatments and not monotherapies.

5. Conclusions

This innovative vaccine is undoubtedly promising; therefore, the methodological issues of the DCVax-L trial deserve our attention and, possibly, another confirmation trial.
Several changes have been made over the years (replacement of a randomized design with a synthetic control arm, removal of PFS as the primary endpoint, addition of a new study population, conduct of unplanned analyses), raising many questions about data interpretability [60].
Although we understand that the development of personalized vaccines is a complex and expensive process, the limitations of this trial reduce the reliability of the results and prevent drawing firm conclusions about the efficacy of the dendritic cell vaccine.
To confirm the results of DCVax-L, it would be ideal to design two new trials, one in the newly diagnosed GB setting and another in the relapsed-disease setting, both randomized, for comparison with the standard of care. For example, it could be proposed to conduct a new randomized trial in the setting of recurrent GB, comparing the standard of care (lomustine) with the combination of DCVax-L plus an immune checkpoint inhibitor (PD-1/PD-L1 blocker or CTLA-4 inhibitor) as a strategy for boosting anti-tumor immune responses [28]. As expected in phase III trials, the primary endpoint of the study should be OS; secondary endpoints of the study should be PFS, quality of life, and patients’ reported outcomes.
We understand that randomized controlled trials strictly follow a pre-established protocol and risk being an overly rigid model because they fail to incorporate information that gradually becomes available. To overcome these rigidities, at least in part, adaptive trial designs can be explored, but randomized trials remain the gold standard for finding optimal answers to clinical questions. In considering the possibility of starting new trials; however, we should not overlook the great difficulties that dendritic cell vaccine entails: manufacturing is very expensive, requires adequate infrastructure, and involves an enormous expenditure of time, as demonstrated by the very long time required to conclude enrollment in the DCVax-L trial. Even in a hypothetical drug’s marketing phase, the problem of costs could be fundamental and demand a discussion between health authorities and pharmaceutical companies to ensure accessibility of the product to patients.

Author Contributions

L.G. and E.F.: Conceptualization, L.G., V.D.N., S.B., L.R. and E.F.: Investigation, L.G.: Writing and draft preparation, V.D.N., E.F., S.B., L.G., L.R. and A.T.: review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

The publication of this article was supported by the “Ricerca Corrente” funding from the Italian Ministry of Health.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is unavailable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ostrom, Q.T.; Gittleman, H.; Farah, P.; Ondracek, A.; Chen, Y.; Wolinsky, Y.; Stroup, N.E.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006–2010. Neuro Oncol. 2013, 15 (Suppl. S2), ii1–ii56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef]
  3. Park, J.K.; Hodges, T.; Arko, L.; Shen, M.; Dello Iacono, D.; McNabb, A.; Olsen Bailey, N.; Kreisl, T.N.; Iwamoto, F.M.; Sul, J.; et al. Scale to predict survival after surgery for recurrent glioblastoma multiforme. J. Clin. Oncol. 2010, 28, 3838–3843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Stupp, R.; Wong, E.T.; Kanner, A.A.; Steinberg, D.; Engelhard, H.; Heidecke, V.; Kirson, E.D.; Taillibert, S.; Liebermann, F.; Dbalý, V.; et al. NovoTTF-100A versus physician’s choice chemotherapy in recurrent glioblastoma: A randomised phase III trial of a novel treatment modality. Eur. J. Cancer 2012, 48, 2192–2202. [Google Scholar] [CrossRef] [Green Version]
  6. Stupp, R.; Taillibert, S.; Kanner, A.; Read, W.; Steinberg, D.; Lhermitte, B.; Toms, S.; Idbaih, A.; Ahluwalia, M.S.; Fink, K.; et al. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients with Glioblastoma: A Randomized Clinical Trial. JAMA 2017, 318, 2306–2316. [Google Scholar] [CrossRef] [Green Version]
  7. Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Hegg, R.; Im, S.A.; Shaw Wright, G.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef]
  8. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Long-Term Outcomes with Nivolumab Plus Ipilimumab or Nivolumab Alone Versus Ipilimumab in Patients with Advanced Melanoma. J. Clin. Oncol. 2022, 40, 127–137. [Google Scholar] [CrossRef]
  9. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef]
  10. Motzer, R.J.; Powles, T.; Burotto, M.; Escudier, B.; Bourlon, M.T.; Shah, A.Y.; Suárez, C.; Hamzaj, A.; Porta, C.; Hocking, C.M.; et al. Nivolumab plus cabozantinib versus sunitinib in first-line treatment for advanced renal cell carcinoma (CheckMate 9ER): Long-term follow-up results from an open-label, randomised, phase 3 trial. Lancet Oncol. 2022, 23, 888–898. [Google Scholar] [CrossRef]
  11. Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Arén Frontera, O.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.; Barthélémy, P.; Porta, C.; George, S.; et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2018, 378, 1277–1290. [Google Scholar] [CrossRef]
  12. Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [Green Version]
  13. Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bähr, O.; et al. Effect of Nivolumab vs Bevacizumab in Patients with Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef] [PubMed]
  14. Omuro, A.; Brandes, A.A.; Carpentier, A.F.; Idbaih, A.; Reardon, D.A.; Cloughesy, T.; Sumrall, A.; Baehring, J.; van den Bent, M.; Bähr, O.; et al. Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated MGMT promoter: An international randomized phase III trial. Neuro Oncol. 2023, 25, 123–134. [Google Scholar] [CrossRef]
  15. Lim, M.; Weller, M.; Idbaih, A.; Steinbach, J.; Finocchiaro, G.; Raval, R.R.; Ansstas, G.; Baehring, J.; Taylor, J.W.; Honnorat, J.; et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro Oncol. 2022, 24, 1935–1949. [Google Scholar] [CrossRef]
  16. Jackson, C.M.; Choi, J.; Lim, M. Mechanisms of immunotherapy resistance: Lessons from glioblastoma. Nat. Immunol. 2019, 20, 1100–1109. [Google Scholar] [CrossRef] [PubMed]
  17. Qazi, M.A.; Vora, P.; Venugopal, C.; Sidhu, S.S.; Moffat, J.; Swanton, C.; Singh, S.K. Intratumoral heterogeneity: Pathways to treatment resistance and relapse in human glioblastoma. Ann. Oncol. 2017, 28, 1448–1456. [Google Scholar] [CrossRef]
  18. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef]
  19. Joyce, J.A.; Fearon, D.T. T cell exclusion, immune privilege, and the tumor microenvironment. Science 2015, 348, 74–80. [Google Scholar] [CrossRef] [Green Version]
  20. Desland, F.A.; Hormigo, A. The CNS and the Brain Tumor Microenvironment: Implications for Glioblastoma Immunotherapy. Int. J. Mol. Sci. 2020, 21, 7358. [Google Scholar] [CrossRef] [PubMed]
  21. Akintola, O.O.; Reardon, D.A. The Current Landscape of Immune Checkpoint Blockade in Glioblastoma. Neurosurg. Clin. N. Am. 2021, 32, 235–248. [Google Scholar] [CrossRef] [PubMed]
  22. McGranahan, N.; Furness, A.J.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [Green Version]
  23. Pauken, K.E.; Wherry, E.J. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 2015, 36, 265–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Chi, X.; Luo, S.; Ye, P.; Hwang, W.L.; Cha, J.H.; Yan, X.; Yang, W.H. T-cell exhaustion and stemness in antitumor immunity: Characteristics, mechanisms, and implications. Front. Immunol. 2023, 14, 1104771. [Google Scholar] [CrossRef] [PubMed]
  25. Litak, J.; Mazurek, M.; Grochowski, C.; Kamieniak, P.; Roliński, J. PD-L1/PD-1 Axis in Glioblastoma Multiforme. Int. J. Mol. Sci. 2019, 20, 5347. [Google Scholar] [CrossRef] [Green Version]
  26. DeMatos, P.; Abdel-Wahab, Z.; Vervaert, C.; Hester, D.; Seigler, H. Pulsing of dendritic cells with cell lysates from either B16 melanoma or MCA-106 fibrosarcoma yields equally effective vaccines against B16 tumors in mice. J. Surg. Oncol. 1998, 68, 79–91. [Google Scholar] [CrossRef]
  27. Liu, B.Y.; Chen, X.H.; Gu, Q.L.; Li, J.F.; Yin, H.R.; Zhu, Z.G.; Lin, Y.Z. Antitumor effects of vaccine consisting of dendritic cells pulsed with tumor RNA from gastric cancer. World J. Gastroenterol. 2004, 10, 630–633. [Google Scholar] [CrossRef]
  28. Najafi, S.; Mortezaee, K. Advances in dendritic cell vaccination therapy of cancer. Biomed. Pharmacother. 2023, 164, 114954. [Google Scholar] [CrossRef]
  29. Polyzoidis, S.; Ashkan, K. DCVax®-L—developed by Northwest Biotherapeutics. Hum. Vaccines Immunother. 2014, 10, 3139–3145. [Google Scholar] [CrossRef] [Green Version]
  30. Qian, D.; Li, J.; Huang, M.; Cui, Q.; Liu, X.; Sun, K. Dendritic cell vaccines in breast cancer: Immune modulation and immunotherapy. Biomed. Pharmacother. 2023, 162, 114685. [Google Scholar] [CrossRef]
  31. Zhou, Q.; Guo, A.L.; Xu, C.R.; An, S.J.; Wang, Z.; Yang, S.Q.; Wu, Y.L. A dendritic cell-based tumour vaccine for lung cancer: Full-length XAGE-1b protein-pulsed dendritic cells induce specific cytotoxic T lymphocytes in vitro. Clin. Exp. Immunol. 2008, 153, 392–400. [Google Scholar] [CrossRef] [PubMed]
  32. Van de Velde, A.L.; Berneman, Z.N.; Van Tendeloo, V.F. Immunotherapy of hematological malignancies using dendritic cells. Bull. Cancer 2008, 95, 320–326. [Google Scholar] [PubMed]
  33. Heimberger, A.B.; Crotty, L.E.; Archer, G.E.; McLendon, R.E.; Friedman, A.; Dranoff, G.; Bigner, D.D.; Sampson, J.H. Bone marrow-derived dendritic cells pulsed with tumor homogenate induce immunity against syngeneic intracerebral glioma. J. Neuroimmunol. 2000, 103, 16–25. [Google Scholar] [CrossRef] [PubMed]
  34. Insug, O.; Ku, G.; Ertl, H.C.; Blaszczyk-Thurin, M. A dendritic cell vaccine induces protective immunity to intracranial growth of glioma. Anticancer Res. 2002, 22, 613–621. [Google Scholar]
  35. Pellegatta, S.; Poliani, P.L.; Corno, D.; Grisoli, M.; Cusimano, M.; Ubiali, F.; Baggi, F.; Bruzzone, M.G.; Finocchiaro, G. Dendritic cells pulsed with glioma lysates induce immunity against syngeneic intracranial gliomas and increase survival of tumor-bearing mice. Neurol. Res. 2006, 28, 527–531. [Google Scholar] [CrossRef]
  36. Fujita, M.; Zhu, X.; Ueda, R.; Sasaki, K.; Kohanbash, G.; Kastenhuber, E.R.; McDonald, H.A.; Gibson, G.A.; Watkins, S.C.; Muthuswamy, R.; et al. Effective immunotherapy against murine gliomas using type 1 polarizing dendritic cells—Significant roles of CXCL10. Cancer Res. 2009, 69, 1587–1595. [Google Scholar] [CrossRef] [Green Version]
  37. Liau, L.M.; Prins, R.M.; Kiertscher, S.M.; Odesa, S.K.; Kremen, T.J.; Giovannone, A.J.; Lin, J.W.; Chute, D.J.; Mischel, P.S.; Cloughesy, T.F.; et al. Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clin. Cancer Res. 2005, 11, 5515–5525. [Google Scholar] [CrossRef] [Green Version]
  38. Liau, L.M.; Ashkan, K.; Brem, S.; Campian, J.L.; Trusheim, J.E.; Iwamoto, F.M.; Tran, D.D.; Ansstas, G.; Cobbs, C.S.; Heth, J.A.; et al. Association of Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination with Extension of Survival Among Patients With Newly Diagnosed and Recurrent Glioblastoma: A Phase 3 Prospective Externally Controlled Cohort Trial. JAMA Oncol. 2023, 9, 112–121. [Google Scholar] [CrossRef]
  39. Liau, L.M.; Ashkan, K.; Tran, D.D.; Campian, J.L.; Trusheim, J.E.; Cobbs, C.S.; Heth, J.A.; Salacz, M.; Taylor, S.; D’Andre, S.D.; et al. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J. Transl. Med. 2018, 16, 142. [Google Scholar] [CrossRef] [Green Version]
  40. Gilbert, M.R.; Dignam, J.J.; Armstrong, T.S.; Wefel, J.S.; Blumenthal, D.T.; Vogelbaum, M.A.; Colman, H.; Chakravarti, A.; Pugh, S.; Won, M.; et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 2014, 370, 699–708. [Google Scholar] [CrossRef] [Green Version]
  41. Gilbert, M.R.; Wang, M.; Aldape, K.D.; Stupp, R.; Hegi, M.E.; Jaeckle, K.A.; Armstrong, T.S.; Wefel, J.S.; Won, M.; Blumenthal, D.T.; et al. Dose-dense temozolomide for newly diagnosed glioblastoma: A randomized phase III clinical trial. J. Clin. Oncol. 2013, 31, 4085–4091. [Google Scholar] [CrossRef] [Green Version]
  42. Weller, M.; Butowski, N.; Tran, D.D.; Recht, L.D.; Lim, M.; Hirte, H.; Ashby, L.; Mechtler, L.; Goldlust, S.A.; Iwamoto, F.; et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Wen, P.Y.; Reardon, D.A.; Armstrong, T.S.; Phuphanich, S.; Aiken, R.D.; Landolfi, J.C.; Curry, W.T.; Zhu, J.J.; Glantz, M.; Peereboom, D.M.; et al. A Randomized Double-Blind Placebo-Controlled Phase II Trial of Dendritic Cell Vaccine ICT-107 in Newly Diagnosed Patients with Glioblastoma. Clin. Cancer Res. 2019, 25, 5799–5807. [Google Scholar] [CrossRef] [PubMed]
  44. Brandes, A.A.; Carpentier, A.F.; Kesari, S.; Sepulveda-Sanchez, J.M.; Wheeler, H.R.; Chinot, O.; Cher, L.; Steinbach, J.P.; Capper, D.; Specenier, P.; et al. A Phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro Oncol. 2016, 18, 1146–1156. [Google Scholar] [CrossRef] [Green Version]
  45. Brandes, A.A.; Gil-Gil, M.; Saran, F.; Carpentier, A.F.; Nowak, A.K.; Mason, W.; Zagonel, V.; Dubois, F.; Finocchiaro, G.; Fountzilas, G.; et al. A Randomized Phase II Trial (TAMIGA) Evaluating the Efficacy and Safety of Continuous Bevacizumab Through Multiple Lines of Treatment for Recurrent Glioblastoma. Oncologist 2019, 24, 521–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Cloughesy, T.; Finocchiaro, G.; Belda-Iniesta, C.; Recht, L.; Brandes, A.A.; Pineda, E.; Mikkelsen, T.; Chinot, O.L.; Balana, C.; Macdonald, D.R.; et al. Randomized, Double-Blind, Placebo-Controlled, Multicenter Phase II Study of Onartuzumab Plus Bevacizumab Versus Placebo Plus Bevacizumab in Patients With Recurrent Glioblastoma: Efficacy, Safety, and Hepatocyte Growth Factor and O(6)-Methylguanine-DNA Methyltransferase Biomarker Analyses. J. Clin. Oncol. 2017, 35, 343–351. [Google Scholar]
  47. Galanis, E.; Anderson, S.K.; Twohy, E.L.; Carrero, X.W.; Dixon, J.G.; Tran, D.D.; Jeyapalan, S.A.; Anderson, D.M.; Kaufmann, T.J.; Feathers, R.W.; et al. A phase 1 and randomized, placebo-controlled phase 2 trial of bevacizumab plus dasatinib in patients with recurrent glioblastoma: Alliance/North Central Cancer Treatment Group N0872. Cancer 2019, 125, 3790–3800. [Google Scholar] [CrossRef]
  48. Lee, E.Q.; Zhang, P.; Wen, P.Y.; Gerstner, E.R.; Reardon, D.A.; Aldape, K.D.; deGroot, J.F.; Pan, E.; Raizer, J.J.; Kim, L.J.; et al. NRG/RTOG 1122: A phase 2, double-blinded, placebo-controlled study of bevacizumab with and without trebananib in patients with recurrent glioblastoma or gliosarcoma. Cancer 2020, 126, 2821–2828. [Google Scholar] [CrossRef]
  49. Lombardi, G.; De Salvo, G.L.; Brandes, A.A.; Eoli, M.; Rudà, R.; Faedi, M.; Lolli, I.; Pace, A.; Daniele, B.; Pasqualetti, F.; et al. Regorafenib compared with lomustine in patients with relapsed glioblastoma (REGOMA): A multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2019, 20, 110–119. [Google Scholar] [CrossRef]
  50. Narita, Y.; Arakawa, Y.; Yamasaki, F.; Nishikawa, R.; Aoki, T.; Kanamori, M.; Nagane, M.; Kumabe, T.; Hirose, Y.; Ichikawa, T.; et al. A randomized, double-blind, phase III trial of personalized peptide vaccination for recurrent glioblastoma. Neuro Oncol. 2019, 21, 348–359. [Google Scholar] [CrossRef] [Green Version]
  51. Taal, W.; Oosterkamp, H.M.; Walenkamp, A.M.; Dubbink, H.J.; Beerepoot, L.V.; Hanse, M.C.; Buter, J.; Honkoop, A.H.; Boerman, D.; de Vos, F.Y.; et al. Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): A randomised controlled phase 2 trial. Lancet Oncol. 2014, 15, 943–953. [Google Scholar] [CrossRef] [PubMed]
  52. Wick, W.; Gorlia, T.; Bendszus, M.; Taphoorn, M.; Sahm, F.; Harting, I.; Brandes, A.A.; Taal, W.; Domont, J.; Idbaih, A.; et al. Lomustine and Bevacizumab in Progressive Glioblastoma. N. Engl. J. Med. 2017, 377, 1954–1963. [Google Scholar] [CrossRef] [PubMed]
  53. Wick, W.; Puduvalli, V.K.; Chamberlain, M.C.; van den Bent, M.J.; Carpentier, A.F.; Cher, L.M.; Mason, W.; Weller, M.; Hong, S.; Musib, L.; et al. Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J. Clin. Oncol. 2010, 28, 1168–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Preusser, M.; van den Bent, M.J. Autologous tumor lysate-loaded dendritic cell vaccination (DCVax-L) in glioblastoma: Breakthrough or fata morgana? Neuro Oncol. 2023, 25, 631–634. [Google Scholar] [CrossRef] [PubMed]
  55. Pasqualetti, F.; Zanotti, S. Nonrandomised controlled trial in recurrent glioblastoma patients: The promise of autologous tumour lysate-loaded dendritic cell vaccination. Br. J. Cancer, 2023; epub ahead of print. [Google Scholar]
  56. Mishra-Kalyani, P.S.; Amiri Kordestani, L.; Rivera, D.R.; Singh, H.; Ibrahim, A.; DeClaro, R.A.; Shen, Y.; Tang, S.; Sridhara, R.; Kluetz, P.G.; et al. External control arms in oncology: Current use and future directions. Ann. Oncol. 2022, 33, 376–383. [Google Scholar] [CrossRef] [PubMed]
  57. Prasad, V. Reliable, cheap, fast and few: What is the best study for assessing medical practices? Randomized controlled trials or synthetic control arms? Eur. J. Clin. Investig. 2021, 51, e13580. [Google Scholar] [CrossRef] [PubMed]
  58. Rahman, R.; Ventz, S.; McDunn, J.; Louv, B.; Reyes-Rivera, I.; Polley, M.C.; Merchant, F.; Abrey, L.E.; Allen, J.E.; Aguilar, L.K.; et al. Leveraging external data in the design and analysis of clinical trials in neuro-oncology. Lancet Oncol. 2021, 22, e456–e465. [Google Scholar] [CrossRef]
  59. Platten, M.; Bunse, L.; Wick, A.; Bunse, T.; Le Cornet, L.; Harting, I.; Sahm, F.; Sanghvi, K.; Tan, C.L.; Poschke, I.; et al. A vaccine targeting mutant IDH1 in newly diagnosed glioma. Nature 2021, 592, 463–468. [Google Scholar] [CrossRef]
  60. Olivier, T.; Migliorini, D. Autologous tumor lysate-loaded dendritic cell vaccination in glioblastoma: What happened to the evidence? Rev. Neurol 2023, 179, 502–505. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gatto, L.; Di Nunno, V.; Tosoni, A.; Bartolini, S.; Ranieri, L.; Franceschi, E. DCVax-L Vaccination in Patients with Glioblastoma: Real Promise or Negative Trial? The Debate Is Open. Cancers 2023, 15, 3251. https://doi.org/10.3390/cancers15123251

AMA Style

Gatto L, Di Nunno V, Tosoni A, Bartolini S, Ranieri L, Franceschi E. DCVax-L Vaccination in Patients with Glioblastoma: Real Promise or Negative Trial? The Debate Is Open. Cancers. 2023; 15(12):3251. https://doi.org/10.3390/cancers15123251

Chicago/Turabian Style

Gatto, Lidia, Vincenzo Di Nunno, Alicia Tosoni, Stefania Bartolini, Lucia Ranieri, and Enrico Franceschi. 2023. "DCVax-L Vaccination in Patients with Glioblastoma: Real Promise or Negative Trial? The Debate Is Open" Cancers 15, no. 12: 3251. https://doi.org/10.3390/cancers15123251

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop