Summary
Background and Objectives
We examined retrospectively whether the combination of standard dacarbazine (DTIC) and/or fotemustine chemotherapy and autologous peptide‐loaded dendritic cell (DC) vaccination may improve survival of stage IV melanoma patients. Furthermore, a small cohort of long‐term survivors was studied in more detail.
Patients and methods
Between 1998 and 2008, 41 patients were vaccinated at least three times with DCs while receiving chemotherapy and compared to all other 168 patients in our database who only received chemotherapy (1993–2008).
Results
Median life expectancy of patients receiving additional DC‐vaccination was 18 months, compared to eleven months for patients under standard chemotherapy alone. In contrast to patients with other haplotypes, the HLA‐A1/A1 subset of DC‐treated patients showed significantly lower median survival (12 vs. 25 months). Autoantibodies were frequently detected in serum of both vaccinated and non‐vaccinated patients, and there was no correlation between titers, loss or appearance of autoantibodies and survival. Additionally, phenotyping of DCs and PBMCs also did not reveal any conspicuous correlation with survival.
Conclusions
Combining standard chemotherapy and DC vaccination appears superior to chemotherapy alone. The impact of HLA haplotypes on survival emphasizes the importance of a careful selection of patients with specific, well‐defined HLA haplotypes for future vaccination trials using peptide‐pulsed DCs, possibly combined with checkpoint inhibitors.
Introduction
Skin cancer, prominently including cutaneous melanoma, is rising in incidence and continues to pose a major medical challenge [1]. Prior to the introduction of modern checkpoint inhibitor therapy [2], treatment efficiency of metastatic stage IV melanoma was unsatisfactory with median survival times ranging from four to nine months. Dacarbazine (DTIC) and fotemustine were the only approved chemotherapies for disseminated melanoma, although none of these agents led to an improvement in overall survival [3, 4]. The high immunogenicity of melanoma, which can induce cytotoxic T lymphocyte (CTL)‐dependent spontaneous regression, prompted a number of different immunotherapeutic approaches to increase immunological responses [2, 5, 6]. In particular, dendritic cell (DC) vaccination showed promise for treating patients with advanced melanoma [7]. Indeed, small, initial trials using DC vaccines reported the regression of individual metastases [8, 9]. Although an initial, large, double‐blind trial reported DC vaccination responses comparable to chemotherapy in metastatic melanoma [10] (critically discussed in [11]), importantly, a recent, thorough 12‐year follow‐up of DC monotherapy‐treated patients revealed that survival periods were in the same range as with ipilimumab, but lacking major toxicity [11]. In spite of the recent decline in the number of DC therapy trials (concomitant with the advent of immune checkpoint therapy) there is continued interest and activity in DC‐based anti‐cancer immunization [12, 13, 14, 15], an important reason being that, despite impressive therapeutic success of immune checkpoint blockade, not all patients respond. The problem of resistance development also remains to be solved [16]. Therefore, further research efforts are needed. General agreement exists that combining different treatment strategies will be beneficial. Dendritic cells could be an important component of such combinatory approaches, not least because of the crucial role they play in checkpoint blockade immunotherapy, as described in recent reports [17, 18, 19].
The purpose of this retrospective study was therefore to elucidate whether one such combination, namely standard chemotherapy plus autologous tumor peptide‐loaded DC vaccination, could provide a survival benefit over standard chemotherapy alone in stage IV melanoma patients. In addition, the incidence, development and prognostic significance of autoimmunity were evaluated in DC‐vaccinated patients and compared with those treated with conventional chemotherapy alone. A limited number of long‐term survivors were studied in greater depth for indicative DC features associated with improved survival.
Material and Methods
Patients
All patients with stage IV melanoma who had been vaccinated with DCs at least three times since initiation of vaccination therapy in our department from October 1998 until May 2008 were included (n = 41) and compared to all other stage IV melanoma patients in our database who had been treated with standard chemotherapy (DTIC, 1000 mg/m2 i.v. in 4‐week intervals, and/or fotemustine, 100 mg/m2 i.v. weekly for 3 weeks, then every 4 weeks) from December 1993 (start of our database) to May 2008 (n = 168). One limitation of our study is, that no randomization of treatment arms was performed, therefore a selection bias cannot be ruled out. Selection of patients occurred according to their HLA status and was further limited by lab capacities.
Of note, none of these patients received checkpoint blockade immunotherapy. In addition, for some patients from this cohort DC treatment was continued until 2014, at most. Again, no checkpoint blockade immunotherapy was applied during this period of continued DC treatment. Most patients started out with DC monotherapy. Additional chemotherapy was only administered upon progression of the melanoma. Injections of DC and chemotherapy were never given simultaneously. Instead, whenever possible, a DC injection was placed exactly between two chemotherapy cycles, or alternatively, one day before chemotherapy.
A description of all methods can be found in Supplementary Materials.
Results
Patient characteristics
Patient demographics are listed in Table 1. All patients were of white/Caucasian background and had AJCC stage IV melanoma. We compared two patient cohorts, group 1 with 41 patients receiving standard chemotherapy plus tumor peptide‐pulsed DC vaccination and group 2 with 168 patients receiving DTIC and/or fotemustine only. Group 1 included 16 females and 25 males with a mean age of 59 years: group 2 contained 94 females and 74 males with a mean age of 65 years. Group 1 patients were HLA‐typed: 18 patients had at least one allele for HLA‐A1, 17 for HLA‐A2, 9 for HLA‐A3 and three for HLA‐24, whereas 12 patients were homozygous for HLA‐A1, 12 for HLA‐A2 and 6 for HLA‐A3 (Table S1; online Supporting Information).
Table 1.
Patient characteristics
Group 1: additional DC vaccination n (%) | Group 2: standard chemotherapy n (%) | |
---|---|---|
Randomized (n = 209) | 41 (100 %) | 168 (100 %) |
Caucasian | 41 (100 %) | 168 (100 %) |
Median age (years) | 59 | 65 |
Sex | ||
Male | 25 (61 %) | 74 (44 %) |
Female | 16 (39 %) | 94 (56 %) |
AJCC M category | ||
M1a | 3 (7 %) | 35 (21 %) |
M1b | 2 (5 %) | 20 (12 %) |
M1c | 36 (88 %) | 113 (67 %) |
In a small, exploratory follow‐up cohort we further investigated three patients that survived under continuous DC therapy for 11 (57 vaccinations), 14 (59 vaccinations), and 14.5 (51 vaccinations; still alive in 2020) years and compared them to three patients that died within 14 months from the start of DC vaccinations.
Survival of patients
The median overall survival of all stage IV melanoma patients within the observation period from 10/1998 to 05/2008 was 12 months (95 % confidence interval [CI]: 10.0–14.0) (Figure 1). Patients who were additionally vaccinated with autologous peptide‐pulsed DCs (group 1) had a median life expectancy of 18 months (95 % CI: 7.4–28.5), which was significantly higher than the eleven months – life expectancy of stage IV melanoma patients who received standard chemotherapy alone (group 2) (95 % CI: 8.5–13.5; log rank test P = 0.01) (Figure 1). Multivariate Cox regression analysis revealed a relative risk for worse outcome in male patients aged over 60 years (95 % CI: 0.6–1.2 and 0.8–1.5, respectively). As the unvaccinated patients were significantly older than the vaccinated ones we performed univariate and multivariate Cox regression analysis for survival, showing that the patients’ age did not impact the survival once having reached stage IV melanoma (Table S2; online Supporting Information). Importantly, the homozygous HLA‐A1/A1 subset of DC‐treated patients showed a significantly lower median survival of 12 months (95 % CI: 7.5–16.5) compared to patients carrying other haplotypes with a median survival of 25 months (95 % CI: 16.1–33.9; log rank test P = 0.015) (Figure 2).
Figure 1.
Kaplan‐Meier curves of overall survival by treatment arm. Arm A (in green: standard chemotherapy with DTIC and/or fotemustine), 168 patients and 136 deaths; arm B (in blue: standard chemotherapy plus DC vaccination), 41 patients and 31 deaths. P = 0.01 by log rank test. Censored observations are indicated by vertical bars.
Figure 2.
Kaplan‐Meier curves of overall survival of additionally DC‐vaccinated patients (group 1) by HLA haplotype. Arm A (in green: HLA‐A1/A1 haplotype), 12 patients and 12 deaths; arm B (in blue: all other HLA haplotypes), 29 patients and 19 deaths. P = 0.015 by log rank test. Censored observations are indicated by vertical bars.
Development of autoimmunity: autoantibodies and vitiligo
The presence, new development and loss of autoantibodies within the ten years observation period from 10/1998 to 05/2008 are summarized in Table 2. We analyzed 25 patients from group 1 and 21 from group 2. From the date of diagnosis autoantibodies (one or more) were detected in 13 of 25 (52 %) DC‐vaccinated (group 1) and in 11 of 21 (52 %) chemotherapy‐only‐treated (group 2) stage IV melanoma patients (P = 0.7). Between the first and the second measurement (18 months) autoantibodies appeared in eight (32 %) DC‐vaccinated and four (19 %) chemotherapy‐treated patients (P = 0.2). A loss in autoantibodies was observed in three patients of each treatment arm (P = 0.6). Two patients treated with standard chemotherapy alone had vitiligo at the time of progression to stage IV, whereas two DC‐vaccinated patients developed vitiligo during treatment. Notably, the presence, new development and loss of autoantibodies were not associated with a survival benefit, neither within each treatment group nor when all patients were considered together (Figure 3).
Table 2.
Presence, new development and loss of autoantibodies in stage IV melanoma patients receiving standard chemotherapy plus DC vaccination (group 1) or standard chemotherapy alone (group 2)
Autoantibodies | Group 1 (standard chemotherapy plus additional DC vaccination) (n = 25) | Group 2 (standard chemotherapy only) (n = 21) |
---|---|---|
First measurement | 13 (53 %) | 11 (52 %) |
Second measurement | 14 (56 %) | 8 (36 %) |
New development of autoantibodies | 8 (32 %) | 4 (21 %) |
Loss of autoantibodies | 3 (12 %) | 3 (14 %) |
Figure 3.
Kaplan‐Meier curves of overall survival by the presence of autoantibodies. Arm A (in green: patients with autoantibodies at second measurement), 22 patients and 15 deaths; arm B (in blue: patients without autoantibodies at second measurement), 24 patients and 20 deaths. P = 0.67 by log rank test. Censored observations are indicated by vertical bars.
Influence of dendritic cell quality on clinical outcome
Populations of peptide‐loaded mature DCs administered to the patients were routinely monitored and found to be of high quality as described in detail in Supplementary Materials.
Immunological follow‐up of a limited number of long‐term surviving patients
Immunohistochemistry: PD‐L1 expression on tumor cells is widely used as an – albeit imperfect – prognostic marker for responsiveness to immune checkpoint inhibitor therapy [20, 21]. Similarly, we hypothesized that high PD‐L1 expression on tumor cells may correlate with short survival, and vice versa. A small, exploratory sample of pre‐DC treatment tumors from five short‐term (< 2 years) and three long‐term (> 11 years) survivors was immunohistochemically probed for PD‐L1. Results were not conclusive. Further studies with larger samples are needed.
Flow cytometry: Cryopreserved peripheral blood mononuclear cells (PBMCs) from time points of the first leukapheresis, that is, before the start of DC vaccinations (for 3 short and 2 long‐term survivors) and, in addition, from 15, 55 and 82 months after the start of DC therapy for either of the three the long‐term survivors were analyzed with large panels of antibodies (Table S3, online Supplement Information). Peripheral blood mononuclear cells of patients before treatment showed no differences in the relative proportions of lymphoid cell populations (CD4+, CD8+ T cells and total CD56+CD3– NK cells) (Figure S1, online Supplement Information). Percentages of activated T cells, as determined by HLA‐DR staining, were also similar between the two groups (data not shown). Likewise, proportions of myeloid cell types (CD14+ monocytic cells, CD11c+CD1c–CD141+ cDC1 and CD11c+CD1c+ cDC2) did not differ grossly between long‐term and short‐term survivors (Figure S2, online Supplement Information). Percentages of activated CD14+ cells and cDC2 (as determined by CD83 expression) were also unchanged, and so was the expression of PD‐L1 on DCs (data not shown), the latter being a DC feature that, when experimentally blocked, was found to enhance T‐cell responses [22]. For long term survivors, percentages of lymphoid and myeloid cells were unaltered during treatment (data not shown).
Regarding immune effector cells, it is noteworthy that NK cells may have been critical in one of the patients (14 yrs. survival) who had markedly elevated numbers of CD56+ NK cells in the blood throughout the course of therapy. This, however, was already evident before the start of DC vaccination and can therefore not be ascribed to the DC therapy. The other long‐term survivors did not show such conspicuously and persistently elevated NK levels. Nevertheless, one observation might provide a “lead” to further analyses on subsets of NK cells, namely the CD56‐bright, cytolytically potent minority of NK cells in the blood versus the majority of CD56‐dim NK cells [23]. In our small cohort the long (> 10 years) surviving patients had proportionally slightly more CD56‐bright NK cells in their blood as compared to the short (< 2 years) surviving patients (ratios of CD56‐bright to CD56‐dim: 0.36 for long and 0.10 for short term survivors) (Figure S3, online Supplement Information).
Discussion
The principal finding of this retrospective study was that in contrast to the standard chemotherapy group, patients survived significantly longer under additional DC vaccination. The median life expectancy of patients who received additional DC‐vaccination was 18 months, compared to 11 months in patients who were treated with standard chemotherapy alone. Of note, these data are from the period from 1998 to 2008, and therefore none of the patients had received immune checkpoint immunotherapy or kinase inhibitors. Although DC vaccination can induce antigen specific immune responses in melanoma patients, only a few trials have shown a correlation between survival and detectable immune responses [24]. Numerous reasons were proposed for this discordance, ranging from the wide variety of methods for generating DCs to the routes of DC administration. Livingston et al. [25] and Wallack et al. [26], for example, found statistically significant survival benefits in the active specific immunotherapy group. Likewise, Kyte et al. [27] reported that DC‐vaccinated patients experienced a prolonged overall survival when retrospectively compared to a control group of non‐vaccinated patients receiving standard treatment. Although the first large‐scale, comparative clinical study by Schadendorf et al. [10] failed to demonstrate that DC‐vaccination was superior to DTIC treatment, explorative subgroup analyses indicated that HLA‐A2+/HLA‐B44– vaccinated patients survived longer than DTIC treated patients. This difference was not observed in the DTIC arm. Similarly, Sosman et al. [28] documented a similarly beneficial effect of an allogeneic tumor vaccine in patients positive for HLA‐A2.
Our findings are consistent with these data, and with recent observations by Gross et al. [11], who found an association of HLA‐A1/A1 and unfavorable clinical outcome, with other haplotypes being more frequent in long‐term survivors. One of several possible reasons for the better response of HLA‐A2 patients may be that more peptides on more DC batches were administered per vaccination for the A2 haplotype. Based on these observations, we strongly suggest that the expression pattern of HLA haplotypes could have a major impact on vaccination efficacy. Therefore, we recommend that only patients with specific, well‐defined HLA haplotype characteristics should be enrolled in future vaccination trials using peptide‐loaded DCs.
There is increasing evidence for a potential synergistic effect of DC vaccination and conventional chemotherapy [29, 30, 31, 32]. The development of drug resistance to conventional chemotherapy is considered to be the major cause for treatment failure in many types of cancer. One way in which cancer cells become resistant is through increased expression of regulator proteins of apoptosis (e.g., survivin). In a combination scenario, chemotherapy would kill most cancer cells, leaving behind only resistant cells expressing high levels of these proteins. This would render them particularly susceptible to killing by DC vaccination‐induced T cells (e.g., against survivin) [31, 33].
In addition, immunogenic cell death may be brought about by certain forms of chemotherapy, which may provide copious tumor antigen for DCs to present to T cells [34, 35, 36]. The improved efficacy of DC vaccination might also be due to the depletion of CD4+, CD25+, Foxp3+ regulatory T cells (Treg) by the administration of chemotherapy prior to immunotherapy [37]. These cells were shown to play an inhibitory role in anti‐tumor responses.
As has been shown by others, our retrospective study revealed that vaccination with autologous tumor peptide‐pulsed DCs is a safe procedure and does not induce more autoimmune phenomena [8, 9, 38] as compared to the control chemotherapy treatment. This contrasts with the well‐known occurrence of autoantibodies [39] and autoimmune pathology [40, 41] in immune checkpoint immunotherapy. In general, autoantibodies were frequently detected in both vaccinated and non‐vaccinated stage IV melanoma patients. Also, vitiligo occurred in only two patients of each treatment group. It should be noted that there was no correlation between development of autoimmunity and a better prognosis, strongly indicating that the survival benefit seen in vaccinated stage IV melanoma patients was due to the induction of specific antitumor immunity.
In addition to mere efficacy considerations, but also in view of our autoantibody findings, it should be emphasized that DC‐based immunotherapy is well tolerated. We observed this in our patients over the years, and it could also be objectified in quality of life (QoL) studies in melanoma [42] and – much earlier – in renal cell carcinoma [43].
In search for possible immune correlates for prolonged survival we followed up a small, exploratory cohort of DC plus chemo‐treated stage IV metastatic melanoma patients representing the extremes of the observed spectrum, that is, patients who survived for more than eleven years versus patients who died within 14 months of the onset of DC therapy. PD‐L1 expression patterns on the primary melanomas of five short and three long‐term survivors were not conclusive. More samples are needed to reveal a possible correlation. Nor did the flow cytometry analysis of lymphoid and myeloid leukocytes subsets in pre‐therapy PBMCs, including DC subsets (cDC1, as defined by CD141 and cDC2) of three short and two long‐term survivors provide a strong and reliable prognostic marker for responsiveness to DC therapy, specifically survival. The only possible “lead” was found in NK cells of three short and three long‐term survivors, where long‐term survivors had proportionally more CD56‐bright, strongly cytotoxic NK cells [23]. This observation, however, needs more investigation. Interestingly, the recent 12‐years follow‐up analysis of DC‐vaccinated melanoma patients by the Erlangen group [11] could not corroborate some correlations that would have seemed obvious at first glance: Prolonged survival did not correlate with numbers of regulatory T cells or myeloid‐derived suppressor cells (MDSC) cells, or with frequencies of vaccine‐specific T cells (as determined by Elispot and tetramer staining) in the blood, but rather with strong reactions at the vaccine injection sites, and with blood eosinophilia of 20 % or more at any time point during (though not before) vaccination therapy [44]. Similarly, a substantial proportion (50 %) of our patients treated with DC‐based immunotherapy plus chemotherapy experienced at least once an eosinophilia of 5 % or more during the course of therapy, but only one of 41 (2.4 %) reached levels of more than 10 %, and none had more than 20 %. However, a trend, although not statistically significant, appeared. Eosinophilia of 5–10 % occurred in eleven of 14 (79 %) long‐term surviving patients (> 18 months) as compared to 10 of 28 (36 %) patients surviving less than 18 months. The reason for the absence of eosinophilias beyond 20 % remains elusive. Speculatively, the concomitant chemotherapy might have had an influence. Local reactions in response to the vaccinations were occasional reddening of the injection site and occasional sub‐febrile temperature. However, these observations were not systematically correlated with clinical responses. An elevated ratio of myeloid to lymphoid (T, B, NK) cells in the blood was reported to correlate with poor survival and low levels of PEBP1 expression [45]. Our low sample size did not allow substantiation of this observation. Interestingly, patient survival also did not correlate with the phenotypical quality of administered mature DCs. This observation emphasizes, however, that monocyte‐derived DCs generated from the blood of cancer patients can mature perfectly well and are fully intact.
In summary, we showed herein that standard chemotherapy in combination with autologous tumor peptide‐loaded DC vaccination is more effective in stage IV melanoma patients than standard chemotherapy alone. Moreover, the response to DC vaccination appears to be influenced by the HLA haplotype. In this respect, combining DC vaccination with treatment regimens other than chemotherapy, such as immune checkpoint inhibitors and inhibitors of the RAS/RAF/MEK/ERK pathway, may be worthy of further consideration to circumvent the complex tumor escape mechanisms and thus to achieve better clinical responses. Indeed, some initial trials have already been performed, albeit with limited success [14, 46]. This underscores the need for further optimization and standardization of current DC vaccination protocols, especially in terms of DC generation, administration (e.g., targeting of antigen to DC subsets [47, 48, 49, 50]) and antigen preparation. DC will remain an important component of the (immuno)therapeutic “toolbox” against cancer.
Funding and Acknowledgement
Personnel (S.E., D.R., M.F.) and infrastructure for the manufacturing and quality control of DC for therapy was financed over the years by Kompetenzzentrum Medizin Tirol and the Oncotyrol research consortium. Oncotyrol was funded by the Austrian Research Promotion Agency (FFG) and the Regional Government (Standortagentur Tirol). We highly appreciate the continuous participation of tirol‐kliniken, who served as a partner of Oncotyrol. Additional support came from Austrian Science Fund projects P29919 to N.R. and P27001 to P.S.
This paper is dedicated to Prof. Peter Fritsch on the occasion of his 80th birthday. He was Director of the Department of Dermatology & Venereology from 1981 to 2008. Under his auspices and with his input and support the largest part of the reported studies was performed.
Conflict of interest
None.
Supporting information
Supplement Information
Supplement Information
Table S1
Table S2
Table S3
Table S4
Figure S1
Figure S2
Figure S3
References
- 1. Krensel M, Petersen J, Mohr P et al. Estimating prevalence and incidence of skin cancer in Germany. J Dtsch Dermatol Ges 2019; 17: 1239–49. [DOI] [PubMed] [Google Scholar]
- 2. Fritz JM, Lenardo MJ. Development of immune checkpoint therapy for cancer. J Exp Med 2019; 216: 1244–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Atkins MB. The treatment of metastatic melanoma with chemotherapy and biologics. Curr Opin Oncol 1997; 9: 205–13. [DOI] [PubMed] [Google Scholar]
- 4. Eigentler TK, Caroli UM, Radny P et al. Palliative therapy of disseminated malignant melanoma: a systematic review of 41 randomised clinical trials. Lancet Oncol 2003; 4: 748–59. [DOI] [PubMed] [Google Scholar]
- 5. Celluzzi CM, Mayordomo JI, Storkus WJ et al. Peptide‐pulsed dendritic cells induce antigen‐specific CTL‐mediated protective tumor immunity. J Exp Med 1996; 183: 283–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Hodi FS, O’Day SJ, McDermott DF et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363: 711–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Schuler G, Steinman RM. Dendritic cells as adjuvants for immune‐mediated resistance to tumors. J Exp Med 1997; 186: 1183–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Nestle FO, Alijagic S, Gilliet M et al. Vaccination of melanoma patients with peptide‐ or tumor lysate‐pulsed dendritic cells. Nat Med 1998; 4: 328–32. [DOI] [PubMed] [Google Scholar]
- 9. Thurner B, Haendle I, Röder C et al. Vaccination with mage‐3A1 peptide‐pulsed mature, monocyte‐derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J Exp Med 1999; 190: 1669–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Schadendorf D, Ugurel S, Schuler‐Thurner B et al. Dacarbazine (DTIC) versus vaccination with autologous peptide‐pulsed dendritic cells (DC) in first‐line treatment of patients with metastatic melanoma: a randomized phase III trial of the DC study group of the DeCOG. Ann Oncol 2006; 17: 563–70. [DOI] [PubMed] [Google Scholar]
- 11. Gross S, Erdmann M, Haendle I et al. Twelve‐year survival and immune correlates in dendritic cell‐vaccinated melanoma patients. JCI Insight 2017; 2: e91438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Sprooten J, Ceusters J, Coosemans A et al. Trial watch: dendritic cell vaccination for cancer immunotherapy. Oncoimmunology 2019; 8: e1638212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Wculek SK, Cueto FJ, Mujal AM et al. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol 2020; 20: 7–24. [DOI] [PubMed] [Google Scholar]
- 14. Bassani‐Sternberg M, Digklia A, Huber F et al. A phase Ib study of the combination of personalized autologous dendritic cell vaccine, aspirin, and standard of care adjuvant chemotherapy followed by nivolumab for resected pancreatic adenocarcinoma‐a proof of antigen discovery feasibility in three patients. Front Immunol 2019; 10: 1832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Garg AD, Coulie PG, Van den Eynde BJ et al. Integrating next‐generation dendritic cell vaccines into the current cancer immunotherapy landscape. Trends Immunol 2017; 38: 577–93. [DOI] [PubMed] [Google Scholar]
- 16. Sharma P, Allison JP. Dissecting the mechanisms of immune checkpoint therapy. Nat Rev Immunol 2020; 20: 75–6. [DOI] [PubMed] [Google Scholar]
- 17. Garris CS, Arlauckas SP, Kohler RH et al. Successful anti‐PD‐1 cancer immunotherapy requires t cell‐dendritic cell crosstalk involving the cytokines IFN‐gamma and IL‐12. Immunity 2018; 49: 1148–61 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Moussion C, Mellman I. The dendritic cell strikes back. Immunity 2018; 49: 997–9. [DOI] [PubMed] [Google Scholar]
- 19. Oh SA, Wu D‐C, Cheung J et al. PD‐L1 expression by dendritic cells is a key regulator of T‐cell immunity in cancer. Nature Cancer 2020; 1: 681–91. [DOI] [PubMed] [Google Scholar]
- 20. Robert C, Long GV, Brady B et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015; 372: 320–30. [DOI] [PubMed] [Google Scholar]
- 21. Daud AI, Wolchok JD, Robert C et al. Programmed death‐ligand 1 expression and response to the anti‐programmed death 1 antibody pembrolizumab in melanoma. J Clin Oncol 2016; 34: 4102–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Brown JA, Dorfman DM, Ma FR et al. Blockade of programmed death‐1 ligands on dendritic cells enhances T cell activation and cytokine production. J Immunol 2003; 170: 1257–66. [DOI] [PubMed] [Google Scholar]
- 23. Fehniger TA, Cooper MA, Nuovo GJ et al. CD56bright natural killer cells are present in human lymph nodes and are activated by T cell‐derived IL‐2: a potential new link between adaptive and innate immunity. Blood 2003; 101: 3052–7. [DOI] [PubMed] [Google Scholar]
- 24. Engell‐Noerregaard L, Hansen TH, Andersen MH et al. Review of clinical studies on dendritic cell‐based vaccination of patients with malignant melanoma: assessment of correlation between clinical response and vaccine parameters. Cancer Immunol Immunother 2009; 58: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Livingston PO, Wong GY, Adluri S et al. Improved survival in stage III melanoma patients with GM2 antibodies: a randomized trial of adjuvant vaccination with GM2 ganglioside. J Clin Oncol 1994; 12: 1036–44. [DOI] [PubMed] [Google Scholar]
- 26. Wallack MK, Sivanandham M, Balch CM et al. Surgical adjuvant active specific immunotherapy for patients with stage III melanoma: the final analysis of data from a phase III, randomized, double‐blind, multicenter vaccinia melanoma oncolysate trial. J Am Coll Surg 1998; 187: 69–77; discussion 77–9. [DOI] [PubMed] [Google Scholar]
- 27. Kyte JA, Mu L, Aamdal S et al. Phase I/II trial of melanoma therapy with dendritic cells transfected with autologous tumor‐mRNA. Cancer Gene Ther 2006; 13: 905–18. [DOI] [PubMed] [Google Scholar]
- 28. Sosman JA, Unger JM, Liu PY et al. Adjuvant immunotherapy of resected, intermediate‐thickness, node‐negative melanoma with an allogeneic tumor vaccine: impact of HLA class I antigen expression on outcome. J Clin Oncol 2002; 20: 2067–75. [DOI] [PubMed] [Google Scholar]
- 29. Truxova I, Hensler M, Skapa P et al. Rationale for the combination of dendritic cell‐based vaccination approaches with chemotherapy agents. Int Rev Cell Mol Biol 2017; 330: 115–56. [DOI] [PubMed] [Google Scholar]
- 30. Gabrilovich DI. Combination of chemotherapy and immunotherapy for cancer: a paradigm revisited. Lancet Oncol 2007; 8: 2–3. [DOI] [PubMed] [Google Scholar]
- 31. Andersen MH, Sorensen RB, Schrama D et al. Cancer treatment: the combination of vaccination with other therapies. Cancer Immunol Immunother 2008; 57: 1735–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Boudewijns S, Bloemendal M, de Haas N et al. Autologous monocyte‐derived DC vaccination combined with cisplatin in stage III and IV melanoma patients: a prospective, randomized phase 2 trial. Cancer Immunol Immunother 2020; 69: 477–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Becker JC, Andersen MH, Hofmeister‐Muller V et al. Survivin‐specific T‐cell reactivity correlates with tumor response and patient survival: a phase‐II peptide vaccination trial in metastatic melanoma. Cancer Immunol Immunother 2012 61: 2091–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Pitt JM, Kroemer G, Zitvogel L. Immunogenic and non‐immunogenic cell death in the tumor microenvironment. Adv Exp Med Biol 2017; 1036: 65–79. [DOI] [PubMed] [Google Scholar]
- 35. Chen DS, and Mellman I. Oncology meets immunology: the cancer‐immunity cycle. Immunity 2013; 39: 1–10. [DOI] [PubMed] [Google Scholar]
- 36. Belvin M, Mellman I. Is all cancer therapy immunotherapy? Sci Transl Med 2015; 7: 315fs48. [DOI] [PubMed] [Google Scholar]
- 37. Lutsiak ME, Semnani RT, De Pascalis R et al. Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low‐dose cyclophosphamide. Blood 2005; 105: 2862–8. [DOI] [PubMed] [Google Scholar]
- 38. Hersey P, Menzies SW, Halliday GM et al. Phase I/II study of treatment with dendritic cell vaccines in patients with disseminated melanoma. Cancer Immunol Immunother 2004; 53: 125–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Tahir SA, Gao J, Miura Y et al. Autoimmune antibodies correlate with immune checkpoint therapy‐induced toxicities. Proc Natl Acad Sci U S A 2019; 116: 22246–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Postow MA, Hellmann MD. Adverse events associated with immune checkpoint blockade. N Engl J Med 2018; 378: 1165. [DOI] [PubMed] [Google Scholar]
- 41. Gutzmer R, Koop A, Meier F et al. Programmed cell death protein‐1 (PD‐1) inhibitor therapy in patients with advanced melanoma and preexisting autoimmunity or ipilimumab‐triggered autoimmunity. Eur J Cancer 2017; 75: 24–32. [DOI] [PubMed] [Google Scholar]
- 42. Bloemendal M, Rietveld MJA, van Willigen WW et al. Health‐related quality of life analysis in stage III melanoma patients treated with adjuvant dendritic cell therapy. Clin Transl Oncol 2019; 21: 774–80. [DOI] [PubMed] [Google Scholar]
- 43. Leonhartsberger N, Ramoner R, Falkensammer C et al. Quality of life during dendritic cell vaccination against metastatic renal cell carcinoma. Cancer Immunol Immunother 2012; 61: 1407–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Moreira A, Erdmann M, Uslu U et al. Blood eosinophilia is an on‐treatment biomarker in patients with solid tumors undergoing dendritic cell vaccination with autologous tumor‐RNA. Pharmaceutics 2020; 12: 210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Buschow SI, Ramazzotti M, Reinieren‐Beeren IMJ et al. Survival of metastatic melanoma patients after dendritic cell vaccination correlates with expression of leukocyte phosphatidylethanolamine‐binding protein 1/Raf kinase inhibitory protein. Oncotarget 2017; 8: 67439–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Nowicki TS, Berent‐Maoz B, Cheung‐Lau G et al. A pilot trial of the combination of transgenic NY‐ESO‐1‐reactive adoptive cellular therapy with dendritic cell vaccination with or without ipilimumab. Clin Cancer Res 2019; 25: 2096–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Dörrie J, Schaft N, Schuler G et al. Therapeutic cancer vaccination with ex vivo RNA‐transfected dendritic cells‐an update. Pharmaceutics 2020; 12: 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Dhodapkar MV, Sznol M, Zhao B et al. Induction of antigen‐specific immunity with a vaccine targeting NY‐ESO‐1 to the dendritic cell receptor DEC‐205. Sci Transl Med 2014; 6: 232ra51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Wamhoff EC, Schulze J, Bellmann L et al. A specific, glycomimetic langerin ligand for human Langerhans cell targeting. ACS Cent Sci 2019; 5: 808–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Stoitzner P, Schaffenrath S, Tripp CH et al. Human skin dendritic cells can be targeted in situ by intradermal injection of antibodies against lectin receptors. Exp Dermatol 2014; 23: 909–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplement Information
Supplement Information
Table S1
Table S2
Table S3
Table S4
Figure S1
Figure S2
Figure S3