Next Article in Journal
Micro RNAs as a Diagnostic Marker between Glioma and Primary CNS Lymphoma: A Systematic Review
Previous Article in Journal
Perceptions of Canadian Radiation Oncologists, Medical Physicists, and Radiation Trainees about the Feasibility and Need of Boron Neutron Capture Therapy (BNCT) in Canada: A National Survey
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Clinical Trial on the Safety and Tolerability of Personalized Cancer Vaccines Using Human Platelet Lysate-Induced Antigen-Presenting Cells

1
Department of Regenerative Medicine, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
2
Center for Regenerative Medicine, Kanazawa Medical University Hospital, Kahoku 920-0293, Ishikawa, Japan
3
Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
4
Department of Hematology and Immunology, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
5
Division of Stem Cell Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
6
Department of Pathology and Laboratory Medicine, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
7
Department of Obstetrics and Gynecology, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
8
Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
9
Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
10
Office of Research & Development, Kaohsiung Medical University, Kaohsiung 807, Taiwan
11
Internal Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
12
Division of Transfusion and Cell Therapy, Tokyo Women’s Medical University, Adachi Medical Center, Adachi 123-8558, Tokyo, Japan
13
Department of Transfusion Medicine and Cell Processing, Tokyo Women’s Medical University, Shinjuku 162-8666, Tokyo, Japan
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(14), 3627; https://doi.org/10.3390/cancers15143627
Submission received: 27 May 2023 / Revised: 27 June 2023 / Accepted: 11 July 2023 / Published: 14 July 2023
(This article belongs to the Section Cancer Immunology and Immunotherapy)

Abstract

:

Simple Summary

In this study, we developed human platelet lysate-induced antigen-presenting cells (HPL-APCs) from peripheral monocytes with a high potency of presentation ability. This study aimed to verify the safety, tolerability, and immunoinducibility of HPL-APCs loaded with cancer candidate antigens as a Phase I study. As a result of the interim analysis, safety and tolerability were confirmed in three enrolled patients, and the immune response to cancer antigen candidate peptides predicted in silico was confirmed in two completed cases. This clinical study is the first to verify the feasibility and immunoinducibility of a personalized cancer vaccine using HPL-APCs that would be expected to demonstrate further antitumor activity through optimized combination therapies.

Abstract

Research and development of personalized cancer vaccines as precision medicine are ongoing. We predicted human leukocyte antigen (HLA)-compatible cancer antigen candidate peptides based on patient-specific cancer genomic profiles and performed a Phase I clinical trial for the safety and tolerability of cancer vaccines with human platelet lysate-induced antigen-presenting cells (HPL-APCs) from peripheral monocytes. Among the five enrolled patients, two patients completed six doses per course (2–3 × 107 cells per dose), and an interim analysis was performed based on the immune response. An immune response was detected by enzyme-linked immunosorbent spot (ELISpot) assays to HLA-A*33:03-matched KRASWT, HLA-DRB1*09:01-compliant KRASWT or G12D, or HLA-A*31:01-matched SMAD4WT, and HLA-DRB1*04:01-matched SMAD4G365D peptides in two completed cases, respectively. Moreover, SMAD4WT-specific CD8+ effector memory T cells were amplified. However, an attenuation of the acquired immune response was observed 6 months after one course of cancer vaccination as the disease progressed. This study confirmed the safety and tolerability of HPL-APCs in advanced and recurrent cancers refractory to standard therapy and is the first clinical report to demonstrate the immunoinducibility of personalized cancer vaccines using HPL-APCs. Phase II clinical trials to determine immune responses with optimized adjuvant drugs and continued administration are expected to demonstrate efficacy.

Graphical Abstract

1. Introduction

Advances in next-generation sequencing technology make it possible to treat cancer as precision medicine based on individual cancer genome information. However, only a limited number of patients can benefit from individualized therapy [1], and cancer vaccines based on patient-specific cancer genome information are expected as novel cancer treatments [2].
Cancer vaccines targeting either tumor-associated antigens or tumor-specific antigens, including neoantigens generated by genetic mutations of cancer cells, are being developed [3]. Among cancer vaccine platforms including RNA, DNA, peptides, and dendritic cells (DCs) [4], DC vaccines have been reported to have strong immunoinductibility [5,6]. Immune memory is important for the sustained efficacy of cancer vaccines [7], and the function of DCs that play a central role in immune acquisition is attracting attention [8].
Clinical trials of neoantigen-pulsed DC vaccines using in silico analysis based on cancer genomic information in patients with advanced lung cancer have been conducted [9], and the immune response to neoantigens targeting compatible peptides on HLA class I or II has been confirmed. On the contrary, the immune memory induced by DC vaccination must be comprehensively investigated.
Clinical trials have been conducted on classical DC vaccines derived from monocytes using granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4) [10,11,12]. GM-CSF and interferon-α-generated DCs (IFN-DCs) are reported to exhibit antigen presentation and termination activity against tumor cells via TRAIL, FAS ligand and granzyme B/ perforin pathway [13,14]. We further developed IFN-DCs using human platelet lysates (HPL), named human platelet lysate-inducible antigen-presenting cells (HPL-APCs) with high endocytic and proteolytic activities [15], which exhibit strong antigen presentation ability compared to IFN-DCs [15]. We detected SMAD4P130L in pancreatic cancer cells in malignant pleural effusion and reported the inducibility of HLA-A*11:01-restricted SMAD4P130L neoantigen-specific CD8+ T cells in vitro using HPL-APCs [16]. Therefore, HPL-APCs are expected to be used as a platform for personalized cancer vaccines.
In this study, we verified the safety and tolerability of HPL-APCs loaded with personalized cancer candidate antigens based on cancer genomic profiles and immune memory in a Phase I trial in advanced and recurrent cancers.

2. Materials and Methods

2.1. Manufacture of HPL-APCs

Peripheral blood mononuclear cell (PBMC)-rich fractions through leukapheresis were harvested using a Spectra Optia® cell separator (Terumo BCT, Inc., Tokyo, Japan). PBMCs were separated with density gradient centrifugation using Ficoll-Plaque Premium (Global Life Sciences Solutions USA LLC, Marlborough, MA, USA) and were washed out with phosphate-buffered saline (Wako Pure Chemicals Ltd., Osaka, Japan). Then, the PBMCs were suspended in serum-free medium DCO-K (Nissui Pharmaceutical Co., Ltd., Tokyo, Japan) supplemented with 5% of GMP graded UltraGROTM-PURE GI (hereinafter called “HPL”; AventaCell BioMedical Corp., Atlanta, GA, USA), which is an HPL reagent approved as a material for regenerative medicine products by the Pharmaceuticals and Medical Devices Agency in Japan. The PBMCs’ suspension was seeded into adherent culture dishes. After 30 min incubation, the nonadherent cells were removed, and the adherent monocytes were cultured in DCO-K medium containing 100 ng/mL recombinant human GM-CSF (Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany) and 1 µg/mL PEGylated-IFN-α-2a (PEGASYS®; Chugai Pharmaceutical Co., Ltd., Tokyo, Japan) with 5% HPL for 3-day cultivation. For maturation, immature APCs were collected and resuspended in DCO-K medium with 5% HPL with 100 ng/mL GM-CSF, 1 µg/mL PEGASYS®, 10 µg/mL OK-432 (Streptococcal Preparation, Chugai), 10 ng/mL prostaglandin E2 (PGE2; Kyowa Pharma Chemical Co., Ltd., Toyama, Japan), and 10 µg/mL cancer antigen candidate peptides (synthesized by GenScript USA Inc., Piscataway, NJ, USA). After cultivation for 24 h in low adherent cell culture dishes, the cells were harvested and cryopreserved at −152 °C. The phenotype of HPL-APCs was determined using BD FACSLyricTM (BD Biosciences, Franklin Lakes, NJ, USA). HPL-APCs were treated with FcR Blocking Reagent (Miltenyi Biotec) at room temperature for 10 min and stained with antibodies (Table S1) at 4 °C for 60 min. Their respective isotype antibodies were used as the negative control. Live cells defined as negative for 7-aminoactinomycin D (7-AAD, Miltenyi Biotec) were analyzed using FACSLyricTM (BD Biosciences). Data were analyzed with FlowJoTM (BD Biosciences).

2.2. Prediction of Cancer Antigen Candidate Peptides

Cancer antigen candidate peptides based on the binding affinities of 8- to 14-mer to HLA-A or 12- to 18-mer to HLA-DPB1/DRB1 were examined using the IEDB analysis resource NetMHCpan (ver. 4.1) [17]. The peptides that predicted the binding affinity the most strongly were selected for the HPL-APC vaccine according to the following priority order: (1) mutated peptides with IC50 < 500 nM and (2) wild-type peptides with IC50 < 500 nM.

2.3. HPL-APC Vaccination

The release criteria of the HPL-APC vaccine were set at CD86+HLA-DR+, >70% viability, negative for bacterial and fungal infections, < 1 EU/mL of endotoxin, and negative for mycoplasma. Cryopreserved HPL-APCs were thawed during each session and suspended in 1 mL of saline. The HPL-APC vaccine was intradermally and bilaterally administered near the axillary region and groin.

2.4. Evaluation of Safety and Tolerability

The evaluation of safety included the following: (1) any systemic reactions at an early phase within 48 h of the intradermal injection of the HLA-APC vaccine (presence of high fever, hypotension, shock tachycardia, bradycardia, breathing difficulties, or skin rash) and (2) local skin reactions at the injection sites; other symptoms, including nausea, vomiting, diarrhea, appetite loss, mucosal ulcer, or central nervous system disorders; and laboratory test dysfunctions: anemia, reduced white blood cells, reduced platelet count, or elevated kidney and liver serum levels during or after the course of vaccination as described previously [10].

2.5. Enzyme-Linked Immunosorbent Spot (ELISpot) Assays

To investigate the antigen-specific IFN-γ production, ELISpot assays were performed using the human IFN-γ ELISpot PLUS kit (Mabtech AB, Nacka Strand, Sweden). Moreover, 1 × 106 PBMCs were incubated in 96 wells with AIM-V medium with 10% fetal bovine serum (FBS) and 10 μM peptides. After 16–20 h of incubation, IFN-γ producing cells were detected according to the manufacturer’s protocol. The spots were calculated using an automatic ELISpot reader (AID ELISpot Reader Classic ELR 07; Autoimmun Diagnostika GmbH, Strassberg, Germany). The counted numbers with cancer candidate peptides were compared with the HLA-matched HIV peptide (Table S2) as a negative control. The immune response to cancer antigen candidate peptides was defined according to the following criteria: (1) at least 15 spots per 1 × 106 PBMCs and (2) at least 1.5-fold more presence of spots than negative control peptide spots [18].

2.6. Detection of Monocytic MDSCs (M-MDSCs) and Regulatory T Cells (Tregs)

PBMCs were treated with FcR Blocking Reagent (Miltenyi Biotec) and stained with antibodies in Table S3 or Table S4. Live cells defined as negative for 7-AAD (Miltenyi Biotec) were analyzed using FACSCanto II (BD Biosciences) or FACSLyric (BD Biosciences). Tregs were defined as CD45+CD3+CD4+CD25+CD127−/low cells [19]. M-MDSCs were defined as CD45+CD11b+CD33+CD14+HLA-DR cells [20].

2.7. Detection of Antigen-Specific Memory T Cell Subsets

PBMCs were incubated with an AIM-V medium with 10% FBS and 10 μM peptides. HLA-matched HIV peptide was used as the negative control. After 16–20 h incubation, cells were washed and treated with FcR Blocking Reagent (Miltenyi Biotec) and stained with antibodies as shown in Table S5 at 4 °C for 60 min. To detect antigen-specific memory T cells, the activation of CD137 on CD8+ T cells was analyzed [21]. Further, naïve cells as CD45ROCCR7+, central memory cells as CD45RO+CCR7+, effector memory cells as CD45RO+CCR7, and effector cells as CD45ROCCR7 were identified for memory T cell subsets [22].

2.8. Case Report

2.8.1. Case 1 (Patient 2)

A male patient in his 50s was diagnosed with pancreatic cancer (stage IV, KRASG12D gene mutation). Apheresis was performed without adverse events such as vasovagal reflexes. A course of HPL-APC vaccination for KRAS combined with gemcitabine + protein-bound paclitaxel chemotherapy was completed in six doses, with a total cell count of 14.0 × 107 cells (mean viable cell rate, 91.9%). No allergic reaction with delayed-type hypersensitivity (DTH) lower than 5 mm to peptides was noted, and a grade 2 fever reaction and redness of grade 2 at the vaccine-injected sites were observed; however, no other adverse reactions or adverse events were reported. No adverse events were found during the observation period, and follow-up continued. Diagnostic imaging after one course revealed a stable disease (SD); however, 4 months after one course, imaging of the metastasized lung showed a progressive disease (PD), and chemotherapy with 5FU + irinotecan hydrochloride hydrate was performed.

2.8.2. Case 2 (Patient 3)

A female patient in her 40s was diagnosed with cervical cancer (adenocarcinoma, stage IV, KRASG12D and SMAD4G365D gene mutations). Apheresis was performed with no adverse events. One course of the HPL-APC vaccine against KRAS and SMAD4 in combination with oral cyclophosphamide was completed, with a total cell count of 17.7 × 107 cells (mean viable cell rate, 93.9%) without any quality problems. No allergic reactions with DTH reaction of peptides lower than 5 mm were noted. Grade 2 fever reactions, chills, arthralgia, and redness of grade 2 in the vaccination area were observed but were well tolerated. At the second dose, vaginal bleeding due to primary disease infiltration was observed; however, no other vaccine-related adverse reactions or adverse events occurred. Diagnostic imaging after one course was SD; however, at 4 months after one course, a colostomy was performed because of cancer progression.

2.8.3. Case 3 (Patient 5)

A female patient in her 70s was diagnosed with pancreatic cancer (postoperative recurrence stage IV, KRASG12D and TP53E258K gene mutations). Apheresis had already been performed in the WT1 clinical study to obtain materials. During 5FU + irinotecan hydrochloride hydrate salvage therapy, she was administered the HPL-APC vaccine against KRAS and TP53. Neither allergic reaction to DTH nor febrile reactions occurred; however, no adverse reactions or adverse events other than redness of grade 1 were found at the vaccination site. Because the cancer progressed to aortic lymph node metastasis, multiple liver metastases, bile duct obstruction, and portal vein invasion, only bile duct stents and conservative treatment including celiac nerve block were administered without anticancer drugs. After four vaccine lots were administered, she died from the cancer’s further progression.

3. Results

3.1. Preparation of Personalized HPL-APC Vaccine with Cancer Antigen Candidate Peptides

By targeting gene products with pathogenic mutations from cancer genomic information, we predicted the affinity of cancer antigen candidate peptides for HLA typing in each patient. Peptides exhibiting an affinity of IC50 < 500 nM were selected for HLA class I and HLA class II (Table 1 and Table 2). In patients 2 and 3, killer peptides for the activation of CD8+ T cells with a high affinity for HLA class I of IC50 < 50 nM were identified (KRASWT-HLA-A*33:03, IC50 = 11 nM; SMAD4WT-HLA-A*31:01, IC50 = 6 nM). Three enrolled patients had HLA-DRB1*09:01 and were identified to have helper peptides for the activation of CD4+ T cells with a moderate affinity for KRASG12D of IC50 < 500 nM (IC50 = 181 nM, KRASG12D-HLA-DRB1*09:01). The quality of the HPL-APC vaccine harboring candidate peptides targeting personalized cancer antigens is verified in Table 3. HPL-APCs showed high yields (>23%), viability (>94%), and purity (>91%) in three patients. Phenotypic analysis using a flow cytometer revealed HLA-ABC and HLA-DR expression with costimulatory molecules CD40, CD80, and CD86. These results confirmed the validated quality of HPL-APCs containing cancer antigen-specific peptides for use in clinical research.

3.2. Detection of Immune Responses to Personalized Cancer Antigen Candidate Peptides

The interim analysis of immune responses was performed on three enrolled participants, including two who completed six doses per course. Immune responses were monitored by ELISpot assays using personalized cancer antigen candidate peptides (Figure 1). The immune response was evaluated according to the criteria for ELISpot assays [18]. In patient 2, an immune response to HLA-A*33:03-matched KRASWT was detected after six doses of the HPL-APC vaccine (Figure 1b). Immune responses to KRASWT peptide compatible with HLA-DRB1*07:01 and KRASG12D to HLA-DRB1*09:01 were detected (Figure 1c). Furthermore, an immune response was detected using a KRASWT-HLA-DRB1*09:01 that had a sequence corresponding to KRASG12D-HLA-DRB1*09:01. These immune responses attenuated 6 months after completion of HPL-APC vaccination. In patient 3 (Figure 1d–f), an immune response to HLA-A*31:01-matched SMAD4WT peptide was strongly detected after three and six doses of the HPL-APC vaccine (Figure 1e). Positive reactions to SMAD4G365D peptide compatible with HLA-DRB1*04:01 and those of KRASG12D or WT to HLA-DRB1*09:01 were detected (Figure 1f). Six months after one course of the HPL-APC vaccine, the immune response was also diminished. An interim analysis of patient 5 after three doses of the HPL-APC vaccine (Figure 1g,h) showed no immune response. When the immunosuppressive factors of Tregs and M-MDSCs were examined (Figure S1), a marked increase in M-MDSCs was observed in patient 5. On the contrary, patients 2 and 3 did not show quantitative fluctuations in Tregs and M-MDSCs.

3.3. Detection of SMAD4WT HLA-A*31:01-Specific CD8+ Effector Memory T Cells

Immunoinductibility of the HPL-APC vaccine was verified from the viewpoint of immune memory. Memory subsets of CD8+ T and CD4+ T cells in PBMCs during HPL-APC vaccination were analyzed (Figure S2). Patient 2, who was positive in the ELISpot assays, showed an increase in CD8+ T effector memory T cells (TEM) after three and six doses of the HPL-APC vaccine (before vaccination, 19.6%; after vaccination #3, 26.6%; and after vaccination #6, 27.3% in CD8+ T cells). Patient 3 also showed an increase in CD8+ TEM after HPL-APC vaccination (before vaccination, 29.3%; after vaccination #3, 43%; and after vaccination #6, 46.5% in CD8+ T cells). However, a significant decrease in TEM was observed 6 months after six doses (6 months from vaccination #6, 25.2% in patient 3). In addition, changes in cancer antigen candidate peptide-specific CD8+ memory T cells were verified. With antigen candidate peptides of KRASWT-HLA-A*33:03 or SMAD4WT-HLA-A*31:01, CD137 expression in CD8+ T cells was analyzed (Figure 2). Patient 2, after six doses of the HPL-APC vaccine, showed few differences in CD137 expression due to KRASWT-HLA-A*33:03 compared with an HIV-negative peptide (CD137+CD8+ T cells: HIV-HLA-A*33:03, 2.09% vs. KRASWT-HLA-A*33:03, 2.24% in Figure 2a). In patient 3, SMAD4WT-HLA-A*31:01 increased CD137 expression (CD137+CD8+ T cells: HIV-HLA-A*31:01, 0.46%; SMAD4WT-HLA-A*31:01, 1.14% in Figure 2b). TCM in CD137+CD8+ T cells decreased, and an increase in TEM was observed (TCM: before vaccination vs. after vaccination #6, 61.5% vs. 43.8%; TEM: before vaccination vs. after vaccination #6, 38.5% vs. 56.2%).

4. Discussion

This study confirmed the safety and tolerability of the HPL-APC vaccine in two patients with pancreatic cancer (patients 2 and 5) and one with cervical cancer (patient 3) and clarified the immunoinductibility to personalized cancer antigen candidate peptides using ELISpot assays. HLA class I-compatible killer peptides or HLA class II-compliant helper peptides targeting KRAS, SMAD4, or TP53 were predicted from cancer genomic profiles (Table 2), which are pancreatic cancer driver genes frequently identified as pathogenic mutations [23]. Immunoinductibility to KRASWT or G12D peptides for HLA class II was confirmed in patient 2 after six sessions, and that to SMAD4WT peptide for HLA class I and SMAD4G365D peptide for HLA class II in patient 3 were detected after three doses (Figure 1). Although these peptides for the induction of antigen-specific immune response to CD8+ and CD4+ T cells in PBMCs after the HPL-APC vaccine were suggested, future analysis is needed to clarify the responding cells from IFN-γ production using flow cytometry. The activation of cancer antigen-specific CD8+ T and CD4+ T cells is a critical issue for the efficacy of cancer vaccines [7]. KRAS is the most frequently mutated isoform present in 22% of all tumors [24], and KARSG12D is the highest frequency of approximately 35% among them [25]. Immunoinductibility could not be observed for KRASG12D-HLA-A*11:01 (VVGADGVGK, 9-mer peptide) in patient 3 (Figure 1d). However, as the TCR repertoire responding to VVVGADGGGK (10-mer peptide) to HLA-A*11:01 binding KRASG12D peptide was reported [26], the immunoinductibility of HPL-APCs to these peptides must be verified in the future with an additional number of cases. On the contrary, KRASG12D-HLA-DRB1*09:01 helper peptide common to all three cases was chosen (TEYKLVVVGADGVGK, 15-mer peptide in Table 2). Although a TCR repertoire for HLA-DRB1*08:01 binding KRASG12D was reported [27], immunoinduction against KRASG12D-HLA-DRB1*09:01 helper peptide was first identified; however, the TCR repertoire has not yet been analyzed. Interestingly, an immune response was also detected when using a KRASWT-HLA-DRB1*09:01 that had a sequence corresponding to KRASG12D-HLA-DRB1*09:01 (Figure 1c,f). The TCR repertoire analysis induced by the KRASG12D-HLA-DRB1*09:01 helper peptide is expected to reveal specificity and further verify the differences in the antitumor response.
DC vaccines can enhance effector and memory T cell responses, which are important for antitumor immunity and have been used in clinical trials of cancer immunotherapy [28,29]. A Phase I clinical trial of a DC vaccine pulsed with Wilms’ tumor 1 (WT1)-specific MHC class I/II-restricted epitope for pancreatic cancer in combination with chemotherapy was reported. An association was found between WT1/HLA-A*24:02-specific memory cells in CD8+ T cells and overall survival (OS) [12]. WT1-specific central and effector memory CD8+ T cells were observed in cancerous pleural effusion in a patient with long-survival pancreatic cancer after WT1-pulsed DC vaccination [16]. In Phase II trials to evaluate the efficacy of personalized cancer vaccines using HPL-APCs, the detection of antigen-specific memory T cells could be a biomarker of therapeutic prognosis. In patients 2 and 3, CD8+ TEM increased after three doses of the HPL-APC vaccine (Figure S2), and in patient 3, an increase in antigen-specific CD8+ TEM was observed with SMAD4WT-HLA-A*31:01 peptide in a small number of cells due to limited clinical specimens (Figure 2b). Because TEM has strong cytotoxicity with high cytokine production capacity [30], it was consistent with the increase in IFN-γ-producing cells observed with SMAD4WT HLA-A*31:01 peptide (Figure 1e). These results confirmed the ability of HPL-APCs to enhance antigen-specific CD8+ memory T cells. On the contrary, the detection of CD8+ memory T cells to KRASWT-HLA-A*33:03 and that of CD4+ memory T cells responding to KRASWT or G12D or SMAD4G365D helper peptides remain to be elucidated. A method for highly efficient detection of neoantigen-responsive T cells and identification of these TCRs has been reported for personalized cancer immunotherapy [31]. Cancer antigen-specific memory T cells induced by the HPL-APC vaccine using a method that can be easily detected from the peripheral blood via TCRs as an indicator must be identified.
In patients 2 and 3, who completed the HPL-APC vaccination, the acquired immune response was attenuated 6 months after HPL-APC vaccination (Figure 1b,c,e,f), and in patient 2, a significant decrease in TEM was observed (Figure S2). In patients with long-term survival receiving continuous WT1-DC vaccine, acquired WT1-CTLs were maintained [32], and booster effects of additional vaccines were observed [33]. As DC vaccines have been expected to have antitumor effects by priming and boosting [34], the immune memory acquired by the HPL-APC vaccine must be also preserved. Therefore, activating the immune response with additional administrations of the HPL-APC vaccine is expected. As the life span of DC in the lymph nodes is limited to a few days [35,36], the persistence of the HPL-APC vaccine also needs to be monitored in patients to ensure long-lasting efficacy. On the contrary, the immunosuppressive factor M-MDSCs increased, and no immune response was observed in patient 5 (Figure 1g,h). The combination of DC vaccines with chemotherapy is important to create a state favoring expansion of antitumor effector cells, and inhibition, depletion, or both, of Tregs and MDSCs [37]. In a future Phase II study to verify the efficacy of the HPL-APC vaccine, the antitumor immune reactions in combination with anticancer drugs that eliminate or abolish the function of M-MDSCs and immune checkpoint inhibitors that restore exhausted T cells must be optimized [38].

5. Conclusions

This is the first Phase I clinical trial that demonstrates the safety, tolerability, and immunoinductibility of the HPL-APC vaccine with personalized cancer antigen candidate peptides. The HPL-APC vaccine can enhance antigen-specific CD8+ memory T cells. The maintenance of immune memory might require continued HPL-APC vaccination and improvement of the immunosuppressive environment. The feasibility of personalized cancer vaccines using HPL-APCs has been clarified, and a future Phase II trial with optimized dosing and combination therapies would be expected to provide progress in antitumor response.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/cancers15143627/s1, Figure S1: Tregs and M-MDSCs in patients treated with HPL-APC vaccines; Figure S2: Memory T cell subsets in patients treated with HPL-APC vaccines; Table S1: Antibodies for HPL-APC; Table S2: Prediction of HLA-matched HIV peptides; Table S3: Antibodies for Tregs; Table S4: Antibodies for M-MDSCs; Table S5: Antibodies for memory T-cell subsets.

Author Contributions

Conceptualization, writing original draft, and funding acquisition, T.K. and S.S.; resources, S.S.; data curation, M.T.; validation, S.M.; methodology, T.K.J.; investigation, T.K., K.Y., Y.N., H.U., S.T. and S.Y.; writing—review and editing, T.S., H.K. (Hirohito Kobayashi), T.U., H.K. (Hitoshi Kanno), Y.-C.L., S.-S.Y., D.-C.W. and S.S.; project administration, S.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the ALP Institute for Regenerative Medicine Inc.; the Program on Open Innovation Platform with Enterprises, Research Institutes, and Academia of the JST under grant number JPMJOP1722; and JSPS KAKENHI under grant number JP22K07244.

Institutional Review Board Statement

The HPL-APC vaccination study in patients with cancer (approval No. PC009; 18 November 2021) was approved by the Kanazawa Medical University Certificated Committee for Regenerative Medicine (Class III technologies) (approval No. of the committee NB4150006) and was registered as jRCTc040210109 in the Japan Registry of Clinical Trials. All investigations were performed according to the Declaration of Helsinki. An accompanying study of HPL-APC vaccination therapy to monitor the immune response was conducted after receiving appropriate informed consent. This study was approved by the Ethical Committee of Kanazawa Medical University (approval No. G156; 8 June 2020).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available in the article and Supplementary Materials.

Acknowledgments

The authors are grateful to Haruhiko Kawaguchi, Biomedica Solution Inc., for the technical assistance in this work. We thank Manami Nakamura, Department of Regenerative Medicine, Kanazawa Medical University, for her skillful secretarial assistance. We also thank Hiroaki Azehara, Department of Clinical Trial Center, Kanazawa Medical University Hospital, for pre-reviewing the document of jRCTc040210109 and Ryoko Sanada, Department of Clinical Trial Center, Kanazawa Medical University Hospital, for monitoring the clinical trial. We also appreciate Tamaki Takano and Taishi Fujii, Department of Cardiovascular Surgery, Kanazawa Medical University Hospital, for their supporting apheresis sessions.

Conflicts of Interest

S.S., T.K. and M.T. are patent inventors for a method for manufacturing human platelet lysate-inducible interferon (IFN)–DCs (PCT/JP/2021/040505). S.S., T.K. and M.T. evaluated the DCO-K serum-free medium (Nissui Pharmaceutical Co., Ltd.) for the HPL-APC culture. Kanazawa Medical University concludes a collaborative investigation contract with Nissui Pharmaceutical Co., Ltd. The funders had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.

References

  1. Zehir, A.; Benayed, R.; Shah, R.H.; Syed, A.; Middha, S.; Kim, H.R.; Srinivasan, P.; Gao, J.; Chakravarty, D.; Devlin, S.M.; et al. Mutational Landscape of Metastatic Cancer Revealed from Prospective Clinical Sequencing of 10,000 Patients. Nat. Med. 2017, 23, 703–713. [Google Scholar] [CrossRef] [PubMed]
  2. Shemesh, C.S.; Hsu, J.C.; Hosseini, I.; Shen, B.Q.; Rotte, A.; Twomey, P.; Girish, S.; Wu, B. Personalized Cancer Vaccines: Clinical Landscape, Challenges, and Opportunities. Molecular. Therapy 2021, 29, 555–570. [Google Scholar] [CrossRef] [PubMed]
  3. Lin, M.J.; Svensson-Arvelund, J.; Lubitz, G.S.; Marabelle, A.; Melero, I.; Brown, B.D.; Brody, J.D. Cancer Vaccines: The next Immunotherapy Frontier. Nat. Cancer 2022, 3, 911–926. [Google Scholar] [CrossRef]
  4. Tang, L.; Zhang, R.; Zhang, X.; Yang, L. Personalized Neoantigen-Pulsed DC Vaccines: Advances in Clinical Applications. Front. Oncol. 2021, 11, 2969. [Google Scholar] [CrossRef] [PubMed]
  5. Parmiani, G.; Russo, V.; Maccalli, C.; Parolini, D.; Rizzo, N.; Maio, M. Peptide-Based Vaccines for Cancer Therapy. Hum. Vaccin Immunother. 2014, 10, 3175–3178. [Google Scholar] [CrossRef] [Green Version]
  6. Dissanayake, D.; Murakami, K.; Tran, M.D.; Elford, A.R.; Millar, D.G.; Ohashi, P.S. Peptide-Pulsed Dendritic Cells Have Superior Ability to Induce Immune-Mediated Tissue Destruction Compared to Peptide with Adjuvant. PLoS ONE 2014, 9, e92380. [Google Scholar] [CrossRef]
  7. Blass, E.; Ott, P.A. Advances in the Development of Personalized Neoantigen-Based Therapeutic Cancer Vaccines. Nat. Rev. Clin. Oncol. 2021, 18, 215–229. [Google Scholar] [CrossRef]
  8. Palucka, K.; Banchereau, J. Dendritic-Cell-Based Therapeutic Cancer Vaccines. Immunity 2013, 39, 38–48. [Google Scholar] [CrossRef] [Green Version]
  9. Ding, Z.; Li, Q.; Zhang, R.; Xie, L.; Shu, Y.; Gao, S.; Wang, P.; Su, X.; Qin, Y.; Wang, Y.; et al. Personalized Neoantigen Pulsed Dendritic Cell Vaccine for Advanced Lung Cancer. Signal Transduct. Target. Ther. 2021, 6, 26. [Google Scholar] [CrossRef]
  10. Yanagisawa, R.; Koizumi, T.; Koya, T.; Sano, K.; Koido, S.; Nagai, K.; Kobayashi, M.; Okamoto, M.; Sugiyama, H.; Shimodaira, S. WT1-Pulsed Dendritic Cell Vaccine Combined with Chemotherapy for Resected Pancreatic Cancer in a Phase I Study. Anticancer Res. 2018, 38, 2217–2225. [Google Scholar] [CrossRef] [Green Version]
  11. Shimodaira, S.; Yanagisawa, R.; Koya, T.; Hirabayashi, K.; Higuchi, Y.; Sakamoto, T.; Togi, M., Jr.; Kobayashi, T.; Koizumi, T.; Koido, S.; et al. In Vivo Administration of Recombinant Human Granulocyte Colony-Stimulating Factor Increases the Immune Effectiveness of Dendritic Cell-Based Cancer Vaccination. Vaccines 2019, 7, 120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Koido, S.; Homma, S.; Okamoto, M.; Takakura, K.; Mori, M.; Yoshizaki, S.; Tsukinaga, S.; Odahara, S.; Koyama, S.; Imazu, H.; et al. Treatment with Chemotherapy and Dendritic Cells Pulsed with Multiple Wilms’ Tumor 1 (WT1)-Specific MHC Class I/II-Restricted Epitopes for Pancreatic Cancer. Clin. Cancer Res. 2014, 20, 4228–4239. [Google Scholar] [CrossRef] [Green Version]
  13. Koya, T.; Yanagisawa, R.; Higuchi, Y.; Sano, K.; Shimodaira, S. Interferon-α-Inducible Dendritic Cells Matured with OK-432 Exhibit TRAIL and Fas Ligand Pathway-Mediated Killer Activity. Sci. Rep. 2017, 7, srep42145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Watanabe, A.; Togi, M.; Koya, T.; Taniguchi, M.; Sakamoto, T.; Iwabuchi, K.; Kato, T.; Shimodaira, S. Identification of CD56dim Subpopulation Marked with High Expression of GZMB/PRF1/PI-9 in CD56+ Interferon-α-Induced Dendritic Cells. Genes Cells 2021, 26, 313–327. [Google Scholar] [CrossRef] [PubMed]
  15. Date, I.; Koya, T.; Sakamoto, T.; Togi, M.; Kawaguchi, H.; Watanabe, A.; Kato, T.; Shimodaira, S. Interferon-α-Induced Dendritic Cells Generated with Human Platelet Lysate Exhibit Elevated Antigen Presenting Ability to Cytotoxic T Lymphocytes. Vaccines 2021, 9, 10. [Google Scholar] [CrossRef]
  16. Koya, T.; Niida, Y.; Togi, M.; Yoshida, K.; Sakamoto, T.; Ura, H.; Togi, S.; Kato, T.; Yamada, S.; Sugiyama, H.; et al. The Detection of Immunity against WT1 and SMAD4P130L of EpCAM+ Cancer Cells in Malignant Pleural Effusion. Int. J. Mol. Sci. 2022, 23, 12177. [Google Scholar] [CrossRef]
  17. Reynisson, B.; Alvarez, B.; Paul, S.; Peters, B.; Nielsen, M. NetMHCpan-4.1 and NetMHCIIpan-4.0: Improved Predictions of MHC Antigen Presentation by Concurrent Motif Deconvolution and Integration of MS MHC Eluted Ligand Data. Nucleic. Acids Res. 2020, 48, W449–W454. [Google Scholar] [CrossRef]
  18. Higuchi, Y.; Koya, T.; Yuzawa, M.; Yamaoka, N.; Mizuno, Y.; Yoshizawa, K.; Hirabayashi, K.; Kobayashi, T.; Sano, K.; Shimodaira, S. Enzyme-Linked Immunosorbent Spot Assay for the Detection of Wilms’ Tumor 1-Specific T Cells Induced by Dendritic Cell Vaccination. Biomedicines 2015, 3, 304–315. [Google Scholar] [CrossRef] [Green Version]
  19. Liu, W.; Putnam, A.L.; Xu-yu, Z.; Szot, G.L.; Lee, M.R.; Zhu, S.; Gottlieb, P.A.; Kapranov, P.; Gingeras, T.R.; Barbara, B.F.; et al. CD127 Expression Inversely Correlates with FoxP3 and Suppressive Function of Human CD4+ T Reg Cells. J. Exp. Med. 2006, 203, 1701. [Google Scholar] [CrossRef] [Green Version]
  20. Van Wigcheren, G.F.; De Haas, N.; Mulder, T.A.; Horrevorts, S.K.; Bloemendal, M.; Hins-Debree, S.; Mao, Y.; Kiessling, R.; van Herpen, C.M.L.; Flórez-Grau, G.; et al. Cisplatin Inhibits Frequency and Suppressive Activity of Monocytic Myeloid-Derived Suppressor Cells in Cancer Patients. Oncoimmunology 2021, 10, 1935557. [Google Scholar] [CrossRef]
  21. Litjens, N.H.R.; de Wit, E.A.; Baan, C.C.; Betjes, M.G.H. Activation-Induced CD137 Is a Fast Assay for Identification and Multi-Parameter Flow Cytometric Analysis of Alloreactive T Cells. Clin. Exp. Immunol. 2013, 174, 179. [Google Scholar] [CrossRef] [PubMed]
  22. Ponnan, S.M.; Hayes, P.; Fernandez, N.; Thiruvengadam, K.; Pattabiram, S.; Nesakumar, M.; Srinivasan, A.; Kathirvel, S.; Shankar, J.; Goyal, R.; et al. Evaluation of Antiviral T Cell Responses and TSCM Cells in Volunteers Enrolled in a Phase I HIV-1 Subtype C Prophylactic Vaccine Trial in India. PLoS ONE 2020, 15, e229461. [Google Scholar] [CrossRef]
  23. Hu, H.-F.; Ye, Z.; Qin, Y.; Xu, X.-W.; Yu, X.-J.; Zhuo, Q.-F.; Ji, S.-R. Mutations in Key Driver Genes of Pancreatic Cancer: Molecularly Targeted Therapies and Other Clinical Implications. Acta Pharmacol. Sinica 2021, 42, 1725–1741. [Google Scholar] [CrossRef] [PubMed]
  24. Prior, I.A.; Lewis, P.D.; Mattos, C. A Comprehensive Survey of Ras Mutations in Cancer. Cancer Res. 2012, 72, 2457. [Google Scholar] [CrossRef] [Green Version]
  25. Stolze, B.; Reinhart, S.; Bulllinger, L.; Fröhling, S.; Scholl, C. Comparative Analysis of KRAS Codon 12, 13, 18, 61, and 117 Mutations Using Human MCF10A Isogenic Cell Lines. Sci. Rep. 2015, 5, 8535. [Google Scholar] [CrossRef] [Green Version]
  26. Poole, A.; Karuppiah, V.; Hartt, A.; Haidar, J.N.; Moureau, S.; Dobrzycki, T.; Hayes, C.; Rowley, C.; Dias, J.; Harper, S.; et al. Therapeutic High Affinity T Cell Receptor Targeting a KRASG12D Cancer Neoantigen. Nat. Commun. 2022, 13, 5333. [Google Scholar] [CrossRef]
  27. Cafri, G.; Yossef, R.; Pasetto, A.; Deniger, D.C.; Lu, Y.C.; Parkhurst, M.; Gartner, J.J.; Jia, L.; Ray, S.; Ngo, L.T.; et al. Memory T Cells Targeting Oncogenic Mutations Detected in Peripheral Blood of Epithelial Cancer Patients. Nat. Commun. 2019, 10, 449. [Google Scholar] [CrossRef] [Green Version]
  28. Saxena, M.; Bhardwaj, N. Re-Emergence of Dendritic Cell Vaccines for Cancer Treatment. Trends Cancer 2018, 4, 119–137. [Google Scholar] [CrossRef]
  29. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic Cells in Cancer Immunology and Immunotherapy. Nat. Rev. Immunol. 2019, 20, 7–24. [Google Scholar] [CrossRef]
  30. Gattinoni, L.; Speiser, D.E.; Lichterfeld, M.; Bonini, C. T Memory Stem Cells in Health and Disease. Nat. Med. 2017, 23, 18. [Google Scholar] [CrossRef]
  31. Yossef, R.; Tran, E.; Deniger, D.C.; Gros, A.; Pasetto, A.; Parkhurst, M.R.; Gartner, J.J.; Prickett, T.D.; Cafri, G.; Robbins, P.F.; et al. Enhanced Detection of Neoantigen-Reactive T Cells Targeting Unique and Shared Oncogenes for Personalized Cancer Immunotherapy. JCI Insight 2018, 3, e122467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Shimodaira, S.; Sano, K.; Hirabayashi, K.; Koya, T.; Higuchi, Y.; Mizuno, Y.; Yamaoka, N.; Yuzawa, M.; Kobayashi, T.; Ito, K.; et al. Dendritic Cell-Based Adjuvant Vaccination Targeting Wilms’ Tumor 1 in Patients with Advanced Colorectal Cancer. Vaccines 2015, 3, 1004–1018. [Google Scholar] [CrossRef] [Green Version]
  33. Shimodaira, S.; Kobayashi, T.; Hirabayashi, K.; Horiuchi, K.; Koya, T.; Mizuno, Y.; Yamaoka, N.; Yuzawa, M.; Ishikawa, S.; Higuchi, Y.; et al. Induction of Antigen-Specific Cytotoxic T Lymphocytes by Chemoradiotherapy in Patients Receiving Wilms’ Tumor 1-Targetted Dendritic Cell Vaccinations for Pancreatic Cancer. OMICS J. Radiol. 2015, 4, 1–6. [Google Scholar] [CrossRef] [Green Version]
  34. Harari, A.; Graciotti, M.; Bassani-Sternberg, M.; Kandalaft, L.E. Antitumour Dendritic Cell Vaccination in a Priming and Boosting Approach. Nat. Rev. Drug Discov. 2020, 19, 635–652. [Google Scholar] [CrossRef] [PubMed]
  35. Kamath, A.T.; Henri, S.; Battye, F.; Tough, D.F.; Shortman, K. Developmental Kinetics and Lifespan of Dendritic Cells in Mouse Lymphoid Organs. Blood 2002, 100, 1734–1741. [Google Scholar] [CrossRef]
  36. Ingulli, E.; Mondino, A.; Khoruts, A.; Jenkins, M.K. In Vivo Detection of Dendritic Cell Antigen Presentation to CD4(+) T Cells. J Exp Med 1997, 185, 2133–2141. [Google Scholar] [CrossRef]
  37. Anguille, S.; Smits, E.L.; Lion, E.; Van Tendeloo, V.F.; Berneman, Z.N. Clinical Use of Dendritic Cells for Cancer Therapy. Lancet Oncol 2014, 15, e257–e267. [Google Scholar] [CrossRef]
  38. Zhao, Y.; Du, J.; Shen, X. Targeting Myeloid-Derived Suppressor Cells in Tumor Immunotherapy: Current, Future and Beyond. Front. Immunol. 2023, 14, 1157537. [Google Scholar] [CrossRef]
Figure 1. Detection of immune responses to cancer antigen candidate peptides. (a) Detection of IFN-γ-producing cells by ELISpot assays using PBMCs and KRASWT-HLA-A*24:02 of six doses (after one course) of the HPL-APC vaccine in patient 2. HIV peptides compatible with each patient’s HLA typing was used as negative controls. Immune response was observed before and at three and six doses after HPL-APC vaccination and 6 months after completion of one course. (b) ELISpot assays using PBMCs and KRASWT-HLA-A*33:03 after one course of HPL-APC vaccination in patient 2. (c) Helper peptides targeting KRASWT or G12D compatible with HLA class II in patient 2. (d) KRASG12D-HLA-A*11:01 in patient 3. (e) SMAD4WT-HLA-A*31:01 in patient 3. (f) HLA class II-compliant SMAD4G365D, KRASWT or G12D targeted helper peptides in patient 3. (g) KRASG12D-HLA-A*02:06 in patient 5. (h) Helper peptide targeting HLA class II-compatible TP53E258K, KRASWT or G12D in patient 5.
Figure 1. Detection of immune responses to cancer antigen candidate peptides. (a) Detection of IFN-γ-producing cells by ELISpot assays using PBMCs and KRASWT-HLA-A*24:02 of six doses (after one course) of the HPL-APC vaccine in patient 2. HIV peptides compatible with each patient’s HLA typing was used as negative controls. Immune response was observed before and at three and six doses after HPL-APC vaccination and 6 months after completion of one course. (b) ELISpot assays using PBMCs and KRASWT-HLA-A*33:03 after one course of HPL-APC vaccination in patient 2. (c) Helper peptides targeting KRASWT or G12D compatible with HLA class II in patient 2. (d) KRASG12D-HLA-A*11:01 in patient 3. (e) SMAD4WT-HLA-A*31:01 in patient 3. (f) HLA class II-compliant SMAD4G365D, KRASWT or G12D targeted helper peptides in patient 3. (g) KRASG12D-HLA-A*02:06 in patient 5. (h) Helper peptide targeting HLA class II-compatible TP53E258K, KRASWT or G12D in patient 5.
Cancers 15 03627 g001aCancers 15 03627 g001bCancers 15 03627 g001cCancers 15 03627 g001dCancers 15 03627 g001e
Figure 2. CD137 expression on CD8+ T cells stimulated with KRASWT-HLA-A*33:03 or SMAD4WT-HLA-A*31:01-compatible peptides. (a) CD137-positive fraction in CD8+ T cells stimulated with KRASWT-HLA-A*33:03 peptide in patient 2 were gated, and the memory subset was further analyzed. HLA-A*33:03-compatible HIV peptide was used as a negative control. (b) Detection of a memory subset of CD137+CD8+ T cells by SMAD4WT-HLA-A*31:01 stimulation in patient 3. CD45RO+CCR7+, central memory T cells (TCM); CD45ROCCR7+, naïve T cells (TN); CD45RO+CCR7, effector memory T cells (TEM); and CD45ROCCR7, terminal effector T cells (TTE).
Figure 2. CD137 expression on CD8+ T cells stimulated with KRASWT-HLA-A*33:03 or SMAD4WT-HLA-A*31:01-compatible peptides. (a) CD137-positive fraction in CD8+ T cells stimulated with KRASWT-HLA-A*33:03 peptide in patient 2 were gated, and the memory subset was further analyzed. HLA-A*33:03-compatible HIV peptide was used as a negative control. (b) Detection of a memory subset of CD137+CD8+ T cells by SMAD4WT-HLA-A*31:01 stimulation in patient 3. CD45RO+CCR7+, central memory T cells (TCM); CD45ROCCR7+, naïve T cells (TN); CD45RO+CCR7, effector memory T cells (TEM); and CD45ROCCR7, terminal effector T cells (TTE).
Cancers 15 03627 g002
Table 1. Cancer genomic profile of patients treated with the HPL-APC vaccine.
Table 1. Cancer genomic profile of patients treated with the HPL-APC vaccine.
Patient No.Age (Years)SexDiseaseCancer Genomic Profiles
257Mpancreatic cancerKRAS G12D
348Fcervical cancerKRAS G12D, SMAD4 G365D
575Fpancreatic cancerKRAS G12D, TP53 E258K
†, evaluated as a pathogenic mutation by FoundationOne CDx.
Table 2. Prediction of cancer antigen candidate peptides for HPL-APC vaccines.
Table 2. Prediction of cancer antigen candidate peptides for HPL-APC vaccines.
Patient No.HLA TypeSequencePeptide NameAffinity to HLA
(IC50 nM)
2HLA-A*24:02QYMRTGEGFKRASWT-HLA-A*24:0237
HLA-A*33:03SFEDIHHYRKRASWT-HLA-A*33:0311
HLA-DRB1*07:01KQAQDLARSYGIPFIKRASWT-HLA-DRB1*07:0140
3HLA-A*11:01VVGADGVGKKRASG12D-HLA-A*11:01172
HLA-A*31:01CVNPYHYERSMAD4WT-HLA-A*31:016
HLA-DRB1*04:01GDRFCLDQLSNVHRTSMAD4G356D-HLA-DRB1*04:01355
5HLA-A*02:06LVVVGADGVKRASG12D-HLA-A*02:06164
HLA-DRB1*01:01TIITLKDSSGNLLGRTP53E258K-HLA-DRB1*01:0171
2, 3, 5HLA-DRB1*09:01TEYKLVVVGADGVGKKRASG12D-HLA-DRB1*09:01181
Table 3. Quality of the HPL-APC vaccine.
Table 3. Quality of the HPL-APC vaccine.
Patient No.Yield (%)Viability (%)Purity (%)Phenotype (%)
CD40CD80CD86HLA-ABCHLA-DR
223949389.950.583.799.961.7
326949681.432.180.2100.086.7
538959176.234.192.699.892.0
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

Koya, T.; Yoshida, K.; Togi, M.; Niida, Y.; Togi, S.; Ura, H.; Mizuta, S.; Kato, T., Jr.; Yamada, S.; Shibata, T.; et al. Clinical Trial on the Safety and Tolerability of Personalized Cancer Vaccines Using Human Platelet Lysate-Induced Antigen-Presenting Cells. Cancers 2023, 15, 3627. https://doi.org/10.3390/cancers15143627

AMA Style

Koya T, Yoshida K, Togi M, Niida Y, Togi S, Ura H, Mizuta S, Kato T Jr., Yamada S, Shibata T, et al. Clinical Trial on the Safety and Tolerability of Personalized Cancer Vaccines Using Human Platelet Lysate-Induced Antigen-Presenting Cells. Cancers. 2023; 15(14):3627. https://doi.org/10.3390/cancers15143627

Chicago/Turabian Style

Koya, Terutsugu, Kenichi Yoshida, Misa Togi, Yo Niida, Sumihito Togi, Hiroki Ura, Shuichi Mizuta, Tomohisa Kato, Jr., Sohsuke Yamada, Takeo Shibata, and et al. 2023. "Clinical Trial on the Safety and Tolerability of Personalized Cancer Vaccines Using Human Platelet Lysate-Induced Antigen-Presenting Cells" Cancers 15, no. 14: 3627. https://doi.org/10.3390/cancers15143627

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