ABSTRACT
Conventional cancer treatments remain insufficient to treat many therapy-resistant tumors.1 Cancer vaccines attempt to overcome this resistance by activating the patient's immune system to eliminate tumor cells without the toxicity of systemic chemotherapy and radiation. Nanoparticles (NPs) are promising as customizable, immunostimulatory carriers to protect and deliver antigen. Although many NP vaccines have been investigated in preclinical settings, a few have advanced into clinical application, and still fewer have demonstrated clinical benefit. This review incorporates observations from NP vaccines that have been evaluated in early phase clinical trials to make recommendations for the next generation of NP-based cancer vaccines.
KEYWORDS: Cancer immunotherapy, clinical trials, cancer vaccines, liposomes, nanoparticles
Introduction
Despite advances in chemotherapy, radiation, and surgical resection, cancer is the second leading cause of death in the United States.1 Cancer immunotherapy has recently produced significant advancements in treatment and is widely recognized as one of the major recent breakthroughs in clinical oncology.2 Adoptive cellular therapies, involving the transfer of ex vivo expanded tumor-reactive lymphocytes, have produced substantial clinical responses in humans, but are complex modalities that require extensive cell preparation ex vivo, resulting in high cost, limited availability, and significant regulatory hurdles to clinical approval.3 Vaccine therapies hold the promise of engendering long-lasting immunity by inducing innate cells including dendritic cells (DCs) to prime and activate T cells in situ. However, to date, cancer vaccines have failed to achieve meaningful and durable responses in the majority of treated patients. Early vaccine development focused on the use of peptides as antigens. However, peptide-based vaccines require immunogenic carriers to activate DCs and are often limited to presentation by specific HLA alleles.4 Nucleic acid vaccines have been proposed as “universal” vaccines that bypass HLA restriction, but these require protection from proteases and entry into cytoplasm and/or nuclear membrane for translation into immunogenic peptides. NPs have been used to protect cargo from degradation, permit entry into cells, and stimulate DC maturation.5 As a result, immunostimulatory NP vaccines have been proposed as “off the shelf” vehicles to deliver antigen directly to antigen presenting cells (APCs) in vivo.6,7 Although preclinical work has produced a multitude of NPs capable of delivering antigen to APCs in vitro and in mouse models, only a few have been approved for investigational use in humans. Those that have been investigated have largely failed to produce significant clinical benefit in late phase clinical trials.8-10
This trend is consistent across NP disciplines. Despite massive increases in the number of articles published each year with the search term “nanoparticle*” in the biomedical literature, a few applications have progressed into clinical evaluation.11 Analysis of the traits that facilitate rapid translation could inform development of nanotherapeutics likely to reach clinical application and ultimately improve patient outcomes.
This review provides a critical analysis of NPs that have been used as tumor vaccines in humans. Clinical and preclinical literature are synthesized to make recommendations on NP engineering and trial design criteria for optimal antitumor efficacy and translation. Application of the insights gained in this review to early NP development may lower regulatory barriers and hasten the development of effective NP vaccines for cancer treatment.
Overview of nanoparticle vaccines investigated in humans
Of 1,564 clinical trials listed on ClinicalTrials.gov with “(nanoparticle OR liposome) AND cancer” as search terms, only 76 utilize delivery of antigen for cancer treatment. Within this group, clinical trial results are reported in PubMed for only nine nanoparticle products (Table 1). Although this sample size is small, the similarities of these NPs may be useful to develop treatments of rapid clinical use. This review begins with a description of each vaccine and its use in clinical trials. Critical analysis of all of these NPs is then used to develop design criteria for future NP vaccines for cancer therapy.
Table 1.
NP-based cancer vaccines for antigen delivery currently being used in humans.
| NP composition | Adjuvant | Antigen | Size (nm) | Disease | Trials | Ref. | |
|---|---|---|---|---|---|---|---|
| Tecemotide (L-BLP25, Stimuvax) | Cholesterol, DMPG, DPPC | BLP25, Monophosphoryl Lipid A | MUC1 | 150–580 | Breast cancer, NSCLC, prostate cancer, CRC | 14 | 8,12,17,19,82 |
| AS15 | Monophosphoryl Lipid A, QS-21 | CpG7909 | MAGE-A3, dHER2 | ND | Metastatic melanoma, NSCLC, breast cancer | 24 | 9,10,20,28,77 |
| Lipovaxin-MM | POPC, 3NTA-DTDA, NiSO4 | IFNγ | Recombinant proteins from MM200 cells | 240 | Stage IV melanoma | 1 | 51 |
| DepoVax | Phosphatidyl choline:Cholesterol 10:1 | Montanide ISA 51, tetanus toxoid | 7 HLA-A2 restricted TAAs or survivin peptides | 120 | Breast, ovarian, and prostate cancer | 6 | 7,29 |
| RNA-LPX | DOTMA, DOPE | None | MAGEA3, tyrosinase, NY-ESO1, TPTE mRNA | 200–400 | Stage IIIB–IV melanoma | 1 | 6 |
| OncoVax - Id/IL-2 | DMPC | IL-2 | Autologous idiotype protein | NF | Follicular lymphoma | 1 | 50 |
| CHP | Pullalan, cholesterol isocyanate | +/- GMCSF or OK-432 | Recombinant MAGE-A4, truncated 146HER2, or NY-ESO-1 protein | 20–50 | Breast, esophageal, stomach, lung cancer | 4 | 31-34,36 |
| ISCOMATRIX | Cholesterol, phospholipid | Quillaia saponin | Recombinant NY-ESO-1 protein, E6/E7 peptide | 40–50 | NY-ESO-1 expressing tumors | 2 | 41,43,44 |
| VLP | Qβ bacteriophage | A-type CpG | Melan-A/MART-1 peptides | 30 | Stage I–IV melanoma | 5 | 47,48 |
Composition, vaccine design, size, disease targeted, and number of trials registered on ClinicalTrials.gov evaluating use in cancer patients for each of the nine NP vaccines. DMPG, dimyristoyl-phosphatidylglycerol; DPPC, dipalmitoyl phosphatidylcholine; DOPE, 1, 2-dioleoyl-sn-glycero-3-phosphoethanolamine; DOTMA,1, 2-di-O-octadecenyl-3-trimethylammonium propane; ND, not determined; POPC, α-palmitoyl-β-oleoyl-phosphatidylcholine; 3NTA-DTDA, 3(nitriilotriacetic acid)-ditetradecylamine.
Tecemotide
Vaccine design
Tecemotide (L-BLP25, StimuVax) is a 150–580 nm liposome composed of cholesterol, dimyristoyl phosphatidylglycerol (DMPG), and dipalmitoyl phosphatidylcholine (DPPC).12,13 Tecemotide delivers MUC1 glycoprotein, which is overexpressed on the apical surfaces of epithelia in many mucosal cancers, in the presence of the immunostimulatory lipid BLP25 and TLR4 agonist Monophosphoryl Lipid A, which is known to induce a shift toward Th1 polarization and CD8+ T-cell response.12,14-16
Clinical studies
Tecemotide is safe, immunogenic, and may provide clinical benefit for subsets of patients with non-small cell lung cancer (NSCLC) (Table 2). Although phase II and III trials failed to demonstrate significant survival benefit for patients with NSCLC, subgroup analyses found significant survival benefit in patients with stage IIIB locoregional disease and patients treated concurrently with cyclophosphamide.8,12 However, a subsequent study to evaluate Tecemotide with concurrent cyclophosphamide treatment was stopped prematurely after a fatal encephalitis that may have been caused by multiple repeated doses of cyclophosphamide and Tecemotide.17 Nevertheless, Tecemotide is currently being investigated in an ongoing multinational phase III trial of colorectal cancer following curative resection of hepatic metastases and as maintenance therapy for patients with phase III NSCLC.18,19
Table 2.
Overview of published clinical trials using NP vaccines.
| Vaccine | Phase | Design | Patients | Control treatment | Endpoint | Result | Immune response (T cell//Ab) | Disease | Combinatorial treatments | Ref. |
|---|---|---|---|---|---|---|---|---|---|---|
| Tecemotide | Phase I | R*, 2 arm | 17 | NUC | Safety/Immunogenicity | pos/pos | neg//neg | Stage IIIB/IV NSCLC | Cyclophosphamide: single IV dose | 13 |
| Phase IIB | R, NB, 2 arm, ITT | 171 | BSC | Median survival/safety | neg/pos | neg//NR | Stage IIIB/IV NSCLC | Cyclophosphamide: single IV dose | 12 | |
| Phase III | R, DB, 2 arm, MITT | 1513 | Placebo | Overall survival | neg | pos//NR | Stage III NSCLC | Cyclophosphamide: single IV dose | 8 | |
| Phase II | R, NB, 2 arm | 34 | NUC | Antigen-specific T-cell response | pos | pos//NR | Multiple myeloma, stage I/II untreated or stable II/III | Cyclophosphamide: single IV dose or metronomic low doses | 17 | |
| Phase II | NR, NB | 22 | NUC | Safety | pos | NR//NR | Unresectable stage III NSCLC | Cyclophosphamide: single IV dose 3 d before treatment | 83 | |
| Phase I/II | NR, NB | 6 | NUC | Safety | pos | NR//NR | Unresectable stage III NSCLC | Cyclophosphamide: single IV dose 3 d before treatment | 19 | |
| Pilot study | NR, NB, single arm | 16 | NUC | Prolonged PSA doubling time, safety | pos/pos | NR//NR | Prostate cancer | Cyclophosphamide: single IV dose 3 d before treatment | 84 | |
| AS15 | Phase IIB | R, NB, 2 arms (AS15 vs AS02) | 75 | AS02 | Survival/Safety | neg/pos | pos//pos | Stage IIIB/IV melanoma | 10 | |
| Phase III | R, DB, 2 arms, T | 2,312 | Placebo | Disease-free survival | neg | //pos | Stage IB, II, IIIA NSCLC | With or without cisplatin | 9,21 | |
| Phase 0 | R, 2 arms | 25 | NUC | Safety/ T-cell response | pos/pos | pos//pos | Stage IIB–IV | 77 | ||
| Phase I/II | 4 arms, NR, NB | 67 | NUC | Safety/T-cell response | pos/pos | pos//pos | Stage IB–III NSCLC | Concurrent with, after, or without cisplatin and vinorelbine | 28 | |
| Phase I | 4 arms, NR, DE | 61 | NUC | Safety/Humoral response | pos/pos | NR//pos | Stage II–III HER2-+ breast cancer | 20 | ||
| Phase I | NR, NB | 12 | NUC | Safety/Humoral immunity | pos/pos | neg//pos | HER2+ breast cancer | Lapatinib | 85 | |
| CHP-MAGE-A4 | Phase I | NR, NB | 20 | NUC | Safety/Immunogenicity | pos/pos | neg//neg | Advanced esophageal, stomach, and lung cancer refractory to surgery | 33 | |
| CHP-NY-ESO-1 | Phase I | NR, NB, 2 doses | 13 | NUC | Safety/humoral immunity | pos/pos | pos//pos | Stage IV esophageal cancer | 34 | |
| Phase I | NR, NB | 9 | NUC | Humoral immunity | pos | ND//pos | Esophageal cancer, prostate cancer, and melanoma | 39 | ||
| Phase I | 2 Arm NR, DE, T | 25 | NUC | Safety/Humoral immunity/overall survival | pos/pos/pos | ND//pos | Esophageal cancer | 40 | ||
| CHP-NY-ESO-1 and CHP-HER2 | Phase I | NR, NB, single arm | 8 | NUC | Safety/Immunogenicity | pos/pos | ND//pos | NY-ESO-1+ HER-2− esophageal cancer | OK-432 | 36 |
| CHP-HER2 | Phase I | NR, NB, 2 arm | 9 | NUC | Safety/TC response | pos/pos | pos//ND | HER2+ cancers | GMCSF or OK-432 | 31 |
| CHP-HER2 | Phase I | NR, NB, 2 arms | 15 | NUC | Safety/Humoral immunity | pos/pos | pos//pos | HER2+ cancers | GMCSF (75 µg × 5 d) | 32 |
| ISCOMATRIX | Phase I | NR, DB, DE,2 arms | 46 | Placebo | Safety/Immunogenicity | pos/pos | pos//pos | NY-ESO-1+ tumors (melanoma, bladder, rectal, breast) | 42 | |
| Phase I | R, B, DE | 31 | Placebo | Safety/Immunogenicity | Pos/pos | pos//pos | Cervical intraepithelial neoplasia | 43 | ||
| Phase II | R, NB, 2 arms | 46 | NUC | Safety/Immunogenicity | pos/pos | pos//pos | Stage IV or unresectable stage III NY-ESO-1 or LAGE-1+ melanoma | Cyclophosphamide: single IV dose | 46,57 | |
| Phase II | R, B, 2 arms | 39 | NUC | T-cell response | pos | pos//pos | Resected melanoma patients vaccinated with NY-ESO-1 ISCOMATRIX | Fowlpox virus containing recombinant full length NY-ESO-1 | 44 | |
| DPX-0907 | Phase I | NR, NB, 2 doses | 23 | NUC | Safety | pos | pos//ND | Breast, ovarian, and prostate cancer | 7 | |
| DPX-Survivin | Phase I | NR, NB, 3 arms | 19 | NUC | Safety/Immunogenicity | pos/pos | pos//ND | Ovarian cancer in 1st or 2nd remission | Concurrent metronomic cyclophosphamide or single dose | 29 |
| OncoVAX-Id/IL-2 | Phase I | NR, NB, 1 arm | 10 | NUC | Safety/Immunogenicity | pos/pos | pos//neg | Follicular lymphoma | >6 cycles of PACE regimen completed before treatment | 50 |
| Lipo-MERIT | Phase I | NR, NB, DE | 3 | NUC | Safety | ND | pos//ND | Stage IIIB–IV melanoma | 6 | |
| VLP | Phase I/II | R/NR, NB, 4 arms | 22 | NUC | Safety/Immunogenicity | pos | pos//ND | Stage II–IV melanoma | 47 | |
| Phase IIa | 4 arms | 21 | NUC | Immunogenicity/Safety | pos/pos | pos//ND | Stage III–IV melanoma | Imiquimod cream or IFA | 48 |
Design and outcome information for each nanoparticle vaccine with published clinical trial results. “Results” represents the outcome of the primary endpoint for the general treated population. Therefore, a negative result does not preclude effect in subgroup analysis.
BSC, best supportive care; DB, double blind; DE, dose escalation; ITT, intention to treat analysis; MITT, modified ITT; NB, nonblinded; ND, not determined; neg, no statistically significant improvement or an increase in less than 50% of treated patients if no control cohort is available for comparison; NR, non-randomized; NUC, no untreated control; pos, statistically significant improvement or increase in greater than 50% of patients if no control cohort is available for comparison; R, randomized; T, only treated patients considered in statistical analysis. “T-cell response” includes both flow cytometry to quantify numbers of increased antigen-specific T cells and ELISAs of peripheral blood to evaluate IFNγrelease after restimulation with tumor antigen.
AS15
Vaccine design
AS15 is an “immunostimulatory lipid” that is co-delivered with Melanoma Associated Antigen 3 (MAGE-A3) protein as a treatment of metastatic melanoma and NSCLC or recombinant HER2 protein (dHER2) as treatment of breast cancer.10,20,21 MAGE-A3 is a tumor-associated cancer/testes antigen that is expressed in 24% of patients with NSCLC and is associated with poor prognosis.22,23 Human Epidermal Growth Factor Receptor 2 (HER2) is overexpressed on surfaces of some malignant breast cancer cells.24 AS15 contains the saponin QS21 that activates the inflammasome in APCs when co-administered with Monophosphoryl Lipid A.25,26 AS15 has been combined with CpG analogs to further potentiate response.10 CpG7909 is of particular interest as a TLR9 agonist used to stimulate plasmacytoid DCs to produce robust CD8+ immunity.10,27
Clinical trials
AS15 trended toward benefit to overall survival in Phase II and III studies and produced complete responses in 3/36 patients with stage III or IV metastatic melanoma.9,10 Treated patients demonstrated increased antibody production and CD4+ T-cell activation.10,21 AS15 induced significantly increased CD4+ and CD8+ T-cell responses in patients with unresected stage IB-III MAGE-A3 positive NSCLC, suggesting that tumor tissue may serve to augment immunologic response.28 AS15 also demonstrated safety, antibody responses, and a trend toward increased disease free survival using HER2 peptide as antigen in a Phase I trial.20 However, a large, randomized, double blind Phase III trial to evaluate use of AS15 in NSLC patients failed to improve disease-free survival.9,21
DepoVax
Vaccine design
DepoVax (DPX) is a 120-nm liposomal vaccine composed of a 10:1 mix of Phosphatidyl Choline:Cholesterol that is used in multiple formulations.7,29 DPX-0907 delivers seven tumor-associated antigens (TAAs) that are expressed by MHC I on an ovarian cancer cell line.7 DPX-Survivac delivers survivin, an inhibitor of apoptosis protein that is overexpressed in a variety of cancers and expressed on normal tissues at only low levels.30 Both DPX vaccines are co-delivered with tetanus toxoid, a proprietary TLR9 agonist, and Montanide ISA 51, which is a Freund's Adjuvant called Montanide ISA 51, and a proprietary TLR9 agonist.7,29
Clinical trials
DPX-0907 induced persistent antigen-specific T-cell responses in 39% of patients with breast, ovarian, or prostate cancer in a Phase I trial.7 Interestingly, individual patients responded to unique sets of antigens.7 A trial using DPX-Survivac to treat patients with ovarian cancer included a metronomic cyclophosphamide regimen to selectively eliminate regulatory T cells (TReg) while allowing the proliferation of effector cells targeted against multiple survivin peptides.29 Although no patients demonstrated evidence of objective clinical response, all patients in this trial generated antigen-specific CD8+ T-cell responses in peripheral blood after three subcutaneous vaccines.29
CHP
Vaccine design
Cholesteryl pullalan (CHP) nanogels deliver peptides of MAGE-A4 and NY-ESO-1 or recombinant proteins of HER2.31-34 Preclinical models suggest that these “immunologically stealth” particles act mainly via medullary macrophages in lymph nodes after subcutaneous injection.35 CHP-HER2 NPs are sometimes combined with GMCSF or OK-432, a killed, low virulence strain of Streptococcus pyogenese that activates TLR-4.31,36
Clinical trials
CHP-NY-ESO-1 demonstrated widespread T-cell and antibody responses that correlated with PSA stabilization in prostate cancer patients and tumor regression in esophageal cancer and melanoma patients whose tumors expressed NY-ESO-1 protein.37-39 Subsequent studies found enhanced survival benefit in esophageal cancer patients who received 200 µg dose compared with 100 µg.40 CHP-MAGE-A4 induced substantial antibody responses that correlated with prolonged progression free and overall survival among esophageal cancer patients whose tumors highly expressed MAGE-A4 protein.33 However, treatment also resulted in increased prevalence of CD4+FoxP3hiCD45RA− regulatory T cells in peripheral blood.33 Likewise, CHP-HER2 induced antibody responses in almost all patients but was associated with loss of tumor antigen, presence of CD68+ macrophages, and reduced CD4+ and CD8+ infiltration in tumor after treatment.32,34
ISCOMATRIX
Vaccine design
ISCOMATRIX is a 50-nm particle containing saponin, cholesterol, and phospholipid designed to increase cross-presentation of MHC class I restricted epitopes.41 ISCOMATRIX has been used to deliver recombinant E6/E7 peptide to treat cervical intraepithelial neoplasia (CIN) or NY-ESO-1 protein as treatment of NY-ESO-1 positive melanoma, bladder, rectal, and breast cancer.42,43
Clinical trials
ISCOMATRIX originally garnered enthusiasm after a Phase I trial demonstrated dose-dependent antibody responses and reduced risk of relapse for patients with NY-ESO-1 positive tumors.42 However, Phase II trials failed to demonstrate CD8+ T-cell responses in patients with advanced disease and instead revealed production of an antigen-specific TReg population in vaccinated patients.44-46 A current search on ClinicalTrials.gov revealed two terminated studies and one suspended study with this drug for cancer treatment (NCT01341496, NCT02054104, and NCT01258868).
VLPs
Vaccine design
Virus-like nanoparticles (VLPs), also known as CYT004-MelQbG10, are generated from the coat protein of bacteriophage Qβ and loaded with the CpG “G10.”47 This A-type CpG is known to stimulate IFN-α production by plasmacytoid dendritic cells (pDCs) to a greater degree than the B-Type CpG found in Montanide via stimulation of TLR9, but was previously unable to be used in humans due to rapid degradation by DNAse.47 VLPs deliver peptides of melanoma-associated antigens Melan-A/MART-1.47,48
Clinical trials
A randomized Phase I trial found that VLPs safely generate T-cell responses in most patients with Stage II–IV melanoma.47 Comparisons of multiple vaccination routes demonstrated increased production of antigen-specific T cells and effector memory and central memory subsets of antigen-specific T cells with intradermal or subcutaneous injection compared with intranodal injection.48
OncoVAX-Id/IL-2
Vaccine design
OncoVAX-Id/IL-2 was designed to generate more robust responses and a more homogeneous product after a protein conjugate vaccine demonstrated efficacy in follicular lymphoma patients in complete remission.49 OncoVAX-Id/IL-2 incorporates protein for autologous lymphoma idiotypes and recombinant IL-2 as adjuvant inside a dimyristoylphosphatidylcholine (DMPC) liposome.50
Clinical trials
A Phase I trial with 10 patients found that OncoVAX-Id/IL-2 safely induced T-cell responses in 100% of patients and antibody responses in 40% of follicular lymphoma patients in clinical remission.50
Lipovaxin
Vaccine design
Lipovaxin is a 240-nm liposomal NP that delivers recombinant protein antigen and IFNγ intravenously (IV) to treat Stage IV melanoma.51 This vaccine is composed of membrane vesicles from MM200 melanoma cells fused to POPC (α-palmitoyl-β-oleoyl-phosphatidylcholine) liposomes for stability and an anti-DC SIGN (DC-specific intercellular adhesion molecule 3 grabbing nonintegrin) immunoglobulin single variable domain to increase DC uptake.51
Clinical trials
A Phase I trial evaluating Lipovaxin was completed in 2012, but no results have been reported (NCT01052142).54
Lipo-MERIT
Vaccine design
Lipo-MERIT is a 200-nm liposomal vaccine that delivers mRNA encoding four common melanoma antigens (NY-ESO-1, MAGE-A3, tyrosinase, and TPTE) to APCs.6 The 200–400 nm particle contains DOTMA (1,2-di-O-octadecenyl-3-trimethylammonium propane) and DOPE (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine) and is targeted to lymphoid organs after systemic injection by modifying lipid:RNA ratios, so that the net particle charge is negative.6
Clinical trials
Lipo-MERIT is currently in a Phase I trial to assess safety and dosing for patients with stage IV melanoma (NCT02410733). Although final results have not been reported, the first three patients developed flu-like symptoms, released IFN-α and IP-10, and increased levels of antigen-specific CD4+ or CD8+ T cells.6
Critical analysis of clinical outcomes
These trials and NPs have many similarities. While all studies report either antibody or T-cell response, no trials demonstrated statistically significant survival benefit in primary analysis.8-10 The lack of clinical benefit despite immune outputs indicates that responses are either of the wrong character, insufficient magnitude, or too brief in duration. These insufficient outcomes may result from poor biodistribution or immunosuppression via tumor-induced mechanisms such as immune checkpoint activation. Understanding the impact of particle characteristics, combination therapies, and study design may inform development of successful therapeutics.
Study design
Outcome variable
Primary endpoints for each trial are included in Table 2. Although T cells are intended mediators of antitumor responses, their activation and proliferation is often not a direct predictor of clinical response. As an example, clinical responders to AS15 did not have increased rates of CD4+ or CD8+ T-cell responses.10 Similarly, although proliferative response of antigen-specific T cells in the blood correlated with increased median survival among all patients vaccinated with Tecemotide (n = 88), the subset of patients that showed a survival benefit compared with controls (n = 35) included only two patients with proliferative T-cell responses.12 Both analyses are limited by sample size and tepid responses to treatment, but this combined evidence may indicate that detectable levels of antigen-specific T cells are neither necessary nor sufficient to produce clinical response to vaccination.
Another possibility is that T-cell characterization as CD4+ and CD8+ is insufficient to predict outcome. Instead, a tipping of the balance between effector, memory, and regulatory T cells may be necessary for rational vaccine design to induce clinical antitumor immunity.55 This effect was evaluated in Tecemotide, in which immunologic responders generated reduced effector and effector memory CD4+ T cells.17 In another approach, Berinstein and colleagues differentiated T cells functionally and found that DepoVax induced increased production of polyfunctional central and effector memory T cells.7,29 Both analyses were applied to evaluate responses to VLPs, which induce production of multifunctional effector memory and central memory antigen-specific T cells.47
Furthermore, regulatory responses may counteract effector T cells. CHP-MAGE-A4 nanogels and NY-ESO-1/ISCOMATRIX both induced significant formation of regulatory T cells in many patients.33,45 In one patient with metastatic melanoma, CHP-NY-ESO-1 induced systemic humoral and cellular responses but also high levels of CD4+CD25+Foxp3+ regulatory T cells and macrophages at the tumor sites.56 This patient ultimately succumbed to metastatic pulmonary infiltrates.56 Likewise, histologic analysis of tumor from a small number of patients treated with CHP-HER2 revealed increased CD68+ macrophages, reduced CD4+ and CD8+ infiltration, and loss of antigen expression after treatment.34 Interestingly, clinical responses to CHP-MAGE-A4 correlated with antibody production.33
Studies to corroborate the relationship between these immunological classifications and clinical outcomes will likely demonstrate roles for CD8+ T-cell phenotype, regulatory cells, and CD4+ T cells in evaluation of vaccine efficacy.
Combinatorial treatments
Synergy between antigen-specific T-cell activation and chemotherapy has significant implications on future trial design. Combination chemotherapy improved quality or quantity of T cell responses generated by AS15, Tecemotide, ISCOMATRIX, and DepoVax.7,8,10,29,57,58 One proposed explanation is that cyclophosphamide selectively reduces the regulatory T-cell compartment 59 Additionally, the lymphodepletion induced by chemotherapy may induce homeostatic proliferative responses that benefit vaccine-induced T cells. Although somewhat counterintuitive, we also observed enhancing effects of lymphodepletive chemotherapy in vaccinated patients with glioblastoma.60-62 Murine experiments suggest that lymphodepletion is followed by a surge of IL-7 that stimulates lymphocyte proliferation..62,63 In the setting of vaccination, IL-7 can be co-opted to dramatically enhance expansion of tumor-reactive T cells.62,64
Combination therapy with other vaccine approaches may also avoid barriers to efficacy. ISCOMATRIX vaccine demonstrated this principle in showing increased generation of CD8+ T-cell responses when paired with a fowlpox virus vaccine bearing the same antigen.44 Although unexplored for NP-based vaccines, treatment with immune checkpoints may also potentiate vaccine efficacy by reducing the effect of vaccine-generated TRegs and potentially overcoming tumor-induced immunosuppression.
Vaccine design
Antigen selection
Choice of antigen may also explain low treatment efficacy. None of the described trials targeted tumor-specific neoantigens. Instead, these studies generated responses against overexpressed antigens (Her2, survivin, and telomerase), lineage-restricted antigens (tyrosinase), and cancer-testis antigens (MAGE and NY-ESO). The low immunogenicity of overexpressed peptide antigens may explain the presence of antibody and CD4+ T-cell responses without clinical response.3,8,9,12,20,28 Selection of appropriate peptides within a protein antigen may also improve vaccinations. The importance of peptide selection was demonstrated with ISCOMATRIX, in which some epitopes stimulated antitumor T-cell responses to cryptic antigens and others generated functional TRegs specific for tumor antigens.45,65
Many vaccines contained only a single cancer antigen. However, this strategy allows tumor cells to escape immune detection upon loss of the selected vaccine target.66 This “antigen loss” was recorded after use of both VLRs and CHP nanogels.34,48 Efforts to reduce this risk focus on the use of multiple antigens. Individual patients vaccinated with the seven TAA's encoded within DPX-0907 responded to unique subsets of these antigens.7 The same was true of responses to Lipo-MERIT, which went further by using a variety of types of cancer antigens.6 Although sample size is small, each of the three patients responded to a unique set of two of the four antigens in the vaccine.6 However, studies with CHP showed that vaccination with multiple antigens may reduce the magnitude of humoral responses to specific antigens compared with single antigen vaccination.36 Therefore, multiple antigens are likely needed to induce clinically significant responses, but further studies are needed to understand how distinct cancer antigens can be combined effectively.
Antigen form
Peptides bind avidly to MHC molecules but are limited to patients who express certain HLA molecules. Recombinant protein allows presentation of additional MHC I and MHC II epitopes, but requires a carrier that facilitates antigen presentation on both MHC molecules.42 While protein and peptide vaccines have consistently achieved measurable immunologic responses, low clinical efficacy and HLA restriction encouraged the development of nucleic acid based vaccination strategies.6,7,10 Although the bulk of work with nucleic acid vaccines uses DNA, mRNA is attractive for NP delivery because it is innately immunogenic and does not require entry into the nuclear membrane. LipoMERIT was the first human trial to evaluate the systemic administration of mRNA liposomes as vaccines in humans.6 While preliminary data are promising, survival benefit will be necessary to determine the utility of this development.
Adjuvant
Multiple adjuvants have been used to bolster clinical responses. These include IFA, which increased numbers of antigen-specific and effector memory CCR7−CD45RA− T cells in response to VLPs, GMCSF, which accelerated antibody production after vaccination with CHP-HER2, and CPG7909, which trended toward providing survival benefit when combined with AS15.10,32,48 Other adjuvants produced mixed results, such as the TLR7 agonist Imiquimod, which reduced percentages of antigen-specific T cells but increased CCR7+/CD45RA− central memory phenotype and CD127+ T cells.48
Effects of NP composition on DC activation
While insufficient information on characteristics of the available NP constructs makes clinical comparisons difficult, preclinical evidence suggests that NP composition can significantly alter DC response. Many NPs, including Lipo-MERIT, are known to be taken up by DCs via multiple endocytosis pathways including macropinocytosis, but definitive analysis of the best pathway for subsequent antigen presentation remains elusive.6,67 Studies in other cell types have suggested that addition of cationic lipids to otherwise neutral NPs changed the intracellular destination of NPs.68 Notwithstanding the lack of mechanistic explanation, cationic NPs have generally emerged as more effective than neutral or anionic NPs for stimulating DC activation in vitro.69,70 Soema et al. recently explored multiple variables with a Design of Experiment model to determine that optimal DC activation occurs at a NP charge of +30 mV.71 Other groups found similar benefits of cationic NPs with diverse starting materials using elegant screening methods in vitro.72,73 However, in vitro studies are likely not sufficient to draw conclusions on translational potential since the characteristics that determine particle uptake are also thought to govern NP localization after injection.
Delivery method
Effect of composition on localization
Clinical trials included subcutaneous (SC), intramuscular (IM), intradermal (ID), and intravenous (IV) vaccine administration (Table 3). Differences in trial outcomes may be reflected by the different cell populations targeted with each method. ID and SC administration are thought to lead to NP uptake by immature DC subsets including Langerhans cells and CD14+ dermal DCs that migrate to lymph nodes and stimulate CD8+ T-cell responses.74 However, CD8+ T-cell response may be more readily generated with transfection of multiple DC subtypes including those deep within lymph nodes.75,76 IV vaccines, on the other hand, are thought to circulate before transfecting APCs in lymphoid organs.6 Consideration of the DC subsets that will be activated by each injection route should influence selection of injection method. Although CD8+ T-cell responses were seen with IM administration of AS15, a recently reported human study of AS15 found that while no routes induced robust CD8+ T-cell responses, ID and SC administration tended toward increased CD4+ T-cell responses compared with IM vaccination.77 Studies with VLP corroborate a lack of difference between ID and SC injection and demonstrate reduced antigen-specific, effector, and memory T cells after direct intranodal injection.48 Other injection methods have been explored preclinically, including intranasal vaccination, which has been shown to generate robust DC accumulation in pulmonary LNs and produce CD8+ T-cell and antibody responses in peripheral blood in mice.78 Future human studies should consider all available forms of vaccination and select the most optimal based on NP design and desired response.
Table 3.
Injection methods and vaccination schedule for NP vaccines in clinical trials.
| NP | Vaccination schedule | Vaccination method | References |
|---|---|---|---|
| Tecemotide | 8 doses weekly, then continued doses every 6 weeks until progression | 4 SC sites | 8,12,17,83,84 |
| Weeks 0, 2, 5, and 9 | Upper arm, anterolateral thigh | 13 | |
| > 4 weekly doses | 4 SC sites | 19 | |
| AS15 | Biweekly for 12 weeks, triweekly for 18 weeks, every 6 weeks for 4 mo, quarterly for 1 y, and biannually for 3 y | IM | 10 |
| 13 injections in 27 mo | IM | 9 | |
| 8 doses triweekly | IM | 28 | |
| 6 doses over 14 weeks | IM or ID/SC | 20,77 | |
| Biweekly for 12 weeks | IM | 85 | |
| DepoVax | 3 doses triweekly | SC | 7, 29 |
| LipoVaxin | ND | IV | 54 |
| Lipo-MERIT | Weekly vaccinations with increasing dose | IV | 6 |
| OncoVAX - Id/IL-2 | Months 0, 1, 2, 3, and 5 | 4 SC sites | 50 |
| CHP-NY-ESO-1 or CHP-HER2 | 6 doses biweekly | SC | 31,33,36,40 |
| 14 biweekly doses | SC | 34 | |
| 3 doses biweekly | SC | 31 | |
| 4 doses biweekly, then regularly up to 12 vaccinations | SC | 32,39 | |
| ISCOMATRIX | 3–6 doses monthly | IM | 42,44,51 |
| Weeks 1, 3, 5, 9, then 3 monthly, then 1 every 12 weeks until progression | IM | 46,57 | |
| 1–3 doses weekly | IM | 43,57 | |
| VLP | Weekly or daily for 14 weeks | SC or IM | 47 |
| 3 weekly, then 3 monthly | SC, ID, or intranodal | 48 |
ID, intradermal; IM, intramuscular; SC, subcutaneous; triweekly, every third week.
Once a delivery method is decided, simple modifications can be made to enhance delivery to target organs. None of the clinical studies track NP fate, but lessons from preclinical work may be extrapolated to understand results of clinical trials. Although cationic liposomes increase DC function and activation in culture, positively charged particles accumulate in lungs in murine models after IV injection, precluding immune responses in lymphoid organs.6 LipoMERIT avoids this problem by increasing the concentration of negatively charged RNA molecules, effectively reducing net particle charge to negative values.6 The same principle applies to subcutaneous administration in murine models, where reducing surface charge by addition of PEG to cationic liposomes dramatically increases their uptake in lymph nodes.79 However, changing route of administration may avoid this dilemma. After intranasal administration, cationic NPs are more readily taken up by nasopharyngeal-associated lymphoreticular tissue (NALT) DCs than anionic NPs.80 Therefore, NP charge should be selected based on the desired injection method, with neutral NPs for ID/SC administration, cationic NPs for nasal administration, and cationic NPs with net negative charge for systemic administration.
NP size also dictates localization within lymph nodes.75 However, pleiotropic effects make guidelines difficult. As an example, in a study using poly(lactic-co-glycolic acid) (PLGA) NPs to deliver antigen to DCs, 40 nm NPs migrated to lymph nodes more effectively after SC administration, but 100 and 200 nm NPs each delivered more DNA per NP.81 However, many treatments used in humans appear to be in the range of 100 nm to 1 µm, which is larger than recommended for optimal migration to LN after SC injection in murine models.81 Analysis of the effects of size and charge in humans will require consistent reporting of size and charge in ongoing trials.
Despite the clear importance of particle localization, none of the evaluated trials track particle fate in humans. Future particle development will require adoption of clinically relevant methods to determine effects of particle characteristics on particle localization in humans.
Future trial design
Although promising, nanoparticle vaccines face many challenges in practice. Tolerance induced by long-term exposure to tumor antigens and the immunoregulatory tumor microenvironment may contribute significantly to this underwhelming clinical benefit. However, combination with immune checkpoint inhibitors may overcome these barriers. By “turning off the brakes” on the immune system, anti-CTLA-4, anti-PDL-1, and anti-PD-1 antibodies prevent tolerance induction and allow unfettered T-cell activation. Although combination therapies are more challenging to evaluate in clinical trials, the clinical approval and acceptance of ipilimumab (anti-CTLA-4), pembrolizumab (anti-PD-1), and nivolumab (anti-PD-1) for a variety of advanced malignancies has paved the way for the initiation of a myriad of combinatorial therapeutic trials. Therefore, to unleash the full therapeutic potential of nanoparticle vaccines, future trials will likely be designed in combination with agents that remodel the intratumoral microenvironment.
Conclusions
NP vaccines that have been investigated in humans have many similarities. Most contain components that were approved for use in other products and are co-delivered with previously approved immunostimulatory adjuvants. Analysis of these early studies indicates the need for multiple, tumor-specific antigens, rational selection of combinatorial treatments, and NP design specific to route of administration. Future studies should consider the impact of NP characteristics on particle localization, determine immune correlates that accurately predict clinical outcomes, and consider combination therapy with immune checkpoint inhibitors. Consistent reporting of NP characteristics and immunological outcome variables in human and preclinical trials would facilitate this work and inform a generation of NP vaccines that provide significant survival advantage to patients with malignant tumors.
Disclosure of potential conflicts of interest
DAM receives research funding from Immunomic Technologies, Inc., a cancer therapeutics company. AJG and EJS declare no conflicts of interest.
Funding
This work was supported in part by the National Institutes of Health grants R01CA175517 (DAM) and R01CA195563 (DAM).
References
- 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016; 66:7-30; https://doi.org/ 10.3322/caac.21332 [DOI] [PubMed] [Google Scholar]
- 2.Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013; 342:1432-3; PMID:24357284; https://doi.org/ 10.1126/science.342.6165.1432 [DOI] [PubMed] [Google Scholar]
- 3.Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR et al.. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011; 17:4550-7; PMID:21498393; https://doi.org/ 10.1158/1078-0432.CCR-11-0116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Gulukota K, DeLisi C. HLA allele selection for designing peptide vaccines. Genet Anal 1996; 13:81-6; PMID:8931995; https://doi.org/ 10.1016/1050-3862(95)00156-5 [DOI] [PubMed] [Google Scholar]
- 5.Cobaleda-Siles M, Henriksen-Lacey M, Ruiz de Angulo A, Bernecker A, Gomez Vallejo V, Szczupak B, Llop J, Pastor G, Plaza-Garcia S, Jauregui-Osoro M et al.. An iron oxide nanocarrier for dsRNA to target lymph nodes and strongly activate cells of the immune system. Small 2014; 10:5054-67; PMID:25123704; https://doi.org/ 10.1002/smll.201470156 [DOI] [PubMed] [Google Scholar]
- 6.Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Meng M, Fritz D, Vascotto F, Hefesha H et al.. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 2016; 534:396-401; PMID:27281205; https://doi.org/ 10.1038/nature18300 [DOI] [PubMed] [Google Scholar]
- 7.Berinstein NL, Karkada M, Morse MA, Nemunaitis JJ, Chatta G, Kaufman H, Odunsi K, Nigam R, Sammatur L, MacDonald LD et al.. First-in-man application of a novel therapeutic cancer vaccine formulation with the capacity to induce multi-functional T cell responses in ovarian, breast and prostate cancer patients. J Transl Med 2012; 10:156; PMID:22862954; https://doi.org/ 10.1186/1479-5876-10-156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Butts C, Socinski MA, Mitchell PL, Thatcher N, Havel L, Krzakowski M, Nawrocki S, Ciuleanu T-E, Bosquée L, Trigo JM et al.. Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): a randomised, double-blind, phase 3 trial. Lancet Oncol 2014; 15:59-68; PMID:24331154; https://doi.org/ 10.1016/S1470-2045(13)70510-2 [DOI] [PubMed] [Google Scholar]
- 9.Vansteenkiste JF, Vanakesa T, De Pas T, Zielinski M, Kim MS, Jassem J, Yoshimura M, Dahabreh J, Nakayama H, Havel L et al.. MAGRIT, a double-blind, randomized, placebo-controlled Phase III study to assess the efficacy of the RecMAGE-A3 + AS15 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-Positive non-small cell lung cancer (NSCLC). Ann Oncol 2014; 25:409-16; PMID:24368400; https://doi.org/ 10.1093/annonc/mdu089 [DOI] [PubMed] [Google Scholar]
- 10.Kruit WHJ, Suciu S, Dreno B, Mortier L, Robert C, Chiarion-Sileni V, Maio M, Testori A, Dorval T, Grob J-J et al.. Selection of immunostimulant AS15 for active immunization with MAGE-A3 protein: results of a randomized phase II study of the European Organisation for Research and Treatment of Cancer Melanoma Group in Metastatic Melanoma. J Clin Oncol 2013; 31:2413-20; PMID:23715572; https://doi.org/ 10.1200/JCO.2012.43.7111 [DOI] [PubMed] [Google Scholar]
- 11.Etheridge ML, Campbell SA, Erdman AG, Haynes CL, Wolf SM, McCullough J. The big picture on nanomedicine: the state of investigational and approved nanomedicine products. Nanomed: Nanotechnol Biol Med 2013; 9:1-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Butts C, Murray N, Maksymiuk A, Goss G, Marshall E, Soulières D, Cormier Y, Ellis P, Price A, Sawhney R et al.. Randomized Phase IIB trial of BLP25 liposome vaccine in stage IIIB and IV non-small-cell lung cancer. J Clin Oncol 2005; 23:6674-81; https://doi.org/ 10.1200/JCO.2005.13.011 [DOI] [PubMed] [Google Scholar]
- 13.Palmer M, Parker J, Modi S, Butts C, Smylie M, Meikle A, Kehoe M, MacLean G, Longenecker M. Phase I study of the BLP25 (MUC1 peptide) liposomal vaccine for active specific immunotherapy in stage IIIB/IV non-small-cell lung cancer. Clin Lung Cancer 2001; 3:49-57; discussion 8; PMID:14656392; https://doi.org/ 10.3816/CLC.2001.n.018 [DOI] [PubMed] [Google Scholar]
- 14.Vlad AM, Kettel JC, Alajez NM, Carlos CA, Finn OJ. MUC1 immunobiology: from discovery to clinical applications. Adv Immunol 2004; 82:249-93; PMID:14975259 [DOI] [PubMed] [Google Scholar]
- 15.Ten Brinke A, Karsten ML, Dieker MC, Zwaginga JJ, van Ham SM. The clinical grade maturation cocktail monophosphoryl lipid A plus IFNgamma generates monocyte-derived dendritic cells with the capacity to migrate and induce Th1 polarization. Vaccine 2007; 25:7145-52; PMID:17719152; https://doi.org/ 10.1016/j.vaccine.2007.07.031 [DOI] [PubMed] [Google Scholar]
- 16.Sangha R, North S. L-BLP25: a MUC1-targeted peptide vaccine therapy in prostate cancer. Expert Opin Biol Ther 2007; 7:1723-30; PMID:17961094; https://doi.org/ 10.1517/14712598.7.11.1723 [DOI] [PubMed] [Google Scholar]
- 17.Rossmann E, Osterborg A, Lofvenberg E, Choudhury A, Forssmann U, von Heydebreck A, Schroder A, Mellstedt H. Mucin 1-specific active cancer immunotherapy with tecemotide (L-BLP25) in patients with multiple myeloma: an exploratory study. Hum Vaccin Immunother 2014; 10:3394-408; PMID:25483677; https://doi.org/ 10.4161/hv.29918 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Schimanski CC, Mohler M, Schon M, van Cutsem E, Greil R, Bechstein WO, Hegewisch-Becker S, von Wichert G, Vohringer M, Heike M et al.. LICC: L-BLP25 in patients with colorectal carcinoma after curative resection of hepatic metastases: a randomized, placebo-controlled, multicenter, multinational, double-blinded phase II trial. BMC Cancer 2012; 12:144; PMID:22494623; https://doi.org/ 10.1186/1471-2407-12-144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ohyanagi F, Horai T, Sekine I, Yamamoto N, Nakagawa K, Nishio M, Senger S, Morsli N, Tamura T. Safety of BLP25 liposome vaccine (L-BLP25) in Japanese patients with unresectable stage III NSCLC after primary chemoradiotherapy: preliminary results from a Phase I/II study. Jpn J Clin Oncol 2011; 41:718-22; PMID:21393255; https://doi.org/ 10.1093/jjco/hyr021 [DOI] [PubMed] [Google Scholar]
- 20.Limentani SA, Campone M, Dorval T, Curigliano G, de Boer R, Vogel C, White S, Bachelot T, Canon JL, Disis M et al.. A non-randomized dose-escalation Phase I trial of a protein-based immunotherapeutic for the treatment of breast cancer patients with HER2-overexpressing tumors. Breast Cancer Res Treat 2016; 156:319-30; PMID:26993131; https://doi.org/ 10.1007/s10549-016-3751-x [DOI] [PubMed] [Google Scholar]
- 21.Vansteenkiste JF, Cho BC, Vanakesa T, De Pas T, Zielinski M, Kim MS, Jassem J, Yoshimura M, Dahabreh J, Nakayama H et al.. Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer (MAGRIT): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2016; 17:822-35; PMID:27132212; https://doi.org/ 10.1016/S1470-2045(16)00099-1 [DOI] [PubMed] [Google Scholar]
- 22.Shigematsu Y, Hanagiri T, Shiota H, Kuroda K, Baba T, Mizukami M, So T, Ichiki Y, Yasuda M, So T et al.. Clinical significance of cancer/testis antigens expression in patients with non-small cell lung cancer. Lung Cancer 2010; 68:105-10; PMID:19545928; https://doi.org/ 10.1016/j.lungcan.2009.05.010 [DOI] [PubMed] [Google Scholar]
- 23.Gure AO, Chua R, Williamson B, Gonen M, Ferrera CA, Gnjatic S, Ritter G, Simpson AJ, Chen YT, Old LJ et al.. Cancer-testis genes are coordinately expressed and are markers of poor outcome in non-small cell lung cancer. Clin Cancer Res 2005; 11:8055-62; PMID:16299236; https://doi.org/ 10.1158/1078-0432.CCR-05-1203 [DOI] [PubMed] [Google Scholar]
- 24.Moasser MM. The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene 2007; 26:6469-87; PMID:17471238; https://doi.org/ 10.1038/sj.onc.1210477 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Marty-Roix R, Vladimer GI, Pouliot K, Weng D, Buglione-Corbett R, West K, MacMicking JD, Chee JD, Wang S, Lu S et al.. Identification of QS-21 as an inflammasome-activating molecular component of saponin adjuvants. J Biol Chem 2016; 291:1123-36; PMID:26555265; https://doi.org/ 10.1074/jbc.M115.683011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Prieels J-P, Garcon-Johnson NM-JC, Slaoui M, Pala P. Vaccine composition containing adjuvants. 2003 [Google Scholar]
- 27.Krieg AM. CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol 2002; 20:709-60; PMID:11861616; https://doi.org/ 10.1146/annurev.immunol.20.100301.064842 [DOI] [PubMed] [Google Scholar]
- 28.Pujol J-L, Vansteenkiste JF, De Pas TM, Atanackovic D, Reck M, Thomeer M, Douillard J-Y, Fasola G, Potter V, Taylor P et al.. Safety and immunogenicity of MAGE-A3 cancer immunotherapeutic with or without adjuvant chemotherapy in patients with resected stage IB to III MAGE-A3-positive non-small-cell lung cancer. J Thorac Oncol 2015; 10:1458-67; PMID:26309191; https://doi.org/ 10.1097/JTO.0000000000000653 [DOI] [PubMed] [Google Scholar]
- 29.Berinstein NL, Karkada M, Oza AM, Odunsi K, Villella JA, Nemunaitis JJ, Morse MA, Pejovic T, Bentley J, Buyse M et al.. Survivin-targeted immunotherapy drives robust polyfunctional T cell generation and differentiation in advanced ovarian cancer patients. Oncoimmunology 2015; 4:e1026529; PMID:26405584; https://doi.org/ 10.1080/2162402X.2015.1026529 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Fukuda S, Pelus LM. Survivin, a cancer target with an emerging role in normal adult tissues. Mol Cancer Ther 2006; 5:1087-98; PMID:16731740; https://doi.org/ 10.1158/1535-7163.MCT-05-0375 [DOI] [PubMed] [Google Scholar]
- 31.Kitano S, Kageyama S, Nagata Y, Miyahara Y, Hiasa A, Naota H, Okumura S, Imai H, Shiraishi T, Masuya M et al.. HER2-specific T-cell immune responses in patients vaccinated with truncated HER2 protein complexed with nanogels of cholesteryl pullulan. Clin Cancer Res 2006; 12:7397-405; PMID:17189412; https://doi.org/ 10.1158/1078-0432.CCR-06-1546 [DOI] [PubMed] [Google Scholar]
- 32.Kageyama S, Kitano S, Hirayama M, Nagata Y, Imai H, Shiraishi T, Akiyoshi K, Scott AM, Murphy R, Hoffman EW et al.. Humoral immune responses in patients vaccinated with 1–146 HER2 protein complexed with cholesteryl pullulan nanogel. Cancer Sci 2008; 99:601-7; PMID:18081877; https://doi.org/ 10.1111/j.1349-7006.2007.00705.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Saito T, Wada H, Yamasaki M, Miyata H, Nishikawa H, Sato E, Kageyama S, Shiku H, Mori M, Doki Y. High expression of MAGE-A4 and MHC class I antigens in tumor cells and induction of MAGE-A4 immune responses are prognostic markers of CHP-MAGE-A4 cancer vaccine. Vaccine 2014; 32:5901-7; PMID:25218300; https://doi.org/ 10.1016/j.vaccine.2014.09.002 [DOI] [PubMed] [Google Scholar]
- 34.Wada H, Sato E, Uenaka A, Isobe M, Kawabata R, Nakamura Y, Iwae S, Yonezawa K, Yamasaki M, Miyata H et al.. Analysis of peripheral and local anti-tumor immune response in esophageal cancer patients after NY-ESO-1 protein vaccination. Int J Cancer 2008; 123:2362-9; PMID:18729190; https://doi.org/ 10.1002/ijc.23810 [DOI] [PubMed] [Google Scholar]
- 35.Muraoka D, Harada N, Hayashi T, Tahara Y, Momose F, Sawada S, Mukai SA, Akiyoshi K, Shiku H. Nanogel-based immunologically stealth vaccine targets macrophages in the medulla of lymph node and induces potent antitumor immunity. ACS Nano 2014; 8:9209-18; PMID:25180962; https://doi.org/ 10.1021/nn502975r [DOI] [PubMed] [Google Scholar]
- 36.Aoki M, Ueda S, Nishikawa H, Kitano S, Hirayama M, Ikeda H, Toyoda H, Tanaka K, Kanai M, Takabayashi A et al.. Antibody responses against NY-ESO-1 and HER2 antigens in patients vaccinated with combinations of cholesteryl pullulan (CHP)-NY-ESO-1 and CHP-HER2 with OK-432. Vaccine 2009; 27:6854-61; PMID:19761832; https://doi.org/ 10.1016/j.vaccine.2009.09.018 [DOI] [PubMed] [Google Scholar]
- 37.Kawada J, Wada H, Isobe M, Gnjatic S, Nishikawa H, Jungbluth AA, Okazaki N, Uenaka A, Nakamura Y, Fujiwara S et al.. Heteroclitic serological response in esophageal and prostate cancer patients after NY-ESO-1 protein vaccination. Int J Cancer 2012; 130:584-92; PMID:21413013; https://doi.org/ 10.1002/ijc.26074 [DOI] [PubMed] [Google Scholar]
- 38.Uenaka A, Wada H, Isobe M, Saika T, Tsuji K, Sato E, Sato S, Noguchi Y, Kawabata R, Yasuda T et al.. T cell immunomonitoring and tumor responses in patients immunized with a complex of cholesterol-bearing hydrophobized pullulan (CHP) and NY-ESO-1 protein. Cancer Immun 2007; 7:9; PMID:17441676 [PMC free article] [PubMed] [Google Scholar]
- 39.Kawabata R, Wada H, Isobe M, Saika T, Sato S, Uenaka A, Miyata H, Yasuda T, Doki Y, Noguchi Y et al.. Antibody response against NY-ESO-1 in CHP-NY-ESO-1 vaccinated patients. Int J Cancer 2007; 120:2178-84; PMID:17278093; https://doi.org/ 10.1002/ijc.22583 [DOI] [PubMed] [Google Scholar]
- 40.Kageyama S, Wada H, Muro K, Niwa Y, Ueda S, Miyata H, Takiguchi S, Sugino SH, Miyahara Y, Ikeda H et al.. Dose-dependent effects of NY-ESO-1 protein vaccine complexed with cholesteryl pullulan (CHP-NY-ESO-1) on immune responses and survival benefits of esophageal cancer patients. J Transl Med 2013; 11:246; PMID:24093426; https://doi.org/ 10.1186/1479-5876-11-246 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Maraskovsky E, Schnurr M, Wilson NS, Robson NC, Boyle J, Drane D. Development of prophylactic and therapeutic vaccines using the ISCOMATRIX adjuvant. Immunol Cell Biol 2009; 87:371-6; PMID:19381160; https://doi.org/ 10.1038/icb.2009.21 [DOI] [PubMed] [Google Scholar]
- 42.Davis ID, Chen W, Jackson H, Parente P, Shackleton M, Hopkins W, Chen Q, Dimopoulos N, Luke T, Murphy R et al.. Recombinant NY-ESO-1 protein with ISCOMATRIX adjuvant induces broad integrated antibody and CD4(+) and CD8(+) T cell responses in humans. Proc Natl Acad Sci USA 2004; 101:10697-702; PMID:15252201; https://doi.org/ 10.1073/pnas.0403572101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Frazer IH, Quinn M, Nicklin JL, Tan J, Perrin LC, Ng P, O'Connor VM, White O, Wendt N, Martin J et al.. Phase 1 study of HPV16-specific immunotherapy with E6E7 fusion protein and ISCOMATRIX adjuvant in women with cervical intraepithelial neoplasia. Vaccine 2004; 23:172-81; PMID:15531034; https://doi.org/ 10.1016/j.vaccine.2004.05.013 [DOI] [PubMed] [Google Scholar]
- 44.Chen JL, Dawoodji A, Tarlton A, Gnjatic S, Tajar A, Karydis I, Browning J, Pratap S, Verfaille C, Venhaus RR et al.. NY-ESO-1 specific antibody and cellular responses in melanoma patients primed with NY-ESO-1 protein in ISCOMATRIX and boosted with recombinant NY-ESO-1 fowlpox virus. Int J Cancer 2015; 136:E590-601; PMID:25081390; https://doi.org/ 10.1002/ijc.29118 [DOI] [PubMed] [Google Scholar]
- 45.Ebert LM, MacRaild SE, Zanker D, Davis ID, Cebon J, Chen W. A cancer vaccine induces expansion of NY-ESO-1-specific regulatory T cells in patients with advanced melanoma. PLoS One 2012; 7:e48424; PMID:23110239; https://doi.org/ 10.1371/journal.pone.0048424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Nicholaou T, Ebert LM, Davis ID, McArthur GA, Jackson H, Dimopoulos N, Tan B, Maraskovsky E, Miloradovic L, Hopkins W et al.. Regulatory T-cell-mediated attenuation of T-cell responses to the NY-ESO-1 ISCOMATRIX vaccine in patients with advanced malignant melanoma. Clin Cancer Res 2009; 15:2166-73; PMID:19276262; https://doi.org/ 10.1158/1078-0432.CCR-08-2484 [DOI] [PubMed] [Google Scholar]
- 47.Speiser DE, Schwarz K, Baumgaertner P, Manolova V, Devevre E, Sterry W, Walden P, Zippelius A, Conzett KB, Senti G et al.. Memory and effector CD8 T-cell responses after nanoparticle vaccination of melanoma patients. J Immunother 2010; 33:848-58; PMID:20842051; https://doi.org/ 10.1097/CJI.0b013e3181f1d614 [DOI] [PubMed] [Google Scholar]
- 48.Goldinger SM, Dummer R, Baumgaertner P, Mihic-Probst D, Schwarz K, Hammann-Haenni A, Willers J, Geldhof C, Prior JO, Kundig TM et al.. Nano-particle vaccination combined with TLR-7 and −9 ligands triggers memory and effector CD8(+) T-cell responses in melanoma patients. Eur J Immunol 2012; 42:3049-61; PMID:22806397; https://doi.org/ 10.1002/eji.201142361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bendandi M, Gocke CD, Kobrin CB, Benko FA, Sternas LA, Pennington R, Watson TM, Reynolds CW, Gause BL, Duffey PL et al.. Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma. Nat Med 1999; 5:1171-7; PMID:10502821; https://doi.org/ 10.1038/13928 [DOI] [PubMed] [Google Scholar]
- 50.Neelapu SS, Baskar S, Gause BL, Kobrin CB, Watson TM, Frye AR, Pennington R, Harvey L, Jaffe ES, Robb RJ et al.. Human autologous tumor-specific T-cell responses induced by liposomal delivery of a lymphoma antigen. Clin Cancer Res 2004; 10:8309-17; PMID:15623607; https://doi.org/ 10.1158/1078-0432.CCR-04-1071 [DOI] [PubMed] [Google Scholar]
- 51.Altin J, Atmosukarto Ines, De Wildt Rudolf Maria, Parish Christopher, Price Jason. Composition for Targeting Dendritic Cells. United States: Lipotek Pty Ltd (Acton, AU), Domantis Limited (Brentwood, GB), 2009 [Google Scholar]
- 52.Muir PD, Gunz FW. A cytogenetic study of eight human melanoma cell lines. Pathology 1979; 11:597-606; PMID:294576; https://doi.org/ 10.3109/00313027909059039 [DOI] [PubMed] [Google Scholar]
- 53.Moris A, Nobile C, Buseyne F, Porrot F, Abastado JP, Schwartz O. DC-SIGN promotes exogenous MHC-I-restricted HIV-1 antigen presentation. Blood 2004; 103:2648-54; PMID:14576049; https://doi.org/ 10.1182/blood-2003-07-2532 [DOI] [PubMed] [Google Scholar]
- 54.Lipotek Pty Ltd., Royal Adelaide Hospital Safety Study of a Liposomal Vaccine to Treat Malignant Melanoma. NCT01052142. Bethesda, MD, USA: National Library of Medicine, 2010. [cited April16, 2016] [Google Scholar]
- 55.Restifo NP, Gattinoni L. Lineage relationship of effector and memory T cells. Curr Opin Immunol 2013; 25:556-63; PMID:24148236; https://doi.org/ 10.1016/j.coi.2013.09.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Tsuji K, Hamada T, Uenaka A, Wada H, Sato E, Isobe M, Asagoe K, Yamasaki O, Shiku H, Ritter G et al.. Induction of immune response against NY-ESO-1 by CHP-NY-ESO-1 vaccination and immune regulation in a melanoma patient. Cancer Immunol Immunother 2008; 57:1429-37; PMID:18311489; https://doi.org/ 10.1007/s00262-008-0478-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Klein O, Davis ID, McArthur GA, Chen L, Haydon A, Parente P, Dimopoulos N, Jackson H, Xiao K, Maraskovsky E et al.. Low-dose cyclophosphamide enhances antigen-specific CD4(+) T cell responses to NY-ESO-1/ISCOMATRIX vaccine in patients with advanced melanoma. Cancer Immunol Immunother 2015; 64:507-18; PMID:25662405; https://doi.org/ 10.1007/s00262-015-1656-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Mehta NR, Wurz GT, Burich RA, Greenberg BE, Griffey S, Gutierrez A, Bell KE, McCall JL, Wolf M, DeGregorio M. L-BLP25 vaccine plus letrozole induces a TH1 immune response and has additive antitumor activity in MUC1-expressing mammary tumors in mice. Clin Cancer Res 2012; 18:2861-71; PMID:22434666; https://doi.org/ 10.1158/1078-0432.CCR-12-0168 [DOI] [PubMed] [Google Scholar]
- 59.Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, Solary E, Le Cesne A, Zitvogel L, Chauffert B. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother 2007; 56:641-8; PMID:16960692; https://doi.org/ 10.1007/s00262-006-0225-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Sampson JH, Aldape KD, Archer GE, Coan A, Desjardins A, Friedman AH, Friedman HS, Gilbert MR, Herndon JE, McLendon RE et al.. Greater chemotherapy-induced lymphopenia enhances tumor-specific immune responses that eliminate EGFRvIII-expressing tumor cells in patients with glioblastoma. Neuro Oncol 2011; 13:324-33; PMID:21149254; https://doi.org/ 10.1093/neuonc/noq157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sanchez-Perez L, Choi BD, Reap EA, Sayour EJ, Norberg P, Schmittling RJ, Archer GE, Herndon JE 2nd, Mitchell DA, Heimberger AB et al.. BLyS levels correlate with vaccine-induced antibody titers in patients with glioblastoma lymphodepleted by therapeutic temozolomide. Cancer Immunol Immunother 2013; 62:983-7; PMID:23591978; https://doi.org/ 10.1007/s00262-013-1405-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mitchell DA, Cui X, Schmittling RJ, Sanchez-Perez L, Snyder DJ, Congdon KL, Archer GE, Desjardins A, Friedman AH, Friedman HS et al.. Monoclonal antibody blockade of IL-2 receptor alpha during lymphopenia selectively depletes regulatory T cells in mice and humans. Blood 2011; 118:3003-12; PMID:21768296; https://doi.org/ 10.1182/blood-2011-02-334565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fry TJ, Mackall CL. Interleukin-7: from bench to clinic. Blood 2002; 99:3892-904; PMID:12010786; https://doi.org/ 10.1182/blood.V99.11.3892 [DOI] [PubMed] [Google Scholar]
- 64.Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA et al.. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol 2005; 23:2346-57; PMID:15800326; https://doi.org/ 10.1200/JCO.2005.00.240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Ebert LM, Liu YC, Clements CS, Robson NC, Jackson HM, Markby JL, Dimopoulos N, Tan BS, Luescher IF, Davis ID et al.. A long, naturally presented immunodominant epitope from NY-ESO-1 tumor antigen: implications for cancer vaccine design. Cancer Res 2009; 69:1046-54; PMID:19176376; https://doi.org/ 10.1158/0008-5472.CAN-08-2926 [DOI] [PubMed] [Google Scholar]
- 66.Sampson JH, Heimberger AB, Archer GE, Aldape KD, Friedman AH, Friedman HS, Gilbert MR, Herndon JE 2nd, McLendon RE, Mitchell DA et al.. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J Clin Oncol 2010; 28:4722-9; PMID:20921459; https://doi.org/ 10.1200/JCO.2010.28.6963 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Silva JM, Vandermeulen G, Oliveira VG, Pinto SN, Rodrigues C, Salgado A, Afonso CA, Viana AS, Jérôme C, Silva LC et al.. Development of functionalized nanoparticles for vaccine delivery to dendritic cells: a mechanistic approach. Nanomedicine 2014; 9:2639-56; PMID:25529568; https://doi.org/ 10.2217/nnm.14.135 [DOI] [PubMed] [Google Scholar]
- 68.Chernenko T, Sawant RR, Miljkovic M, Quintero L, Diem M, Torchilin V. Raman microscopy for non-invasive imaging of pharmaceutical nanocarriers: intracellular distribution of cationic liposomes of different composition. Mol Pharm 2012; 9:930-6; PMID:22376068; https://doi.org/ 10.1021/mp200519y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hamborg M, Rose F, Jorgensen L, Bjorklund K, Pedersen HB, Christensen D, Foged C. Elucidating the mechanisms of protein antigen adsorption to the CAF/NAF liposomal vaccine adjuvant systems: effect of charge, fluidity and antigen-to-lipid ratio. Biochim Biophys Acta 2014; 1838:2001-10; PMID:24769435; https://doi.org/ 10.1016/j.bbamem.2014.04.013 [DOI] [PubMed] [Google Scholar]
- 70.Hu Y, Ehrich M, Fuhrman K, Zhang C. In vitro performance of lipid-PLGA hybrid nanoparticles as an antigen delivery system: lipid composition matters. Nanoscale Res Lett 2014; 9:434; PMID:25232295; https://doi.org/ 10.1186/1556-276X-9-434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Soema PC, Willems GJ, Jiskoot W, Amorij JP, Kersten GF. Predicting the influence of liposomal lipid composition on liposome size, zeta potential and liposome-induced dendritic cell maturation using a design of experiments approach. Eur J Pharm Biopharm 2015; 94:427-35; PMID:26144666; https://doi.org/ 10.1016/j.ejpb.2015.06.026 [DOI] [PubMed] [Google Scholar]
- 72.Kauffman KJ, Dorkin JR, Yang JH, Heartlein MW, DeRosa F, Mir FF, Fenton OS, Anderson DG. Optimization of lipid nanoparticle formulations for mRNA delivery in vivo with fractional factorial and definitive screening designs. Nano Lett 2015; 15:7300-6; PMID:26469188; https://doi.org/ 10.1021/acs.nanolett.5b02497 [DOI] [PubMed] [Google Scholar]
- 73.Tavakoli S, Tamaddon AM, Golkar N, Samani SM. Microencapsulation of (deoxythymidine)20-DOTAP complexes in stealth liposomes optimized by Taguchi design. J Liposome Res 2015; 25:67-77; PMID:24960449; https://doi.org/ 10.3109/08982104.2014.928889 [DOI] [PubMed] [Google Scholar]
- 74.Klechevsky E, Morita R, Liu M, Cao Y, Coquery S, Thompson-Snipes L, Briere F, Chaussabel D, Zurawski G, Palucka AK et al.. Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity 2008; 29:497-510; PMID:18789730; https://doi.org/ 10.1016/j.immuni.2008.07.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Gerner MY, Torabi-Parizi P, Germain RN. Strategically localized dendritic cells promote rapid T cell responses to lymph-borne particulate antigens. Immunity 2015; 42:172-85; PMID:25607462; https://doi.org/ 10.1016/j.immuni.2014.12.024 [DOI] [PubMed] [Google Scholar]
- 76.Sehgal K, Ragheb R, Fahmy TM, Dhodapkar MV, Dhodapkar KM. Nanoparticle-mediated combinatorial targeting of multiple human dendritic cell (DC) subsets leads to enhanced T cell activation via IL-15-dependent DC crosstalk. J Immunol 2014; 193:2297-305; PMID:25080481; https://doi.org/ 10.4049/jimmunol.1400489 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Slingluff CL, Petroni GR, Olson WC, Smolkin ME, Chianese-Bullock KA, Mauldin IS, Smith KT, Deacon DH, Varhegyi NE, Donnelly SB et al.. A randomized pilot trial testing the safety and immunologic effects of a MAGE-A3 protein plus AS15 immunostimulant administered into muscle or into dermal/subcutaneous sites. Cancer Immunol Immunother 2016; 65:25-36; PMID:26581199; https://doi.org/ 10.1007/s00262-015-1770-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Fan Y, Sahdev P, Ochyl LJ, J Akerberg J, Moon JJ. Cationic liposome-hyaluronic acid hybrid nanoparticles for intranasal vaccination with subunit antigens. J Control Rel 2015; 208:121-9; PMID:25869965; https://doi.org/ 10.1016/j.jconrel.2015.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wang C, Liu P, Zhuang Y, Li P, Jiang B, Pan H, Liu L, Cai L, Ma Y. Lymphatic-targeted cationic liposomes: a robust vaccine adjuvant for promoting long-term immunological memory. Vaccine 2014; 32:5475-83; PMID:25110295; https://doi.org/ 10.1016/j.vaccine.2014.07.081 [DOI] [PubMed] [Google Scholar]
- 80.Fromen CA, Rahhal TB, Robbins GR, Kai MP, Shen TW, Luft JC, DeSimone JM. Nanoparticle surface charge impacts distribution, uptake and lymph node trafficking by pulmonary antigen-presenting cells. Nanomed: Nanotechnol Biol Med 2015; PMID:26656533 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Shima F, Uto T, Akagi T, Baba M, Akashi M. Size effect of amphiphilic poly(γ-glutamic acid) nanoparticles on cellular uptake and maturation of dendritic cells in vivo. Acta Biomater 2013; 9:8894-901; PMID:23770225; https://doi.org/ 10.1016/j.actbio.2013.06.010 [DOI] [PubMed] [Google Scholar]
- 82.Wu YL, Park K, Soo RA, Sun Y, Tyroller K, Wages D, Ely G, Yang JC, Mok T. INSPIRE: a phase III study of the BLP25 liposome vaccine (L-BLP25) in Asian patients with unresectable stage III non-small cell lung cancer. BMC Cancer 2011; 11:430; PMID:21982342; https://doi.org/ 10.1186/1471-2407-11-430 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Butts C, Murray RN, Smith CJ, Ellis PM, Jasas K, Maksymiuk A, Goss G, Ely G, Beier F, Soulieres D. A multicenter open-label study to assess the safety of a new formulation of BLP25 liposome vaccine in patients with unresectable stage III non-small-cell lung cancer. Clin Lung Cancer 2010; 11:391-5; PMID:21071331; https://doi.org/ 10.3816/CLC.2010.n.101 [DOI] [PubMed] [Google Scholar]
- 84.North SA, Graham K, Bodnar D, Venner P. A pilot study of the liposomal MUC1 vaccine BLP25 in prostate specific antigen failures after radical prostatectomy. J Urol 2006; 176:91-5; PMID:16753376; https://doi.org/ 10.1016/S0022-5347(06)00494-0 [DOI] [PubMed] [Google Scholar]
- 85.Hamilton E, Blackwell K, Hobeika AC, Clay TM, Broadwater G, Ren XR, Chen W, Castro H, Lehmann F, Spector N et al.. Phase 1 clinical trial of HER2-specific immunotherapy with concomitant HER2 kinase inhibition [corrected]. J Transl Med 2012; 10:28; PMID:22325452; https://doi.org/ 10.1186/1479-5876-10-28 [DOI] [PMC free article] [PubMed] [Google Scholar]
