Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Apr 1.
Published in final edited form as: Curr Opin Immunol. 2018 Mar 16;51:111–122. doi: 10.1016/j.coi.2018.03.001

Cancer Vaccines: Translation from mice to human clinical trials

Hoyoung Maeng 1, Masaki Terabe 1, Jay A Berzofsky 1
PMCID: PMC5943163  NIHMSID: NIHMS950528  PMID: 29554495

Abstract

Therapeutic cancer vaccines have been a long-sought approach to harness the exquisite specificity of the immune system to treat cancer, but until recently have not had much success as single agents in clinical trials. However, new understanding of the immunoregulatory mechanisms exploited by cancers has allowed the development of approaches to potentiate the effect of vaccines by removing the brakes while the vaccines step on the accelerator. Thus, vaccines that had induced a strong T cell response but no clinical therapeutic effect may now reach their full potential. Here, we review a number of promising approaches to cancer vaccines developed initially in mouse models and their translation into clinical trials, along with combinations of vaccines with other therapies that might allow cancer vaccines to finally achieve clinical efficacy against many types of cancer.

Introduction

Cancer vaccines have been long sought as a therapy that could marshal the exquisite specificity of the immune system to fight cancer without the side effects of other less precisely targeted therapies. Despite successes in preventing cancers in mouse models, the difficulty in treating large established tumors in mice, together with failures in translation to humans, led to the expectation that cancer vaccines may not be feasible [1,2]. The successful pivotal trial and licensing of Sipuleucel-T (Provenge) in 2010 reinvigorated the field, despite the limited increase in survival time [3]. More academic labs and companies have invested in cancer vaccines since that milestone event. However, concurrently, other forms of immunotherapy have emerged that have taken hold of the field and attracted most of the new effort. These include checkpoint inhibitors such as anti-CTLA-4, anti-PD-1, and anti-PD-L1 that block immunoregulatory mechanisms inhibiting the immune response to cancer and allow the immune system to reject the cancer in some cases [47], and adoptive T cell therapies, involving T cells from patients selected for their anti-tumor activity or genetically engineered to express a T or B cell receptor targeting a cancer antigen, that are expanded ex vivo and infused [811]. Both of these approaches have now been licensed by the Food and Drug Administration (FDA) and are showing impressive results in certain malignancies. At the same time, newer cancer vaccines are emerging that also show great promise, and it is becoming clear from animal models that cancer vaccines can synergize with both checkpoint inhibitors [12,13] and adoptive cell therapy [2,8,1416]. Moreover, the barrier to success of many cancer vaccines now appears to be the presence of so many immunosuppressive mechanisms in cancer that inhibit the immune response in the tumor microenvironment even when the vaccine can induce a response that is measurable systemically. Besides checkpoint molecules, there are immunosuppressive cytokines like TGF-beta [1719] and IL-10 [20], and immunosuppressive cells like T-regulatory cells [2123], myeloid-derived suppressor cells[2427], regulatory NKT cells [28], tumor-associated M2 macrophages[27,29,30], B-regulatory cells [31], tolerogenic dendritic cells, and others. Therefore, we believe that the future of cancer immunotherapy is to combine cancer vaccines with these other therapies, especially treatments to overcome immunosuppression.

Here, we will review a sampling of cancer vaccines, including two from our own lab and several from others, that were originally developed in mouse models and then successfully translated into human clinical trials showing promising results. While this short review cannot cover a comprehensive survey of the field, these examples will illustrate how preclinical mouse tumor models are effective to develop cancer vaccines that can work in humans, and how different strategies for cancer vaccines can benefit cancer patients and potentially synergize with methods to overcome immunosuppression. Thus, cancer vaccines should soon become the third pillar of cancer immunotherapy and a key component of combination immunotherapy.

Novel promising cancer vaccine strategies from the Vaccine Branch, National Cancer Institute, NIH

Both the cellular and the humoral arms of the immune system can be used to attack cancer. However, antibodies are limited to detecting tumor antigens on the surface of intact tumor cells, whereas T cells can detect any protein made in the cell, because fragments of all proteins made are transported to the surface by Major Histocompatibility Complex (MHC) molecules such as human leukocyte antigens (HLA antigens), which serve an internal surveillance mechanism [32]. Thus, most cancer vaccines have aimed to induce T cells, because these can recognize any protein-based tumor antigen. Here, we will illustrate examples of both.

In order for peptide fragments of protein tumor antigens to bind to MHC molecules, they must fit in the peptide binding groove, and higher affinity binding usually corresponds to greater immunogenicity [33]. Tumor antigens did not evolve to be good MHC binders, so we developed a strategy called “epitope enhancement” in which we modified the sequence to improve affinity without affecting the surface seen by the T cell receptor (TCR), so that we could still elicit T cells recognizing the unmodified antigen [3436]. Others have seen similar improvements by such epitope enhancement of cancer antigens [37,38] or modifications improving T cell recognition [39]. We applied epitope enhancement to peptides we mapped to bind to HLA-A*0201, the most common human class I MHC molecule, present in nearly half the population, from TARP, a prostate and breast cancer antigen described by Ira Pastan’s lab [40]. We showed improved immunogenicity in HLA-A2-transgenic mice and ability to recognize the natural sequence, and confirmed this with human T cells, which could kill human cancer cells [41]. We translated this to a phase I clinical trial in HLA-*0201 stage D0 prostate cancer patients, in which a rising prostate-specific antigen (PSA) indicated microscopic recurrence after prostate removal, despite no radiographic evidence of tumor. In this setting, the rate of rise of PSA is a validated predictor of clinical course and response to therapy [4247]. Patients got immunized 5 times with peptides either in Montanide-ISA51 adjuvant or coated onto autologous dendritic cells (DCs), and since the two arms gave comparable results, they were pooled for statistics. We found that 74% of vaccinated patients had a decreased PSA slope at 1 year compared to their own baseline slope (p = 0.0004), and the median tumor growth rate constant fell in half (p = 0.003), with no significant adverse events [48] 78% also made a new T cell response to the peptides. Based on these results, a randomized placebo-controlled phase II trial, with more peptides to expand beyond HLA-A*0201, has been opened.

Human epidermal growth factor receptor 2 (HER2) is a driver oncogene product expressed on the surface of about 25% of breast cancers and to a lesser extent on many other solid tumors, where it is accessible to antibodies. Monoclonal antibodies to HER2, and other HER2-directed agents, have already shown clinical benefit [4951]. However, no vaccine is available to induce such antibodies. We made an adenovirus expressing the extracellular & transmembrane (ECTM) domains of rodent HER2 and used it to immunize either HER2 transgenic mice that develop autochthonous breast cancers or wild type mice with large established syngeneic TUBO breast tumors [5254]. The vaccine prevented growth of the autochthonous tumors and cured nearly 100% of mice with large established TUBO mammary tumors (up to 2 cm), and established lung metastases[54], but surprisingly was independent of effector T cells, requiring only antibody production. The HER2 antibodies were Fc-receptor (FcR) independent, unlike trastuzumab [55], but rather blocked HER2 phosphorylation [54]. We translated this to a human phase I trial with an adenovirus expressing human HER2 ECTM, used to transduce autologous DCs. First tested in non-breast cancer patients naïve to HER2 therapies (with gastroesophageal, colorectal, ovarian, lung and bladder cancers), it was safe and showed clinical benefit in 5/11 (45%) of evaluable patients at the second and third dose levels [56]. This trial has now been opened to breast cancer patients who have failed other HER2-directed therapies. In addition, there are peptide-based and other vaccines under study to induce T cell responses to HER2 [57,58].

Representative other examples of promising novel cancer vaccines Prostvac

Cancer/testis antigens, whose expression is limited to cancer and non-vital organs, can be a good target for vaccines. A hurdle of targeting such antigens is that we need to break self-tolerance. Prostate-specific antigen (PSA) is such a self-antigen expressed in prostate cancer. Schlom and his coworkers developed PROSTVAC to deal with this problem by incorporating two simultaneous approaches. One is the use of viral vectors expressing PSA, which induces strong inflammation to induce massive T cell responses. The viral vaccines in therapeutic use have been shown to be able to break tolerance in multiple mouse tumor models [5961]. Furthermore, they have demonstrated that the T cell-mediated immune responses induced by a viral vector can be maximized by taking a prime-boost strategy in which immune responses were primed by a recombinant vaccinia virus followed by boosts with a recombinant fowlpox virus [62,63]. Second is to strengthen co-stimulation for T cells by transducing genes of three co-stimulator molecules, LFA-3, ICAM-1 and B7, whose ligands are CD2, LFA-1, and CD28, respectively (together called TRICOM vectors). The effect of the combination is more dramatic when there is a weak TCR signal or low frequency of T cells against target cells [64]. These conditions represent the environment to which T cells are likely to be exposed in tumors, as tumors frequently decrease the expression of class I MHC. The TRICOM vectors have also been shown to improve not only the quantity/magnitude of T cell responses but also the quality of the responses by inducing T cells with higher functional avidity [65,66] .

PROSTVAC was investigated in 2 Phase II studies in metastatic castration-resistant prostate cancer (mCRPC). The first trial was a placebo-controlled, multicenter trial in 125 mCRPC patients with 2:1 randomization that showed overall survival benefit (25.1 months vs. 16.6 months; P = 0.0061) with PROSTVAC compared to the empty poxviral vector. No advantage in time to progression (TTP) was detected as shown similarly in the case of Sipuleucel-T, whereas both did show benefit in overall survival (OSS) [3]. The second trial investigated the influence of GM-CSF with PROSTVAC in a total of only 32 patients that showed no difference in the T cell response or survival between the group receiving GM -CSF and no GM-CSF [67]. PROSTVAC received Special Protocol Assessment (SPA) from the FDA in collaboration with NCI that lead to the PROSPECT study, a global, randomized, double-blind, placebo-controlled phase III trial in patients with mCRPC (NCT01322490). In September 2017, an independent Data Monitoring Committee (DMC) determined that continuation of the trial was futile based on a preplanned interim analysis after fully enrolling approximately 1300 patients with OSS longer than 30 months. It is unclear what led to the divergence in outcome between the phase III trial and the phase II trial. Newly available treatment options could have affected the overall course after the vaccination, in addition to the limited effect of the vaccine as a single agent on the tumor microenvironment during the prolonged treatment course, abrogating the benefit that had been seen in the phase II study. Prostvac might be another example to reinforce previous evidence that a cancer vaccine alone is not sufficient to gain the control of cancer, emphasizing the need for combinations with other agents, such as checkpoint inhibitors, to overcome negative immunoregulatory factors. Additional studies with combination strategies with nivolumab (NCT02933255) in CRPC and with docetaxel in metastatic castration sensitive prostate cancer (NCT02649855) are enrolling the patients. The use of PROSTVAC in the adjuvant setting for high-risk patients and in combination with ipilimumab in the neoadjuvant setting (NCT02506114) as well as in biochemically recurrent or localized prostate cancer is also under investigation.

Human Papillomavirus (HPV) therapeutic vaccines

Among human cancer viruses, HPV has been studied most and most successfully with visible outcome while hepatitis B virus vaccines have certainly reduced incidence of hepatocellular carcinoma, and there are ongoing efforts for hepatitis C virus, Epstein-Barr Virus (EBV), Human T-Lymphotropic Virus (HTLV)-1 and Merkel cell polyoma virus. Even though preventive vaccines covering the high-risk HPV subtypes such as HPV16 and HPV18 can affect the scourge of HPV associated cancer, the vaccination rate has not been satisfactory so far and there are many individuals who already have persistent HPV infection and are at risk of developing cancer, revealing the unmet need of developing therapeutic HPV vaccines.

Early experience with peptide vaccines raised the concern that they may work only in mice but never in human patients. This reputation was set by a long list of clinical trials with peptide vaccines that never achieved success [1]. However, recent work on a long peptide vaccine against HPV, which causes cervical cancer, clearly demonstrated that peptide vaccines work in humans if we design them properly. What made the HPV vaccine different from others? In many previous studies, short peptides with minimal length (9 to 11 amino acid residues long) for binding with a class I MHC, called minimal cytotoxic T lymphocyte (CTL) epitopes, were used as a vaccine. Using minimal CTL epitopes has potential problems. For CD8+ T cells to be fully activated by recognizing a peptide/MHC complex, they need “help” from CD4+ T cells. The help includes 1) “licensing” professional antigen presenting cells (APCs) through CD40-CD40L interaction, which activates and matures them [6870], and 2) supplying a growth factor, IL-2. It has been shown in both viral infection and tumor models that in the absence of CD4+ T cell help long-lasting protection cannot be induced [71,72]. Another potential problem with a short minimal CTL epitope is that it replaces peptides presented by class I MHC on the cell surface, loading directly without processing. As class I MHC molecules are expressed on almost all types of cells, this means that administered peptides will not be presented only by professional APCs but primarily by non-professional APCs that do not provide proper second signals to CD8+ T cells, because they do not have co-stimulatory molecules. Thus, additional CD4+ T cell epitopes peptide must be included in a peptide vaccine, and it may be preferable to use the epitope from vaccine-targeting antigen.

A study by Zwaveling et al using a mouse model with the TC1 cell line, which is an epithelial cell tumor line transformed by HPV16 E6 and E7, demonstrated that a HPV16-derived 35-amino-acid long peptide (HPV E743–77) containing both a CD8+ T cell epitope and a CD4+ T cell epitope induced a far more robust CD8+ T cell response capable of eliminating an established tumor while a minimal CTL epitope (HPV E749–57) did not [73]. One mechanism by which the long peptide can induce such CD8+ T cell responses is that a long peptide requires processing by a mechanism unique to professional APCs, such as cross-presentation, to be presented by class I MHC. In contrast, short minimum CTL epitopes can be loaded onto class I MHC by replacing a peptide already bound to class I MHC molecules on cell surface, and hence can be presented by any cells expressing class I MHC molecules without a requirement for endogenous processing. Thus, non-professional APCs cannot present the antigen. It was also shown that the long peptide increased the duration of antigen presentation in vivo, which contributes to the induction of long-lasting immune response [74].

Based on the unexpected strong efficacy of the long peptide in mouse tumor models, Melief and coworkers launched a development pipeline for ISA101, a mixture of HPV16 E6 and E7 synthetic long peptide (HPV16-SLP) with Montanide ISA-51. A Phase II study in vaginal intraepithelial neoplasia (VIN) Grade III showed viral specific immunogenicity in all vaccinated (n=22) patients and clinical response in 79%, with a complete response in 47% at 12 months of follow-up that was maintained at 24 months of follow-up. No toxicity CTCAE grade III or higher was observed [7577]. Several additional studies as a single agent or in combination using ISA101 are pending final reports. A study of ISA101 in combination with standard of care paclitaxel and carboplatin chemotherapy in advanced cervical cancer was presented during ASCO-SITC 2017 by Melief and Welters [78], reporting an objective response rate (ORR) of 67% vs 25% and disease control rate (ORR+SD) of 94% vs 79% in patients without prior systemic chemotherapy or with prior systemic chemotherapy and survival benefit correlating with HPV-specific immune response.

Another Phase IIb study was successfully conducted by Cornelia Trimble and coworkers using VGX-3100, a plasmid mixture targeting HPV 16 and 18 E6 and E7 proteins delivered by electroporation (Cellectra) for CIN 2/3 patients (N=167), showing 18.2% (95% CI 1.3–34.4; p=0.034) higher histopathologic regression (48.2 vs 30.0%) in modified intention-to-treat analysis with higher concomitant viral clearance in vaccine treated patients (40.2% vs 14.3%, 95% CI 8.0–39.2; p=0.003)[79]. This study laid the groundwork for additional studies in the full spectrum of precancerous and cancerous lesions caused by HPV (NCT03180684, NCT02163057, NCT02172911), including a phase III study for CIN 2/3 (NCT03185013).

MUC1

Finn and coworkers have established the usefulness of the link between tumor-associated antigens (TAA) or tumor specific antigen and disease-associated antigens (DAA) in the research on MUC1, justifying how such an antigen can be a target for a prophylactic vaccine. A DAA is defined as a self-antigen abnormally expressed during acute or chronic inflammation or infection. For example, hypoglycosylated MUC1 is a DAA for mumps that can induce anti-MUC1 antibody but a TAA for pancreatic cancer, ovarian cancer and several other cancer types. Aberrant, hypoglycosylated MUC1 has been identified also in adenocarcinoma and hematologic malignancies. Typically, immune recognition requires an MHC-restricted process to present antigens and only rare situations of MHC-unrestricted TCR recognition have been described. Magarian-Blander et al. published that a repeated MUC1 tandem epitope was able to induce the activation of cytotoxic T cells without presentation by MHC molecules even though antibody to the TCR and CD3 complex blocked the process[80]. Also, mice reconstituted with such TCR-transduced bone marrow cells were able to control MUC1-positive human tumors [81]. A history of childhood mumps might be protective against ovarian cancer according to the case-control studies and the protection seemed to be related to the presence of the anti-MUC1 antibody that rejected tumor cells in mice [82]. If the immunity against MUC1 provided immune protection against the development of ovarian cancer, can the same strategy be used for the cancer prevention by enhancing immune surveillance in high-risk individuals? Accordingly, the Finn group has been focusing on the development of a prophylactic cancer vaccine [83,84].

A MUC1 vaccine, a synthetic 100-amino acid peptide containing 5 MUC1 20-residue repeats, induced antibody responses and long-lasting immune memory in 43.6% of 39 individuals with advanced colonic adenoma[85]. High circulating myeloid-derived suppressor cells correlated with poor antibody response. Based on this work, a randomized, placebo-controlled Phase II study (NCT02134925) is underway in 120 patients with advanced adenomas with the end point of immunogenicity and 25% reduction in polyp recurrence after 3 yrs.

Vaccines for hematologic malignancies

Hematologic malignancies have been the target of many pioneering cancer treatments, exemplified by the first cell therapy (hematopoietic stem cell transplantation), first anti-cancer monoclonal antibody (rituximab), first tyrosine kinase inhibitor (imatinib) and first CAR-T cell therapy (ALL). However, with the success of monoclonal antibodies and kinase inhibitors, the need for vaccine studies has not seemed pressing.

Idiotype vaccines in lymphoma, pioneered by Levy, fascinated many researchers with the idea of providing active immunotherapies evoking polyclonal response while preserving normal B cells as a new concept in cancer immunotherapy from early 1990’s [86]. B cell tumors express unique surface immunoglobulin and idiotype (Id) refers to the unique amino acid sequences within the complementarity determining regions (CDR) of the variable regions of the heavy and light chain of such an Ig. Thus, these are among the early tumor-specific personalized antigens to be studied in humans as targets of cancer vaccines. A study (n=20) lead by Larry Kwak [87] demonstrated not only humoral but also a tumor cell-specific T cell response after vaccination in follicular lymphoma patients in remission after chemotherapy. However, such an id vaccine did not show advantage either in combination with rituximab (n=349) [88] or as a single agent first-line treatment in advanced disease (n=675) [89] in the subsequent two multi-center randomized Phase III studies. Even though Schuster reported improved PFS in advanced stage follicular lymphoma [90] when the vaccine was given when the patients were in remission after chemotherapy, this did not advance for further development. Instead, B cell malignancies became a new focus of adoptive cell therapy.

WT1, the top-ranked cancer antigen in the Prioritization of Cancer antigens proposed by NCI in 2009 considering therapeutic function, immunogenicity, and specificity, is overexpressed both in hematologic malignancies and solid tumors [91]. Oka reported the immunogenicity of the 9-mer WT1 peptide Db126 inducing a CTL response and rejection of WT1-expressing tumor cells in mice immunized with the Db126 peptide [92]. However, no vaccine studies of WT1 have reached Phase III and only a few small studies done in NCI and NHLBI showed disappointing clinical efficacy so far in hematologic malignancies. Currently, studies targeting malignant mesothelioma and ovarian cancer are accruing patients.

In myeloid malignancies, the PR1 vaccine had a long-waited report. PR1 is a HLA-A2 restricted 9-mer peptide derived from proteinase-3 and neutrophil elastase that are normal components of neutrophil azurophilic granules overexpressed in myeloid blast cells. A Phase I/II study treating recurrent or high-risk AML/MDS and CML patients (n=66) with repeated doses of PR1 showed an immune response in 53% and clinical response in 24% with minimal toxicity [93].

Vaccines against neo-antigen epitopes

With the recent discoveries that cancers with multiple somatic mutations are the most immunogenic and thus most responsive to checkpoint inhibitors [6] and that tumor-infiltrating lymphocytes often target such neo-antigens [94,95], there have been recent attempts at developing personalized cancer vaccines targeting such neo-antigen epitopes [96,97]. However, some of the earliest neo-antigens that fit this description are actually mutant Kras and mutant p53, a frequently mutated tumor oncogene and tumor suppressor gene, that were studied as vaccine candidates over a decade earlier [98101]. In these studies, it was found that the most common Kras mutations at codon 12 and 13 fall in a sequence predicted to bind to HLA-A*0201, and indeed these peptides did bind to HLA-A*0201. In addition, many p53 mutations were in such segments that bound to HLA-A*0201 or other common human HLA molecules. Early clinical trials showed immune responses in cancer patients with mutant ras or p53 [102,103] and there was a significant correlation between cytokine responses to these peptides in vaccinated patients and their overall survival, with median survivals of 393 days with a cytotoxic T cell response to these peptides compared to 98 days without, or of 470 days with an interferon-gamma response to the peptides vs 88 days without, although cause and effect could not be proven [103]. Recently, binding of a mutant Kras peptide to HLA-Cw08 with recognition by a protective tumor infiltrating lymphocyte population was also reported [104]. Thus, from these mutant ras and p53 peptides, there is already precedent for use of neo-antigen-based vaccines.

Combinations of vaccines with other cancer therapies

From the history of clinical trials cancer vaccines, we have multiple proofs that vaccines can induce antigen-specific T cells but the response from vaccine alone is not sufficient to control the cancer. From recent breakthroughs in basic studies of immune regulation, such as checkpoint receptors and various types of regulatory T and B cells and myeloid cells, we now understand that the limiting hurdle to cancer vaccine efficacy is not necessarily the induction of an adequate magnitude T cell or antibody response, but the ability of that response to eradicate tumors in the face of these counterposing forces in the hostile tumor microenvironment. Thus, vaccine combinational strategies with cytokines, radiation, chemotherapy or hormonal therapy have been explored for possible synergy, and more recently, vaccine combinations with checkpoint inhibitors such as anti-CTLA-4 or anti-PD-1/PD-L1 are actively investigated to allow T cells induced by the vaccine to enter the tumor and to be effective there without inhibition by ligands such as PD-L1 [105,106]. As CTLA-4 is a receptor that turns off activated T cells and may prevent their entrance into the tumor parenchyma [107], and may also serve as a target for antibody-dependent killing of T-regulatory cells [108], and PD-1/PD-L1 interaction tunes down the T cell receptor and costimulatory signaling cascade by counteracting kinases in these signaling pathways [109], blocking such CTLA-4 or PD-1/PD-L1 interaction is expected to permit the immune responses generated by vaccines to more effectively control cancers. This is an innovative strategy that may reinvigorate the immune cells that were not successful enough in providing adequate immune surveillance.

A combination of PROSTVAC with ipilumumab (anti-CTLA-4) was studied in 30 patients with no increased immune-related adverse events and with a PSA decline in 58% of 24 chemotherapy-naïve metastatic castrate resistant prostate cancer (mCRPC) patients [110]. Vaccines combined with either ipilimumab or nivolumab (anti-PD-1) in recurrent or high-risk melanoma patients showed acceptable toxicity, potential survival benefit, and decreased recurrence[111] [112]. Glisson and coworkers [113] presented an ORR of 33% during ESMO 2017 in a study on ISA101 with Nivolumab in HPV16-positive incurable solid tumors (n=22), mostly oropharyngeal cancers (NCT02426892). Studies of vaccines in combination with checkpoint inhibitors are summarized in Table 1.

Table 1.

Selected therapeutic cancer vaccine trials in combination with checkpoint inhibitors

Checkpoint Inhibitor Vaccine/Target Cancer Types Phase/ClinicalTrials.gov Identifier
Anti-CTLA-4
 Ipilimumab GVAX Pancreas 1, 2 NCT00836407, NCT01896869
NeoVax (neoantigen peptides) Kidney 1 NCT02950766
Pxca-Vec Multiple solid tumors 1 NCT02977156
NY-ESO-1pulsed DC Multiple solid tumors 1 NCT02070406
Prostvac Prostate 1, 2 NCT00113984, NCT02506114
gp100 Melanoma 3 NCT00094653
2 NCT00032045, NCT00077532
gp100, MART-1 Melanoma 1 NCT00025181, NCT00028431
IDO peptide Melanoma 1 NCT02077114
6MHP (MHC Class II peptides) Melanoma 1, 2 NCT02385669
MART-1 Melanoma 2 NCT00084656
 Tremelimumab PrCa VBIR Prostate 1 NCT02616185
MART-1 Melanoma 1 NCT00090896
Anti-PD-1
 Nivolumab GVAX, CRS207/mesothelin Pancreas 2 NCT02243371, NCT03190265
GVAX Pancreas 1, 2 NCT02451982, NCT03161379
ISA 101/HPV16 E6 and E7 HPV16 cancers 2 NCT02426892
CIMAvax/EGF NSCLC 1, 2 NCT02955290
CD40L, GM NSCLC (adenocarcinoma) 1, 2 NCT02466568
NY-ESO-1, MART-1, gp100 Melanoma 1 NCT01176461
PD-L1/IDO peptide Melanoma 1, 2 NCT03047928
NY-ESO-1 pulsed DC Multiple solid tumors 1 NCT02775292
WT1 Ovary 1 NCT02737787
HHS-110 (Viagenpumatucel-L) NSCLC (adeno) 1, 2 NCT02439450
NEO-PV01 Bladder, NSCLC, melanoma 1 NCT02897765
Prostvac Prostate 1, 2 NCT02933255
DCVax-L (tumor lysate) GBM 2 NCT03014804
TG4010/MUC1 NSCLC 2 NCT02823990
Synthetic long peptide Follicular lymphoma 1 NCT03121677
 Pembrolizumab pTVG-HP DNA mCRPC 1, 2 NCT02499835
6MHP (MHC Class II peptides) Melanoma 1, 2 NCT02515227
Synthetic long peptide NSCLC 1 CT03166254
GVAX Pancreas 1 NCT03153410
2 NCT02648282, NCT03006302
GVAX CRC 2 NCT02981524
Peptide Pancreas, CRC 1 NCT02600949
DPX-Survivac Ovary 2 NCT03029403
Oncolytic MG1-MAGE-A3 NSCLC 1, 2 NCT02879760
P53MVA Ovary 2 NCT03113487
Multiple solid tumors 1 NCT02432963
mRNA-4157 Multiple solid tumors 1 NCT03313778
BCG Bladder 1 NCT02808143
Intratumoral DC NHL 1, 2 NCT03035331
HSPPC-96 GBM 2 NCT03018288
Vigil TM bi-shRNAfurin-GM-CSF Melanoma 1 NCT02574533
DC Melanoma 1 NCT03092453
Anti-PD-L1
 Atezolizumab CDX-1401/NY-ESO-1 NSCLC 2 NCT02495636
Ovary, peritoneum 1, 2 NCT03206047
RO7198457 Multiple solid tumors 1 NCT03289962
 Avelumab TG4001 (MVA-HPV-IL2) SCCHN 1, 2 NCT03260023
PVX-410 (tumor antigen peptides) MM 1 NCT02886065
 Durvalumab PVX-410 (tumor antigen peptides) TNBC 1 NCT02826434
Neoantigen DNA TNBC 1 NCT03199040
TPIV200/huFR-1/Folate Receptor Ovary 2 NCT02764333
PVX-410 (tumor antigen peptides) Smoldering myeloma 1 NCT02886065
INO3112/HPV DNA and IL-12 HPV16 and 18 SCCHN 1, 2 NCT03162224
DC/AML fusion cell AML 2 NCT03059485
BCG Bladder 2 NCT02901548
Anti-DLL1/PD-1
 CT-011 DC fusion cells Myeloma, AML 2 NCT01067287, NCT01096602
Two or more checkpoint inhibitors
 Ipi, Nivo GVAX Pancreas 2 NCT03190265
 Ipi, Nivo, Pem HAM (dorgenmeltucel-L) Melanoma 2 NCT02054520
 Ipi, Nivo NY-ESO-1, gp100 Melanoma 1 NCT01176474
 Tre, Dur Poly ICLC Multiple solid tumors 1, 2 NCT02643303
mRNA NSCLC 1, 2 NCT03164772
 Nivo, Ate DSP-7888/WT-1 Multiple solid tumors 1 NCT03311334
DC CRC 1, 2 NCT03152565

Summary and Conclusions

In conclusion, we propose that the previous failures of cancer vaccines to eradicate or control tumors was most often not due to failure to induce an adequate immune response, but rather the inability of that response to achieve its full potential in the face of a myriad of negative regulatory mechanisms invoked by the tumor to counteract the immune system. Most tumors that become clinically evident have already escaped natural immunosurveillance by exploiting host immune regulatory mechanisms that evolved to prevent autoimmunity and inflammation. As the tumor cells are largely self, anti-tumor immunity must overcome these barriers to autoimmunity, and tumors have taken advantage of these mechanism to escape such immunosurveillance. In addition, other aspects of the tumor microenvironment may be hostile to infiltrating T cells, such as hypoxia or extracellular potassium ion concentrations [114]. Thus, such mechanisms are already in place the tumor microenvironment of clinically evident tumors, providing a barrier to the efficacy of tumor-specific T cells, whether the T cells are elicited by a vaccine or exogenously administered as adoptive T cell therapy. Overcoming these barriers with newly developed checkpoint inhibitors or inhibitors of regulatory cells may finally allow the immune response induced by a vaccine to achieve its full potential [107]. Most major efforts to improve the efficacy of cancer vaccine therapy are now focused on such combinations of vaccines with modalities to block checkpoints or remove or block activity of other negative regulatory cells, such as T-regulatory cells, myeloid-derived suppressor cells (MDSCs), regulatory type II NKT cells, M2 macrophages, or other regulatory mechanisms such as indoleamine oxidase (IDO) or cytokines like IL-10, IL-13 or TGF-beta.

We have also learned that while some tumors are naturally immunogenic, and have elicited immune responses that can function on their own as long as immune checkpoints or other regulators are blocked, other tumors (so-called “cold” tumors) [115,116] [6,117,118] fail to elicit immune responses on their own. This may be due to intrinsic lack of immunogenicity as in the case of tumors with low mutational burden [6] or active immunosuppression through activation of pathways such as beta-catenin signaling [118]. In these cases, vaccines can provide the immunogenicity that the tumors lack, inducing an immune response that can then function when combined with such inhibitors of immune regulation. Other interventions are now understood to also function at least in part by inducing an immune response, such as the abscopal effects of radiotherapy [119,120] or the immunogenic cell death induced by some types of chemotherapy [121123], and these modalities may also synergize with vaccines and with checkpoint inhibitors or other strategies to block immunoregulatory mechanisms. New targets for cancer vaccines, such as neo-antigens created by tumor-unique mutations [6,97], may facilitate the development of more effective cancer vaccines. Also, the recent discovery that tumor immunity is strongly influenced by the gut microbiome may provide another approach to potentiating the efficacy of cancer vaccines [124126]. With all of these approaches to improve the efficacy of cancer vaccines, cancer vaccines may finally be in a position to achieve their full potential and to take their rightful place as the third leg of the stool of cancer immunotherapy alongside adoptive cell therapy [8] and checkpoint inhibition that have recently been licensed as novel efficacious treatments for cancer.

Highlights.

  • Animal model successes have been translated to clinical trials from phase I to III.

  • Cancer vaccines have elicited antigen-specific T cells but limited efficacy alone.

  • Overcoming negative regulation may allow vaccine clinical efficacy.

  • Vaccine studies in combination with checkpoint inhibitors are ongoing.

  • Mutation-generated neoantigens provide novel target antigens for cancer vaccines.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

** Highlighted references published since 2015.

  • 1.Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–915. doi: 10.1038/nm1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Klebanoff CA, Finkelstein SE, Surman DR, Lichtman MK, Gattinoni L, Theoret MR, Grewal N, Spiess PJ, Antony PA, Palmer DC, et al. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc Natl Acad Sci U S A. 2004;101:1969–1974. doi: 10.1073/pnas.0307298101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–422. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
  • 4.Robert C, Thomas L, Bondarenko I, O'Day S, Weber J, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–2526. doi: 10.1056/NEJMoa1104621. [DOI] [PubMed] [Google Scholar]
  • 5.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **6.Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128. doi: 10.1126/science.aaa1348. This study describes anti-PD-1-unmasked neoantigen-specific T cell reactivity in non-small cell lung cancer patients as well as a molecular signature in smokers that was associated with improved PFS. It demonstrates that response to PD-1 blockade depends on the frequency of mutation-induced neo-epitopes, a finding of wide importance. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ, Restifo NP, Haworth LR, Seipp CA, Freezer LJ, et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci U S A. 2003;100:8372–8377. doi: 10.1073/pnas.1533209100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **8.Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–68. doi: 10.1126/science.aaa4967. This reference highlights adoptive cell transfer as a personalized strategy for treating human cancer with direct and durable anticancer activity and suggests genetic engineering of antigen receptors for further applications. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Yee C, Thompson JA, Byrd D, Riddell SR, Roche P, Celis E, Greenberg PD. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A. 2002;99:16168–16173. doi: 10.1073/pnas.242600099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **11.Gill S, June CH. Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies. Immunol Rev. 2015;263:68–89. doi: 10.1111/imr.12243. This article explains the concept of CAR T-cells and extant results in hematologic malignancies upon FDA “breakthrough therapy” designation of the anti-CD19 CAR T-cell therapy. [DOI] [PubMed] [Google Scholar]
  • 12.Le DT, Lutz E, Uram JN, Sugar EA, Onners B, Solt S, Zheng L, Diaz LA, Jr, Donehower RC, Jaffee EM, et al. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J Immunother. 2013;36:382–389. doi: 10.1097/CJI.0b013e31829fb7a2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **13.Terabe M, Robertson FC, Clark K, De Ravin E, Bloom A, Venzon D, Kato S, Mirza A, Berzofsky JA. Blockade of only TGF-β 1 and 2 is sufficient to enhance the efficacy of vaccine and PD-1 checkpoint blockade immunotherapy. OncoImmunology. 2017;6:e1308616. doi: 10.1080/2162402X.2017.1308616. This study reports that blockade of TGF-β1 and 2 is sufficient, without blocking TGF-β3, to enhance the efficacy of an HPV16-E7 peptide vaccine in a mouse model and was further potentiated when used with PD-1 blockade, suggesting that the two types of checkpoint inhibitors are complementary and can be used in combination. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Klebanoff CA, Gattinoni L, Torabi-Parizi P, Kerstann K, Cardones AR, Finkelstein SE, Palmer DC, Antony PA, Hwang ST, Rosenberg SA, et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci U S A. 2005;102:9571–9576. doi: 10.1073/pnas.0503726102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hwang LN, Yu Z, Palmer DC, Restifo NP. The in vivo expansion rate of properly stimulated transferred CD8+ T cells exceeds that of an aggressively growing mouse tumor. Cancer Res. 2006;66:1132–1138. doi: 10.1158/0008-5472.CAN-05-1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Klebanoff CA, Gattinoni L, Palmer DC, Muranski P, Ji Y, Hinrichs CS, Borman ZA, Kerkar SP, Scott CD, Finkelstein SE, et al. Determinants of successful CD8+ T-cell adoptive immunotherapy for large established tumors in mice. Clin Cancer Res. 2011;17:5343–5352. doi: 10.1158/1078-0432.CCR-11-0503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Terabe M, Matsui S, Park J-M, Mamura M, Noben-Trauth N, Donaldson DD, Chen W, Wahl SM, Ledbetter S, Pratt B, et al. Transforming Growth Factor-b production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block Cytotoxic T Lymphocyte-mediated tumor immunosurveillance: abrogation prevents tumor recurrence. J Exp Med. 2003;198:1741–1752. doi: 10.1084/jem.20022227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Terabe M, Ambrosino E, Takaku S, O'Konek JJ, Venzon D, Lonning S, McPherson JM, Berzofsky JA. Synergistic enhancement of CD8+ T cell-mediated tumor vaccine efficacy by an anti-transforming growth factor-beta monoclonal antibody. Clin Cancer Res. 2009;15:6560–6569. doi: 10.1158/1078-0432.CCR-09-1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Takaku S, Terabe M, Ambrosino E, Peng J, Lonning S, McPherson JM, Berzofsky JA. Blockade of TGF-beta enhances tumor vaccine efficacy mediated by CD8(+) T cells. Int J Cancer 2009. 2010;126:1666–1674. doi: 10.1002/ijc.24961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Vicari AP, Trinchieri G. Interleukin-10 in viral diseases and cancer: exiting the labyrinth? Immunol Rev. 2004;202:223–236. doi: 10.1111/j.0105-2896.2004.00216.x. [DOI] [PubMed] [Google Scholar]
  • 21.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155:1151–1164. [PubMed] [Google Scholar]
  • 22.Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol. 2004;22:531–562. doi: 10.1146/annurev.immunol.21.120601.141122. [DOI] [PubMed] [Google Scholar]
  • 23.Shevach EM. CD4+ CD25+ suppressor T cells: more questions than answers. Nat Rev Immunol. 2002;2:389–400. doi: 10.1038/nri821. [DOI] [PubMed] [Google Scholar]
  • 24.Bronte V, Wang M, Overwijk WW, Surman DR, Pericle F, Rosenberg SA, Restifo NP. Apoptotic death of CD8+ T lymphocytes after immunization: induction of a suppressive population of Mac-1+/Gr-1+ cells. J Immunol. 1998;15:5313–5320. [PMC free article] [PubMed] [Google Scholar]
  • 25.Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001;166:5398–5406. doi: 10.4049/jimmunol.166.9.5398. [DOI] [PubMed] [Google Scholar]
  • 26.Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–174. doi: 10.1038/nri2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ostrand-Rosenberg S, Sinha P, Beury DW, Clements VK. Cross-talk between myeloid-derived suppressor cells (MDSC), macrophages, and dendritic cells enhances tumor-induced immune suppression. Semin Cancer Biol. 2012;22:275–281. doi: 10.1016/j.semcancer.2012.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Terabe M, Berzofsky JA. The role of NKT cells in tumor immunity. Adv Cancer Res. 2008;101:277–348. doi: 10.1016/S0065-230X(08)00408-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Luo Y, Zhou H, Krueger J, Kaplan C, Lee SH, Dolman C, Markowitz D, Wu W, Liu C, Reisfeld RA, et al. Targeting tumor-associated macrophages as a novel strategy against breast cancer. J Clin Invest. 2006;116:2132–2141. doi: 10.1172/JCI27648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sinha P, Clements VK, Ostrand-Rosenberg S. Reduction of myeloid-derived suppressor cells and induction of M1 macrophages facilitate the rejection of established metastatic disease. J Immunol. 2005;174:636–645. doi: 10.4049/jimmunol.174.2.636. [DOI] [PubMed] [Google Scholar]
  • 31.Bodogai M, Moritoh K, Lee-Chang C, Hollander CM, Sherman-Baust CA, Wersto RP, Araki Y, Miyoshi I, Yang L, Trinchieri G, et al. Immune suppressive and pro-metastatic functions of myeloid-derived suppressive cells rely upon education from tumor-associated B cells. Cancer Res. 2015;75:3456–3465. doi: 10.1158/0008-5472.CAN-14-3077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Germain RN, Margulies DH. The biochemistry and cell biology of antigen processing and presentation. AnnuRev Immunol. 1993;11:403–450. doi: 10.1146/annurev.iy.11.040193.002155. [DOI] [PubMed] [Google Scholar]
  • 33.Sette A, Vitiello A, Reherman B, Fowler P, Nayersina R, Kast WM, Melief CJM, Oseroff C, Yuan L, Ruppert J, et al. The relationship between class I binding affinity and immunogenicity of potential cytotoxic T cell epitopes. J Immunol. 1994;153:5586–5592. [PubMed] [Google Scholar]
  • 34.Berzofsky JA. Epitope selection and design of synthetic vaccines: molecular approaches to enhancing immunogenicity and crossreactivity of engineered vaccines. Ann NY Acad Sci. 1993;690:256–264. doi: 10.1111/j.1749-6632.1993.tb44014.x. [DOI] [PubMed] [Google Scholar]
  • 35.Berzofsky JA. Designing peptide vaccines to broaden recognition and enhance potency. Ann NY Acad Sci. 1995;754:161–168. doi: 10.1111/j.1749-6632.1995.tb44449.x. [DOI] [PubMed] [Google Scholar]
  • 36.Ahlers JD, Takeshita T, Pendleton CD, Berzofsky JA. Enhanced immunogenicity of HIV-1 vaccine construct by modification of the native peptide sequence. Proc Natl Acad Sci US A. 1997;94:10856–10861. doi: 10.1073/pnas.94.20.10856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Parkhurst MR, Salgaller ML, Southwood S, Robbins PF, Sette A, Rosenberg SA, Kawakami Y. Improved induction of melanoma-reactive CTL with peptides from the melanoma antigen gp100 modified at HLA-A*0201-binding residues. J Immunol. 1996;157:2539–2548. [PubMed] [Google Scholar]
  • 38.Overwijk WW, Tsung A, Irvine KR, Parkhurst MR, Goletz TJ, Tsung K, Carroll MW, Liu C, Moss B, Rosenberg SA, et al. gp100/pmel 17 is a murine tumor rejection antigen: induction of “self”-reactive, tumoricidal T cells using high-affinity, altered peptide ligand. J Exp Med. 1998;188:277–286. doi: 10.1084/jem.188.2.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Salazar E, Zaremba S, Arlen PM, Tsang KY, Schlom J. Agonist peptide from a cytotoxic t-lymphocyte epitope of human carcinoembryonic antigen stimulates production of tc1-type cytokines and increases tyrosine phosphorylation more efficiently than cognate peptide. Int J Cancer. 2000;85:829–838. doi: 10.1002/(sici)1097-0215(20000315)85:6<829::aid-ijc16>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
  • 40.Wolfgang CD, Essand M, Vincent JJ, Lee B, Pastan I. TARP: a nuclear protein expressed in prostate and breast cancer cells derived from an alternate reading frame of the T cell receptor gamma chain locus. Proc Natl Acad Sci U S A. 2000;97:9437–9442. doi: 10.1073/pnas.160270597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Oh S, Terabe M, Pendleton CD, Bhattacharyy A, Bera TK, Epel M, Reiter Y, Phillips J, Linehan WM, Kasten-Sportes C, et al. Human CTL to wild type and enhanced epitopes of a novel prostate and breast tumor-associated protein, TARP, lyse human breast cancer cells. Cancer Research. 2004;64:2610–2618. doi: 10.1158/0008-5472.can-03-2183. [DOI] [PubMed] [Google Scholar]
  • 42.Pound CR, Partin AW, Eisenberger MA, Chan DW, Pearson JD, Walsh PC. Natural history of progression after PSA elevation following radical prostatectomy. JAMA. 1999;281:1591–1597. doi: 10.1001/jama.281.17.1591. [DOI] [PubMed] [Google Scholar]
  • 43.Slovin SF, Wilton AS, Heller G, Scher HI. Time to detectable metastatic disease in patients with rising prostate-specific antigen values following surgery or radiation therapy. Clin Cancer Res. 2005;11:8669–8673. doi: 10.1158/1078-0432.CCR-05-1668. [DOI] [PubMed] [Google Scholar]
  • 44.Lee AK, Levy LB, Cheung R, Kuban D. Prostate-specific antigen doubling time predicts clinical outcome and survival in prostate cancer patients treated with combined radiation and hormone therapy. Int J Radiat Oncol Biol Phys. 2005;63:456–462. doi: 10.1016/j.ijrobp.2005.03.008. [DOI] [PubMed] [Google Scholar]
  • 45.Freedland SJ, Humphreys EB, Mangold LA, Eisenberger M, Dorey FJ, Walsh PC, Partin AW. Risk of prostate cancer-specific mortality following biochemical recurrence after radical prostatectomy. JAMA. 2005;294:433–439. doi: 10.1001/jama.294.4.433. [DOI] [PubMed] [Google Scholar]
  • 46.Freedland SJ, Humphreys EB, Mangold LA, Eisenberger M, Dorey FJ, Walsh PC, Partin AW. Death in patients with recurrent prostate cancer after radical prostatectomy: prostate-specific antigen doubling time subgroups and their associated contributions to all-cause mortality. J Clin Oncol. 2007;25:1765–1771. doi: 10.1200/JCO.2006.08.0572. [DOI] [PubMed] [Google Scholar]
  • 47.Antonarakis ES, Zahurak ML, Lin J, Keizman D, Carducci MA, Eisenberger MA. Changes in PSA kinetics predict metastasis- free survival in men with PSA-recurrent prostate cancer treated with nonhormonal agents: combined analysis of 4 phase II trials. Cancer. 2012;118:1533–1542. doi: 10.1002/cncr.26437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **48.Wood LV, Fojo A, Roberson BD, Hughes MSB, Dahut W, Gulley JL, Madan RA, Arlen PM, Sabatino M, Stroncek DF, et al. TARP vaccination is associated with slowing in PSA velocity and decreasing tumor growth rates in patients with Stage D0 prostate cancer. Oncolmmunology. 2016;5:e1197459. doi: 10.1080/2162402X.2016.1197459. This study reports results of a Phase I clinical trial at NCI translated from mice that showed significiant slowing the PSA velocity and tumor growth rate in 74% of stage D0 prostate cancer patients after epitope-enhanced vaccination targeting TARP. a prostate cancer antigen. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–792. doi: 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
  • 50.Kaufman B, Mackey JR, Clemens MR, Bapsy PP, Vaid A, Wardley A, Tjulandin S, Jahn M, Lehle M, Feyereislova A, et al. Trastuzumab plus anastrozole versus anastrozole alone for the treatment of postmenopausal women with human epidermal growth factor receptor 2-positive, hormone receptor-positive metastatic breast cancer: results from the randomized phase III TAnDEM study. J Clin Oncol. 2009;27:5529–5537. doi: 10.1200/JCO.2008.20.6847. [DOI] [PubMed] [Google Scholar]
  • 51.Johnston SR, Gomez H, Stemmer SM, Richie M, Durante M, Pandite L, Goodman V, Slamon D. A randomized and open-label trial evaluating the addition of pazopanib to lapatinib as first-line therapy in patients with HER2-positive advanced breast cancer. Breast Cancer Res Treat. 2013;137:755–766. doi: 10.1007/s10549-012-2399-4. [DOI] [PubMed] [Google Scholar]
  • 52.Sakai Y, Morrison BJ, Burke JD, Park JM, Terabe M, Janik JE, Forni G, Berzofsky JA, Morris JC. Vaccination by Genetically Modified Dendritic Cells Expressing a Truncated neu Oncogene Prevents Development of Breast Cancer in Transgenic Mice. Cancer Research. 2004;64:8022–8028. doi: 10.1158/0008-5472.CAN-03-3442. [DOI] [PubMed] [Google Scholar]
  • 53.Park JM, Terabe M, Sakai Y, Munasinghe J, Forni G, Morris JC, Berzofsky JA. Early Role of CD4+ Th1 cells and antibodies in HER-2 adenovirus-vaccine protection against autochthonous mammary carcinomas. J Immunol. 2005;174:4228–4236. doi: 10.4049/jimmunol.174.7.4228. [DOI] [PubMed] [Google Scholar]
  • 54.Park JM, Terabe M, Steel JC, Forni G, Sakai Y, Morris JC, Berzofsky JA. Therapy of advanced established murine breast cancer with a recombinant adenoviral ErbB-2/neu vaccine. Cancer Res. 2008;68:1979–1987. doi: 10.1158/0008-5472.CAN-07-5688. [DOI] [PubMed] [Google Scholar]
  • 55.Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat Med. 2000;6:443–446. doi: 10.1038/74704. [DOI] [PubMed] [Google Scholar]
  • 56.Wood LV, Roberson BD, Agarwal PK, Apolo AB, Stroncek D, Weiner LM, Morris JC, Terabe M, Lee M-J, Trepel JB, et al. Association of autologous AdHER2 dendritic cell vaccination with antitumor activity and number of circulating tumor cells. Journal of Clinical Oncology. 2017;35:3089–3089. [Google Scholar]
  • 57.Knutson KL, Schiffman K, Disis ML. Immunization with a HER-2/neu helper peptide vaccine generates HER- 2/neu CD8 T-cell immunity in cancer patients. J Clin Invest. 2001;107:477–484. doi: 10.1172/JCI11752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Holmes JP, Gates JD, Benavides LC, Hueman MT, Carmichael MG, Patil R, Craig D, Mittendorf EA, Stojadinovic A, Ponniah S, et al. Optimal dose and schedule of an HER-2/neu (E75) peptide vaccine to prevent breast cancer recurrence: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02. Cancer. 2008;113:1666–1675. doi: 10.1002/cncr.23772. [DOI] [PubMed] [Google Scholar]
  • 59.Bernards R, Destree A, McKenzie S, Gordon E, Weinberg RA, Panicali D. Effective tumor immunotherapy directed against an oncogene-encoded product using a vaccinia virus vector. Proc Natl Acad Sci U S A. 1987;84:6854–6858. doi: 10.1073/pnas.84.19.6854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Irvine KR, Parkhurst MR, Shulman EP, Tupesis JP, Custer M, Touloukian CE, Robbins PF, Yafal AG, Greenhalgh P, Sutmuller RP, et al. Recombinant virus vaccination against “self” antigens using anchor- fixed immunogens. Cancer Res. 1999;59:2536–2540. [PMC free article] [PubMed] [Google Scholar]
  • 61.Kass E, Schlom J, Thompson J, Guadagni F, Graziano P, Greiner JW. Induction of protective host immunity to carcinoembryonic antigen (CEA), a self-antigen in CEA transgenic mice by immunizing with a recombinant vaccinia-CEA virus. Cancer Res. 1999;59:676–683. [PubMed] [Google Scholar]
  • 62.Hodge JW, McLaughlin JP, Kantor JA, Schlom J. Diversified prime and boost protocols using recombinant vaccinia virus and recombinant non-replicating avian pox virus to enhance T-cell immunity and antitumor responses. Vaccine. 1997;15:759–768. doi: 10.1016/s0264-410x(96)00238-1. [DOI] [PubMed] [Google Scholar]
  • 63.Dale CJ, Thomson S, De Rose R, Ranasinghe C, Medveczky CJ, Pamungkas J, Boyle DB, Ramshaw IA, Kent SJ. Prime-boost strategies in DNA vaccines. Methods Mol Med. 2006;127:171–197. doi: 10.1385/1-59745-168-1:171. [DOI] [PubMed] [Google Scholar]
  • 64.Hodge JW, Sabzevari H, Yafal AG, Gritz L, Lorenz MGO, Schlom J. A triad of costimulatory molecules synergize to amplify T-cell activation. Cancer Res. 1999;59:5800–5807. [PubMed] [Google Scholar]
  • 65.Oh S, Hodge JW, Ahlers JD, Burke DS, Schlom J, Berzofsky JA. Selective induction of high avidity CTL by altering the balance of signals from antigen presenting cells. J Immunol. 2003;170:2523–2530. doi: 10.4049/jimmunol.170.5.2523. [DOI] [PubMed] [Google Scholar]
  • 66.Hodge JW, Chakraborty M, Kudo-Saito C, Garnett CT, Schlom J. Multiple costimulatory modalities enhance CTL avidity. J Immunol. 2005;174:5994–6004. doi: 10.4049/jimmunol.174.10.5994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gulley JL, Arlen PM, Madan RA, Tsang KY, Pazdur MP, Skarupa L, Jones JL, Poole DJ, Higgins JP, Hodge JW, et al. Immunologic and prognostic factors associated with overall survival employing a poxviral-based PSA vaccine in metastatic castrate-resistant prostate cancer. Cancer Immunol Immunother. 2010;59:663–674. doi: 10.1007/s00262-009-0782-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Schoenberger SP, Toes REM, van der Voort EIH, Offringa R, Melief CJM. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature. 1998;393:480–483. doi: 10.1038/31002. [DOI] [PubMed] [Google Scholar]
  • 69.Ridge JP, Di Rosa F, Matzinger P. A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell. Nature. 1998;393:474–478. doi: 10.1038/30989. [DOI] [PubMed] [Google Scholar]
  • 70.Bennett SRM, Carbone FR, Karamalis F, Flavell RA, Miller JFAP, Heath WR. Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature. 1998;393:478–480. doi: 10.1038/30996. [DOI] [PubMed] [Google Scholar]
  • 71.Janssen EM, Lemmens EE, Wolfe T, Christen U, von Herrath MG, Schoenberger SP. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature. 2003;421:852–856. doi: 10.1038/nature01441. [DOI] [PubMed] [Google Scholar]
  • 72.Janssen EM, Droin NM, Lemmens EE, Pinkoski MJ, Bensinger SJ, Ehst BD, Griffith TS, Green DR, Schoenberger SP. CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-mediated activation-induced cell death. Nature. 2005;434:88–93. doi: 10.1038/nature03337. [DOI] [PubMed] [Google Scholar]
  • 73.Zwaveling S, Ferreira Mota SC, Nouta J, Johnson M, Lipford GB, Offringa R, van der Burg SH, Melief CJ. Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J Immunol. 2002;169:350–358. doi: 10.4049/jimmunol.169.1.350. [DOI] [PubMed] [Google Scholar]
  • 74.van der Burg SH, Bijker MS, Welters MJ, Offringa R, Melief CJ. Improved peptide vaccine strategies, creating synthetic artificial infections to maximize immune efficacy. Adv Drug Deliv Rev. 2006;58:916–930. doi: 10.1016/j.addr.2005.11.003. [DOI] [PubMed] [Google Scholar]
  • 75.Melief CJ, van der Burg SH. Immunotherapy of established (pre)malignant disease by synthetic long peptide vaccines. Nat Rev Cancer. 2008;8:351–360. doi: 10.1038/nrc2373. [DOI] [PubMed] [Google Scholar]
  • 76.Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, Drijfhout JW, Wafelman AR, Oostendorp J, Fleuren GJ, et al. Phase I immunotherapeutic trial with long peptides spanning the E6 and E7 sequences of high-risk human papillomavirus 16 in end-stage cervical cancer patients shows low toxicity and robust immunogenicity. Clin Cancer Res. 2008;14:169–177. doi: 10.1158/1078-0432.CCR-07-1881. [DOI] [PubMed] [Google Scholar]
  • 77.Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, Essahsah F, Fathers LM, Offringa R, Drijfhout JW, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361:1838–1847. doi: 10.1056/NEJMoa0810097. [DOI] [PubMed] [Google Scholar]
  • 78.Melief CJ, Gerritsen WR, Welters M, Vergote I, Kroep JR, Kenter G, Ottevanger PB, Tjalma WA, Denys H, Nijman H, van Poelgeest MI, Reyners AK, Velu TJ, Blumenstein BA, Goffin F, Lalisang FI, Stead RB, van der Burg S. Correlation between strength of T-cell response against HPV16 and survival after vaccination with HPV16 long peptides in combination with chemotherapy for late-stage cervical cancer. Journal of Clinical Oncology; 2017 ASCO-SITC Clinical Immuno-Oncology Symposium; 02/23/2017; 2017. Abstract 140. [Google Scholar]
  • **79.Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, Edwards L, Parker RL, Denny L, Giffear M, et al. Safety, efficacy, and immunogenicity of VGX-3100; a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet. 2015;386:2078–2088. doi: 10.1016/S0140-6736(15)00239-1. This study presents the first therapeutic vaccine to show efficacy against HPV-16/18-associated CIN2/3, with 48–49% of vaccinees showing histopathological regression to CIN1 or better compared to 30% of controls (p = 0.03) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Magarian-Blander J, Ciborowski P, Hsia S, Watkins SC, Finn OJ. Intercellular and intracellular events following the MHC-unrestricted TCR recognition of a tumor-specific peptide epitope on the epithelial antigen MUC1. J Immunol. 1998;160:3111–3120. [PubMed] [Google Scholar]
  • 81.Alajez NM, Schmielau J, Alter MD, Cascio M, Finn OJ. Therapeutic potential of a tumor-specific, MHC-unrestricted T-cell receptor expressed on effector cells of the innate and the adaptive immune system through bone marrow transduction and immune reconstitution. Blood. 2005;105:4583–4589. doi: 10.1182/blood-2004-10-3848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Vella LA, Yu M, Fuhrmann SR, El-Amine M, Epperson DE, Finn OJ. Healthy individuals have T-cell and antibody responses to the tumor antigen cyclin B1 that when elicited in mice protect from cancer. Proc Natl Acad Sci U S A. 2009;106:14010–14015. doi: 10.1073/pnas.0903225106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Finn OJ, Forni G. Prophylactic cancer vaccines. Curr Opin Immunol. 2002;14:172–177. doi: 10.1016/s0952-7915(02)00317-5. [DOI] [PubMed] [Google Scholar]
  • 84.Finn OJ. Cancer immunology. N Engl J Med. 2008;358:2704–2715. doi: 10.1056/NEJMra072739. [DOI] [PubMed] [Google Scholar]
  • 85.Kimura T, McKolanis JR, Dzubinski LA, Islam K, Potter DM, Salazar AM, Schoen RE, Finn OJ. MUC1 vaccine for individuals with advanced adenoma of the colon: a cancer immunoprevention feasibility study. Cancer Prev Res (Phila) 2013;6:18–26. doi: 10.1158/1940-6207.CAPR-12-0275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kwak LW, Campbell MJ, Czerwinski DK, Hart S, Miller RA, Levy R. Induction of immune responses in patients with B-cell lymphoma against the surface-immunoglobulin idiotype expressed by their tumors. N Engl J Med. 1992;327:1209–1215. doi: 10.1056/NEJM199210223271705. [DOI] [PubMed] [Google Scholar]
  • 87.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;5:1171–1177. doi: 10.1038/13928. [DOI] [PubMed] [Google Scholar]
  • 88.Freedman A, Neelapu SS, Nichols C, Robertson MJ, Djulbegovic B, Winter JN, Bender JF, Gold DP, Ghalie RG, Stewart ME, et al. Placebo-controlled phase III trial of patient-specific immunotherapy with mitumprotimut-T and granulocyte-macrophage colony-stimulating factor after rituximab in patients with follicular lymphoma. J Clin Oncol. 2009;27:3036–3043. doi: 10.1200/JCO.2008.19.8903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Levy R, Ganjoo KN, Leonard JP, Vose JM, Flinn IW, Ambinder RF, Connors JM, Berinstein NL, Belch AR, Bartlett NL, et al. Active idiotypic vaccination versus control immunotherapy for follicular lymphoma. J Clin Oncol. 2014;32:1797–1803. doi: 10.1200/JCO.2012.43.9273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Schuster SJ, Neelapu SS, Gause BL, Janik JE, Muggia FM, Gockerman JP, Winter JN, Flowers CR, Nikcevich DA, Sotomayor EM, et al. Vaccination with patient-specific tumor-derived antigen in first remission improves disease-free survival in follicular lymphoma. J Clin Oncol. 2011;29:2787–2794. doi: 10.1200/JCO.2010.33.3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, Mellman I, Prindiville SA, Viner JL, Weiner LM, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15:5323–5337. doi: 10.1158/1078-0432.CCR-09-0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Oka Y, Udaka K, Tsuboi A, Elisseeva OA, Ogawa H, Aozasa K, Kishimoto T, Sugiyama H. Cancer immunotherapy targeting Wilms' tumor gene WT1 product. J Immunol. 2000;164:1873–1880. doi: 10.4049/jimmunol.164.4.1873. [DOI] [PubMed] [Google Scholar]
  • **93.Qazilbash MH, Wieder E, Thall PF, Wang X, Rios R, Lu S, Kanodia S, Ruisaard KE, Giralt SA, Estey EH, et al. PR1 peptide vaccine induces specific immunity with clinical responses in myeloid malignancies. Leukemia. 2017;31:697–704. doi: 10.1038/leu.2016.254. This reference is the report of a phase 2 trial with a PR1 peptide vaccine in recurrent or high-risk myeloid malignancies showing safety and efficacy. Clinical benefit correlated with the degree of T cell response, demonstrating clinical efficacy of a human cancer vaccine. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **94.Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S, Prickett TD, Gartner JJ, Crystal JS, Roberts IM, et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med. 2016;22:433–438. doi: 10.1038/nm.4051. This article describes the novel noninvasive strategy of using PD-1 staining of peripheral blood CD8+ lymphocytes as an activation biomarker for cells reactive to patient-specific neoantigens in melanoma patients, without the need for a biopsy to isolate TILs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **95.Stevanovic S, Pasetto A, Helman SR, Gartner JJ, Prickett TD, Howie B, Robins HS, Robbins PF, Klebanoff CA, Rosenberg SA, et al. Landscape of immunogenic tumor antigens in successful immunotherapy of virally induced epithelial cancer. Science. 2017;356:200–205. doi: 10.1126/science.aak9510. This study describes the surprising finding after adoptive tumor-infiltrating T cell therapy for advanced HPV-associated cervical cancer that immunodominant T cell reactivities were against mutated neoantigens rather than canonical foreign HPV viral antigens. In both situations, T cells were PD-1–positive suggesting the potential of PD-1 blockade for diverse antitumor T cell reactivities. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, Ivanova Y, Hundal J, Arthur CD, Krebber WJ, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515:577–581. doi: 10.1038/nature13988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **97.Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74. doi: 10.1126/science.aaa4971. This article reviews the mutation-derived neoantigens in human cancer and points out the potential of engineered therapeutic interventions targeting neoantigen-specific T cell reactivity to enhance specificity, safety and efficacy of anti-cancer immunotherapy. [DOI] [PubMed] [Google Scholar]
  • 98.Yanuck M, Carbone DP, Pendleton CD, Tsukui T, Winter SF, Minna JD, Berzofsky JA. A mutant p53 tumor suppressor protein is a target for peptide-induced CD8+ cytotoxic T cells. Cancer Res. 1993;53:3257–3261. [PubMed] [Google Scholar]
  • 99.Carbone DP, Ciernik IF, Yanuck M, Smith MC, Kelley M, Johnson BE, Allegra C, Pendleton CD, Khleif S, Kaye F, et al. Mutant p53 and ras proteins as immunotherapeutic targets. Annals of Oncology. 1994;5:117–110. [Google Scholar]
  • 100.Smith MC, Pendleton CD, Maher VE, Kelley MJ, Carbone DP, Berzofsky JA. Oncogenic mutations in ras create HLA-A2.1 binding peptides but affect their extracellular processing. Internat Immunol. 1997;9:1085–1093. doi: 10.1093/intimm/9.8.1085. [DOI] [PubMed] [Google Scholar]
  • 101.Abrams SI, Hand PH, Tsang KY, Schlom J. Mutant ras epitopes as targets for cancer vaccines. Semin Oncolo. 1996;23:118–134. [PubMed] [Google Scholar]
  • 102.Khleif SN, Abrams SI, Hamilton JM, Bergmann-Leitner E, Chen A, Bastian A, Bernstein S, Chung Y, Allegral CJ, Schlom J. A phase I vaccine trial with peptides reflecting ras oncogene mutations of solid tumors. J Immunotherapy. 1999;22:155–165. doi: 10.1097/00002371-199903000-00007. [DOI] [PubMed] [Google Scholar]
  • 103.Carbone DP, Ciernik IF, Kelley MJ, Smith MC, Nadaf S, Kavanaugh D, Maher VE, Stipanov M, Contois D, Johnson BE, et al. Immunization With Mutant p53- and K-ras-Derived Peptides in Cancer Patients: Immune Response and Clinical Outcome. J Clin Oncol. 2005;23:5099–5107. doi: 10.1200/JCO.2005.03.158. [DOI] [PubMed] [Google Scholar]
  • **104.Tran E, Robbins PF, Lu YC, Prickett TD, Gartner JJ, Jia L, Pasetto A, Zheng Z, Ray S, Groh EM, et al. T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. N Engl J Med. 2016;375:2255–2262. doi: 10.1056/NEJMoa1609279. This study describes a patient with metastatic colorectal cancer harboring a KRAS G12D mutation treated with tumor-infiltrating lymphocytes, including T cells found to be specific for Kras G12D presented by HLA-Cw08. The patient had a partial response for 9 months until a single progressive lung lesion was found to have loss of chromosome 6 haplotype that is the locus for the HLA-Cw08 MHC class I molecule. This result helps validate mutant ras as a tumor neoantigen. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **105.Parchment RE, Voth AR, Doroshow JH, Berzofsky JA. Immuno-pharmacodynamics for evaluating mechanism of action and developing immunotherapy combinations. Semin Oncol. 2016;43:501–513. doi: 10.1053/j.seminoncol.2016.06.008. This reference reviews pharmacodynamic biomarkers in cancer immunotherapy with a focus on the use of immunotherapy combinations in development, including cancer vaccines with checkpoint inhibitors. [DOI] [PubMed] [Google Scholar]
  • **106.Gatti-Mays ME, Redman JM, Collins JM, Bilusic M. Cancer Vaccines: Enhanced Immunogenic Modulation through Therapeutic Combinations. Hum Vaccin Immunother. 2017 doi: 10.1080/21645515.2017.1364322. in press. This reference is a comprehensive review detailing why and how combination strategies with cancer vaccines can lead to enhanced efficacy in tackling cancer to overcome previously identified clinical limitations. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61. doi: 10.1126/science.aaa8172. [DOI] [PubMed] [Google Scholar]
  • 108.Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, Roddie C, Henry JY, Yagita H, Wolchok JD, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210:1695–1710. doi: 10.1084/jem.20130579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **109.Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017;355:1428–1433. doi: 10.1126/science.aaf1292. This study demonstrates that the co-receptor CD28 is preferred over the TCR as a target in response to PD-1 activation by PD-L1 in an in vitro binding system. It implies that the PD-1 signal through SHP2 may target costimulation more than TCR signaling. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Madan RA, Mohebtash M, Arlen PM, Vergati M, Rauckhorst M, Steinberg SM, Tsang KY, Poole DJ, Parnes HL, Wright JJ, et al. Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:501–508. doi: 10.1016/S1470-2045(12)70006-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **111.Wilgenhof S, Corthals J, Heirman C, van Baren N, Lucas S, Kvistborg P, Thielemans K, Neyns B. Phase II Study of Autologous Monocyte-Derived mRNA Electroporated Dendritic Cells (TriMixDC-MEL) Plus Ipilimumab in Patients With Pretreated Advanced Melanoma. J Clin Oncol. 2016;34:1330–1338. doi: 10.1200/JCO.2015.63.4121. This study reports efficacy of a combination therapy using an RNA-transduced dendritic cell vaccine with CTLA-4 blockade, showing a 38% response rate including 8 complete & 7 partial responses among 39 vaccinees. [DOI] [PubMed] [Google Scholar]
  • 112.Gibney GT, Kudchadkar RR, DeConti RC, Thebeau MS, Czupryn MP, Tetteh L, Eysmans C, Richards A, Schell MJ, Fisher KJ, et al. Safety, correlative markers, and clinical results of adjuvant nivolumab in combination with vaccine in resected high-risk metastatic melanoma. Clin Cancer Res. 2015;21:712–720. doi: 10.1158/1078-0432.CCR-14-2468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Glisson B, Massarelli E, William WN, Johnson FM, Kies MS, Ferrarotto R, Guo M, Peng SA, Lee JJ, Tran H, et al. 1136ONivolumab and ISA 101 HPV vaccine in incurable HPV-16+ cancer. Annals of Oncology. 2017;28 mdx376.002-mdx376.002. [Google Scholar]
  • **114.Eil R, Vodnala SK, Clever D, Klebanoff CA, Sukumar M, Pan JH, Palmer DC, Gros A, Yamamoto TN, Patel SJ, et al. Ionic immune suppression within the tumour microenvironment limits T cell effector function. Nature. 2016;537:539–543. doi: 10.1038/nature19364. This study describes the surprising finding that the high level of potassium ion in the tumor microenvironment is a powerful inhibitor of T cell function in the tumor, in addition to any immune checkpoints or other inhibitory mechanisms. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177. [DOI] [PubMed] [Google Scholar]
  • 116.Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, Tosolini M, Camus M, Berger A, Wind P, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–1964. doi: 10.1126/science.1129139. [DOI] [PubMed] [Google Scholar]
  • 117.Fox BA, Schendel DJ, Butterfield LH, Aamdal S, Allison JP, Ascierto PA, Atkins MB, Bartunkova J, Bergmann L, Berinstein N, et al. Defining the critical hurdles in cancer immunotherapy. J Transl Med. 2011;9:214. doi: 10.1186/1479-5876-9-214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **118.Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–235. doi: 10.1038/nature14404. This study reveals a mechanism of melanoma resistance to PD-1/PD-L1 blockade and of inhibition of T cell infiltration through activation of the Wnt/beta-catenin pathway. [DOI] [PubMed] [Google Scholar]
  • 119.Pilones KA, Kawashima N, Yang AM, Babb JS, Formenti SC, Demaria S. Invariant natural killer T cells regulate breast cancer response to radiation and CTLA-4 blockade. Clin Cancer Res. 2009;15:597–606. doi: 10.1158/1078-0432.CCR-08-1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, Inghirami G, Coleman CN, Formenti SC, Demaria S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 2017;8:15618. doi: 10.1038/ncomms15618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, Schmitt E, Hamai A, Hervas-Stubbs S, Obeid M, et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med. 2005;202:1691–1701. doi: 10.1084/jem.20050915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72. doi: 10.1146/annurev-immunol-032712-100008. [DOI] [PubMed] [Google Scholar]
  • **123.Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2016;17:97. doi: 10.1038/nri.2016.107. This review summarizes the mechanisms involved in immunogenic cell death by which killing of tumor cells in vivo can elicit immune responses that contribute to tumor control by other modalities such as chemotherapy and radiotherapy, as well as related mechanisms in infectious disease. [DOI] [PubMed] [Google Scholar]
  • 124.Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342:967–970. doi: 10.1126/science.1240527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **125.Zitvogel L, Galluzzi L, Viaud S, Vetizou M, Daillere R, Merad M, Kroemer G. Cancer and the gut microbiota: an unexpected link. Sci Transl Med. 2015;7:271ps271. doi: 10.1126/scitranslmed.3010473. This study shows the critical role of the gut microbiome in the response of cancers to chemotherapy and immunotherapy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **126.Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–1084. doi: 10.1126/science.aad1329. This study demonstrates a critical role for gut microbiota, especially Bacteroidales, in the efficacy of anti-CTLA-4 blockade, which is found not to work well in germ-free or antibiotic-treated mice. It raises concerns about antibiotic use during cancer therapy. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES