1,612
Views
27
CrossRef citations to date
0
Altmetric
Reviews

Novel GM-CSF-based vaccines: One small step in GM-CSF gene optimization, one giant leap for human vaccines

, , , &
Pages 3020-3028 | Received 02 May 2016, Accepted 30 Jul 2016, Published online: 26 Sep 2016

ABSTRACT

Granulocyte macrophage-colony stimulating factor (GM-CSF) is a potent immunomodulatory cytokine that is known to facilitate vaccine efficacy by promoting the development and prolongation of both humoral and cellular immunity. In the past years we have generated a novel codon-optimized GM-CSF gene as an adjuvant. The codon-optimized GM-CSF gene significantly increased protein expression levels in all cells tested and helped in generating a strong immune responses against HIV-1 Gag and HPV-associated cancer. Here, we review the literature dealing with the adjuvant activity of GM-CSF both in animal models and clinical trials. We anticipate that the codon-optimized GM-CSF gene offers a practical molecular strategy for potentiating immune responses to tumor cell-based vaccinations as well as other immunotherapeutic strategies.

Introduction

Immunotherapies have been utilized to treat cancer for more than a century. A few clinical studies have successfully demonstrated immune activation in the treatment of malignant tumors. Granulocyte–macrophage colony stimulating factor (GM-CSF) is one of the most potent molecules used in these studies.

Granulocyte–macrophage colony stimulating factor (GM-CSF) is a glycoprotein comprising 127 amino acids with 2 potential N-linked glycosylation sites.Citation1,2 GM-CSF was first identified to regulate cell survival, proliferation, and differentiation in granulocyte-macrophage populations.Citation3 GM-CSF also serves as a haematopoietic growth factor for erythroid, megakaryocyte and eosinophil progenitors. The effects of this growth factor on immune system have been studied in murine, ex vivo, and human models.Citation4

GM-CSF is an inflammatory cytokine produced by various cell types, including T cells, B cells, macrophages, mast cells, endothelial cells, fibroblasts, and adipocytes, in response to cytokine or inflammatory stimuli. GM-CSF enhances the function of antigen presenting cells (APCs) by maturing, activating, and conscripting dendritic cells (DCs), macrophages, monocytes and eosinophils.Citation5,6

Persistent inflammation, immunosuppression, and catabolism syndrome (PICS) describes the development of chronic critical illness (CCI)Citation7, which is characterized by increased risks of secondary infections, extended ICU and long-term acute facilities stays, long-term functional or mental impairment, and unexpectedly high post-hospital discharge mortality rates.Citation4 GM-CSF can stimulate and enhance the production and function of neutrophils and monocytes and is used in immunostimulating adjuvant therapies to fight immunosuppressive diseases,Citation4,8,9,10 such as late sepsis. Citation4 GM-CSF has been studied for its effects on sepsis. A GM-CSF-treated group showed remarkably greater increases in total leukocyte counts, enhanced clearance rates and better clinical outcomes. Furthermore, membrane-associated human leukocyte antigen receptors (mHLA-DR), a biomarker for immune suppression Citation11, expression levels increased.Citation4 Lower mHLA-DR expression levels typically indicate inefficient immune cell function.Citation12 This study showed that treatment with GM-CSF resulted in significantly improved clinical outcomes without harmful side effects, including a more rapid recovery from infections, decreased admission lengths, decreased mechanical-ventilation days, and decreased financial costs.Citation13

Disturbed GM-CSF expression has been attributed to the pathogenesis of autoimmune inflammatory diseases.Citation14 Several studies have shown that GMCSF has important therapeutic potential in several inflammatory diseases, especially rheumatoid arthritis (RA).Citation15 RA is characterized by chronic pain and destruction in afflicted joints and is induced by macrophages that produce a variety of inflammatory cytokines, such as TNFα, IL-1β, and IL-6.Citation16 GMCSF was one of the first cytokines detected at high levels in the synovial fluid of inflamed joints.Citation17,18 The correlation between GM-CSF and RA was observed after GM-CSF injections in a mouse model of collagen-induced arthritis (CIA) exacerbated the disease. Treatment of these arthritic mice with neutralizing antibodies against GM-CSF prevented disease progression.Citation19 Thus, the benefits for targeting GMCSF are obvious. The rapid and sustained clinical responses from early phase clinical trials of GMCSF antagonism in patients with RA seem assuring.Citation15

In 1991, GM-CSF was approved for neutropenia associated with stem cell transplantation by the US Food and Drug Administration. Available forms of GM-CSF include sargramostim and molgramostim. Because GM-CSF promotes the function of APCs, GM-CSF has been considered an immunostimulatory adjuvant in numerous clinical trials and has been used to speed up recovery in patients suffering from non-Hodgkin's lymphoma, acute lymphoblastic leukemia and Hodgkin's disease undergoing autologous stem cell transplantation.Citation20 In 1990, a study of Gray collie dogs with cyclic neutropenia, i.e., recurring episodes of neutropenia caused by mutations in the endocytosis gene AP3B1,Citation21 demonstrated the involvement of 3 cytokines, G-CSF, GM-CSF, and IL-3. The study revealed that GM-CSF and G-CSF increased neutrophil counts.Citation22 GM-CSF was also studied as a potential adjunct to immunosuppressive therapyCitation23 for severe aplastic anemia, which is characterized by deficiencies in haematopoietic cell lines due to damaged bone marrow stem cells. GM-CSF immunosuppressive therapy lowered infection complication rates and hospital days than standard therapy alone,Citation24,25 especially for severe aplastic anemia.Citation14

Adverse effects of GM-CSF are dose related. Appropriate and usual dose range being 5–10 micrograms/kg/day either by 4–6 h intravenous infusion or by subcutaneous injection. At such doses, adverse effects are almost reversible, mild-to-moderate in nature, occur in 20–30% of patients and usually comprise fever, myalgia, fatigue, diarrhea and injection site reaction (swelling and tenderness). A rare but significant side effect is thrombosis which rarely can result in pulmonary embolism or stroke. Similarly, a so-called “first-dose effect,” defined as a syndrome of hypoxia, hypotension and tachycardia after the initial but not subsequent doses of GM-CSF. Another serious, but very uncommon side effect of GM-CSF is “capillary leak syndrome” or “vascular leak syndrome.” It is characterized by the presence of 2 or more of the following 3 symptoms: hypotension, edema, and hypoalbuminemia due to the fluids within the vascular system (veins and capillaries) leaks into the tissue outside the bloodstream. Certain patient groups, for example those with myelodysplastic syndrome, acute myeloid leukemia, inflammatory disease and autoimmune thrombocytopenia, require careful clinical monitoring in order to avoid potential complications following the administration of GM-CSF.

GM-CSF in cell-based immunotherapy

GM-CSF advances the function of APCs.Citation5,6 Cytokines, such as IL-2Citation26,27 and GM-CSF,Citation28,29 have been applied in cancer treatment to elicit strong antitumor immune responses.Citation30 Treatments with irradiated GM-CSF-expressing B16 melanoma cells generated more potent and long-lasting anti-tumor responses than other immune modulating gene products. Subcutaneous or intradermal injections of GM-CSF-secreting tumor cells contributed to enhanced infiltration of APCs, such as DCs, macrophages, and granulocytes, which allowed CD4+ and CD8+ T cells to recognize circulating tumor-associated antigens.Citation29,31 DCs are considered as the most potent APCs within the immune system. The recruitment of professional APCs by GM-CSF augmented the intracellular environment for tumor antigen presentation.Citation31

GVAX, a cell-based immunotherapy, is a GM-CSF gene-transduced tumor vaccine. The application of the GVAX system (Cell Genesys, now Aduro Biotech, Berkeley, CA) in preclinical studies has shown no remarkable local or systemic toxicities at clinically relevant doses.Citation31 Murine tumor models and human clinical trials showed that GM-CSF-secreting tumor cells could be used to vaccinate patients with several tumor types, such as non-small cell lung carcinomasCitation32, prostate tumorsCitation33, pancreatic tumorsCitation34, leukemiaCitation35, melanomasCitation36, gliomasCitation37, cervical tumorsCitation38, and renal cell carcinomas.Citation39

In a GVAX prostate vaccine phase II study, 34 hormone refractory prostate cancer patients with metastatic bone diseases at baseline were treated. Nine of the 24 patients at lower boost dosages (41%) and 7 of the 10 patients at the higher boost dosages (70%) survived the disease at the 2-year follow up. Additionally, there was a longer median time to disease progression as measured by bone scans in patients receiving the higher doses of vaccine compared with those at the lower doses (140 d vs. 85 d).Citation40 In a renal cell carcinoma study, 4 patients received GVAX for 48 vaccinations without showing severe adverse events. Two of the 4 patients survived at 58+ and 40+ months after the initial vaccination. These results indicated that GVAX fundamentally enhanced the antitumor cellular and humoral immune responses and may have contributed to the relatively long survival rates of patients.Citation41

Clinical investigations with GVAX continue. While phase II studies have shown significant effects, many steps remain prior to therapeutic approval from the FDA. Prostate cancer has also been considered for tumor immunotherapy; these treatment principles could also be applied to many other tumor types. Citation42

Positive results from phase II trials will likely lead to larger and more expensive phase III studies. However, unlike other medical disciplines, phase III trials often end in failure for cancer patientsCitation43, e.g., GVAX immunotherapy for prostate cancer. Previous phase II studies have shown that GVAX71 immunotherapy was safe. Immunological studies have developed dosing regimens of tumor antigen-specific antibodies.Citation44 Phase II studies have not yet been compared with chemotherapy treatments. Two phase III trials have used chemotherapy comparator groups.Citation42

In the first trial, VITAL-1, a GVAX immunotherapy, was directly compared with chemotherapy in men with asymptomatic, castration-resistant prostate cancer (despite few radiographically detectable responses in phase II GVAX studies). However, VITAL-1 was unsuccessful. Most patients in the trial were considered unlikely to reach the primary survival endpoint and subsequently, the trial was stopped in 2008. In the second study (VITAL-2), a combination of immunotherapy and chemotherapy was hypothesized to increase survival rates in men with more advanced symptoms. No phase II trials were previously carried out to confirm the effects of a combined chemotherapy and immunotherapy treatment in animal or humans. Furthermore, no prior studies determined appropriate dosing schedules or dosing amounts. The VITAL-2 trial resulted in a greater number of deaths in patients treated with the combination of GVAX and chemotherapy, and the trial was subsequently terminated.Citation42,45 Although the positive predictive values of phase II trials for phase III outcomes are not especially vigorous, the negative predictive values are strong. A negative phase II trial in oncology clearly predicts a negative phase III result.Citation46

Combination immunotherapies could be used to maximize cancer patient benefits. Traditional cancer treatments include a combination of chemotherapy drugs or a combination of radiation therapy and chemotherapy.Citation42 GVAX and CRS-207 are cancer vaccines that have been assessed in pancreatic ductal adenocarcinomas (PDAs). GVAX is composed of 2 irradiated, granulocyte-macrophage colony stimulating factor (GM-CSF)–secreting allogeneic PDA cell lines administered 24 hours after treatment with low-dose cyclophosphamide (Cy) to inhibit regulatory T cells. In a prior study, patients with previously treated advanced PDA who received Cy/GVAX had better induction of mesothelin-specific CD8+ T cells than those treated with GVAX alone. Respectively, the median survival rates were 4.3 and 2.3 months.Citation34 In a CRS-207 phase I study in 2012, patients with PDA who received GVAX before entering the study (n = 3) survived for a median of 17 months compared with the 5 months survival rate for those who did not receive prior GVAX (n = 4).Citation45 These observations led to a phase II randomized multicenter study where Cy/GVAX followed by CRS-207 treatments were shown to significantly improve the overall survival rates by 56% (2.2 months) of patients with metastatic PDAS when compared with Cy/GVAX treatments alone. The stable disease rate was 31%, the 1-year survival rate was 24%, and a stabilization or reduction in CA19-9 levels linked to survival was observed. This study encouraged the following combination trial.Citation46

The most common GVAX-related adverse events were local vaccine injection site reactions (90%) followed by fatigue (16%), nausea (12%), and pain and arthralgia (each at 5%). Two grade 4 (pericardial effusion) and 6 grade 3 (dyspnea, fatigue, injection site reaction, hypokalemia, malignant ascites, and pulmonary embolism) events were reported. There was no association between GVAX dose and the total number of adverse events or grade 3 and 4 adverse events.Citation79

A plasmid encoding both GM-CSF and bi-shRNA furin DNA was transfected into harvested tumor cells via electroporation as part of a vaccination termed FANG (Gradalis, Dallas, TX), which provides the afferent arm of the immune system with a full tumor antigen matrix.Citation47,48 This vaccine is a combination immune therapy that produces intra and extra-cellular adjuvant GM-CSF and simultaneously expresses an innovative RNA interference (RNAi) moiety and a bifunctional short hairpin RNA-furin (bi-shRNA-furin).Citation48

In 2013, a long term follow-up phase I study of FANG in advanced cancer casesCitation49 showed encouraging results. The resulting survival rates demonstrated an advantage of FANG over non-FANG treated populations (576 to 205 d and 604 to 228 d). Additionally, no long-term adversely toxic events were reported or observed with FANG.Citation49 The study also showed a remarkable knockdown (>90%) of both TGF-β1 and TGF-β2.Citation48 TGF-β affects a variety of cell types and has been shown to stimulate or inhibit cell growth, induce apoptosis and increase angiogenesis.Citation50,51 The overexpression of TGF-β has been associated with tumor progression and poor prognoses.Citation52,53 Consequently, the inhibitory effects of TGF-β isoforms suggested an immune-regulating function of GM-CSF. The observed FANG-induced immune response was a basic combinatorial TGF-β-suppressing/GM-CSF-expressing immune modulating therapy, which has since advanced to phase II evaluations for treating different tumor types and stages.Citation49

Novel codon-optimized GM-CSF gene

Native GM-CSF protein expression is poor in tissue-specific and activation-dependent forms.Citation54-56 Highly expressed genes are typically biased toward particular codons. These codons are species-dependent.Citation57,58 A number of studies have demonstrated that a good association between biased codon genes and their levels of expression.Citation59-61 The wild-type human and murine GM-CSF cDNA sequences were created by total gene synthesis and optimized by first identifying codons within the cDNA which were not associated with the codon usage in highly expressed genes in humans and mice, respectively. Each suboptimal codon was replaced with those identified from highly expressed genes of the same species. The novel GM-CSF sequences would improve the large-scale protein production without altering the amino acid sequences and biological function. This molecular strategy to enhance immune responses will prove effective for both DNA vaccines and GVAX vaccine. Citation62, 68

The mechanism underlying the improvement of gene expression and induction of immune responses with codon-optimized, GM-CSF-adjuvanted vaccination is still largely unclear.

In a study of DNA vaccines against HIV-1 Gag using codon-optimized GM-CSF, we showed strong antibody, CTL, and protective immune responses against infection with a recombinant vaccine virus expressing HIV-1 Gag.Citation62 To generate pcGM-CSF (plasmid codon-optimized GM-CSF), the codon-optimized form of murine GM-CSF, the researchers used codons of highly expressed human genes.Citation63,64 In initial tests on the protein expression levels of wild-type and codon-optimized murine GM-CSF vectors transfected into mice (NIH3T3), monkey (COS-7) and human (HeLa) cells, the GM-CSF protein was detected in transfected cells with the pcGM-CSF constructs. The pcGM-CSF-transfected cells produced greater amounts of secreted GM-CSF proteins (11,605 pg/ml in NIH3T3, 12,048 pg/ml in COS-7 and 13,250 pg/ml in HeLa cells) than plasmid wild type GM-CSF (pwtGM-CSF)-transfected cells (158 pg/ml, 205 pg/ml and 210 pg/ml, respectively). These results indicated that the novel codon-optimized murine GM-CSF coding sequences increased GM-CSF expression and significantly elevated the release of GM-CSF in NIH3T3, COS-7 and HeLa cells. Using the ELISPOT assay, we measured the number of antigen-specific T cells per million splenocytes and found that pcGM-CSF at least twofold increased the number of Ag-specific responders as measured by IFN-γ production. (1200 SFU versus 500 SFU). Additionally, We observed a high level of Gag-specific CD8+ memory cells reactivated in the spleens of mice immunized with pcGM + Gag (1213 CD8+/IFN-γ) within 4 d of vP1287 challenge compared with pwt-GM+Gag (564 CD8+/IFN-γ).Citation62 The locally sustained release of this cytokine was successfully shown in murine models.Citation62,65

A similar study on codon-optimizing the human IL-15 gene was also recently investigated.Citation66,67 Additional research is required to investigate the effects of homologous and heterologous codon optimizations in mammalian hosts.Citation62

A study explored codon-optimized murine GM-CSF as a prophylactic vaccine adjuvant in cancer immunotherapy for an HPV-16 E6/E7-transformed cell line, TC-1.Citation68 Peak DC recruitment was observed at 72 h post-inoculation in draining lymph nodes (dLNs). This level was significantly increased (p < 0.05) in mice vaccinated with codon-optimized murine GM-CSF in TC-1 cells (TC-1/cGM) when compared with control mice or mice inoculated with wild-type GM-CSF in TC-1 cells (TC-1/wt). Mice vaccinated with irradiated TC-1/cGM cells exhibited increased levels of functional GM-CSF, enhanced immunosurveillance against TC-1 tumors, increased numbers of Ag-specific IFN-γ-producing CD8+T cells, and enhanced recruitment of macrophage-like cells into dLNs and IFN-γ-producing CD8+T cell compared with mice vaccinated with TC-1/wtGM. Using the ELISPOT assay, we measured the number of antigen-specific T cells per million splenocytes and found that TC-1/cGM at least 5-fold increased the number of Ag-specific responders as measured by IFN-γ production. (2800 SFU vs. 480 SFU). Additionally, we observed a high level of E7-specific IFN-γ producing CD8+ cells progressed through cell cycle upon restimulation (20.3% of TC-1/wt versus 30.5% of TC-1/cGM). These results demonstrated that cell-based vaccines secreting the novel c-GM-CSF gene product could prevent the growth of tumors.Citation68

The sustained local releases of GM-CSF at vaccination sites by GM-CSF-secreting cells in the induction of tumor immunity in many animal models were achieved using cells designed to release 90–300 ng/10Citation6 cells per 24 h.Citation31 Between 240 and 3100 ng/10Citation6 irradiated cells was released over 24 h in transgenic TC-1/cGM cancer cells.Citation68 Significantly increased levels in macrophage and DC populations and tumor-specific effector T cell immunity were noted in the immune response. These results were associated with enhanced antitumor therapeutic benefits. Codon-optimized GM-CSF genes offer utilitarian molecular strategies for potential immune responses to tumor cell-based vaccinations and other immunotherapeutic strategies. Here, we review the literature dealing with the adjuvant activity of GM-CSF both in animal models and clinical trials ().

Table 1. A: GMCSF.

Major clinical studies

B codon-optimization GM-CSF. Major preclinical studies

Conclusions

GM-CSF plays a critical role in eliciting tumor-specific immune responses, including activating both antigen-specific CD4+ and CD8+ T cells.Citation69 Our study is the first to demonstrate that changes in GM-CSF gene optimization may be a breakthrough of the anti-HIV and anti-tumor vaccine. Further clinical study involving larger numbers of patients is needed to assess the utility of this novel gene. We anticipate that the development of GM-CSF treatments will lead to a number of effective cancer therapies. The codon-optimized GM-CSF gene offers a practical molecular strategy for potentiating immune responses to tumor cell-based vaccinations as well as other immunotherapeutic strategies.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgments

We are grateful to Long-Ji Chang and Becky Chen for their helpful discussions.

Funding

This study was supported by CMRPG390611 and CMRPG3E1281, which were provided by Chang Gung University Hospital (Taoyuan, Taiwan).

References

  • Kaushansky KOHP, Hart CE, Forstrom JW, Hagen FS. Role of carbohydrate in the function of human granulocyte-macrophage colony-stimulating factor. Biochemistry 1987; 26:4861-7; PMID:3499177
  • Cebon J, Nicola N, Ward M, Gardner I, Dempsey P, Layton J, Dührsen U, Burgess AW, Nice E, Morstyn G. Granulocyte-macrophage colony stimulating factor from human lymphocytes. The effect of glycosylation on receptor binding and biological activity. J Biol Chem 1990; 265:4483-91; PMID:2155231
  • Hamilton JA. Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol 2008; 8:533-44; PMID:18551128; http://dx.doi.org/10.1038/nri2356
  • Mathias B, Szpila BE, Moore FA, Efron PA, Moldawer LL. A review of GM-CSF therapy in sepsis. Medicine 2015; 94:e2044; PMID:26683913; http://dx.doi.org/10.1097/MD.0000000000002044
  • Chang DZ, Lomazow W, Joy Somberg C, Stan R, Perales M-A. Granulocyte-macrophage colony stimulating factor: An adjuvant for cancer vaccines. Hematology 2004; 9:207-15; PMID:15204102; http://dx.doi.org/10.1080/10245330410001701549
  • Fleetwood AJ, Cook AD, Hamilton JA. Functions of granulocyte-macrophage colony-stimulating factor. 2005; 25:405-28; PMID:16167889
  • Gentile LF, Cuenca AG, Efron PA, Ang D, McKinley BA, Moldawer LL, Moore FA. Persistent inflammation and immunosuppression: A common syndrome and new horizon for surgical intensive care. J Trauma Acute Care surg 2012; 72:1491-501; PMID:22695412; http://dx.doi.org/10.1097/TA.0b013e318256e000
  • Landelle C, Lepape A, Voirin N, Tognet E, Venet F, Bohé J, Vanhems P, Monneret G. Low monocyte human leukocyte antigen-DR is independently associated with nosocomial infections after septic shock. Intensive Care Med 2010; 36:1859-66; PMID:20652682; http://dx.doi.org/10.1007/s00134-010-1962-x
  • Reith W, LeibundGut-Landmann S, Waldburger J-M. Regulation of MHC class II gene expression by the class II transactivator. Nat Rev Immunol 2005; 5:793-806; PMID:16200082; http://dx.doi.org/10.1038/nri1708
  • Hotchkiss RS, Moldawer LL. Parallels between cancer and infectious disease. N Engl J Med 2014; 371:380-3; PMID:25054723; http://dx.doi.org/10.1056/NEJMcibr1404664
  • Meisel C, Schefold JC, Pschowski R, Baumann T, Hetzger K, Gregor J, Weber-Carstens S, Hasper D, Keh D, Zuckermann H, et al. Granulocyte–macrophage colony-stimulating factor to reverse sepsis-associated immunosuppression. Am J Respir Critl Care Med 2009; 180:640-8; PMID:19590022; http://dx.doi.org/10.1164/rccm.200903-0363OC
  • Volk HD RP, Docke WD. Clinical aspects: from systemic inflammation to ‘immunoparalysis’. Chem Immunol 2000; 74:162-77; PMID:10608087; http://dx.doi.org/10.1159/000058753
  • Orozco H, Arch J, Medina-Franco H, Pantoja JP, González QH, Vilatoba M, Hinojosa C, Vargas-Vorackova F, Sifuentes-Osornio J. Molgramostim (gm-csf) associated with antibiotic treatment in nontraumatic abdominal sepsis: A randomized, double-blind, placebo-controlled clinical trial. Arch Surg 2006; 141:150-3; PMID:16490891; http://dx.doi.org/10.1001/archsurg.141.2.150
  • Cornish AL, Campbell IK, McKenzie BS, Chatfield S, Wicks IP. G-CSF and GM-CSF as therapeutic targets in rheumatoid arthritis. Nat Rev Rheumatol 2009; 5:554-9; PMID:19798030; http://dx.doi.org/10.1038/nrrheum.2009.178
  • Wicks IP, Roberts AW. Targeting GM-CSF in inflammatory diseases. Nat Rev Rheumatol 2016; 12:37-48; PMID:26633290; http://dx.doi.org/10.1038/nrrheum.2015.161
  • Bischof RJ, Zafiropoulos D, Hamilton JA, Campbell IK. Exacerbation of acute inflammatory arthritis by the colony-stimulating factors CSF-1 and granulocyte macrophage (GM)-CSF: evidence of macrophage infiltration and local proliferation. Clin Exp Immunol 2000; 119:361-7; PMID:10632676; http://dx.doi.org/10.1046/j.1365-2249.2000.01125.x
  • Williamson DJ, Begley CG, Vadas MA, Metcalf D. The detection and initial characterization of colony-stimulating factors in synovial fluid. Clin Exp Immunol 1988; 72:67-73; PMID:3260840
  • Xu WD, Firestein GS, Taetle R, Kaushansky K, Zvaifler NJ. Cytokines in chronic inflammatory arthritis. II. Granulocyte-macrophage colony-stimulating factor in rheumatoid synovial effusions. J Clin Invest 1989; 83:876-82; PMID:2646320; http://dx.doi.org/10.1172/JCI113971
  • Campbell IK, Rich MJ, Bischof RJ, Dunn AR, Grail D, Hamilton JA. Protection from collagen-induced arthritis in granulocyte-macrophage colony-stimulating factor-deficient mice. J Immunol 1998; 161:3639-44; PMID:9759887
  • Mehta HM, Malandra M, Corey SJ. G-CSF and GM-CSF in Neutropenia. J Immunol 2015; 195:1341-9; PMID:26254266; http://dx.doi.org/10.4049/jimmunol.1500861
  • Benson KF, Li F-Q, Person RE, Albani D, Duan Z, Wechsler J, Meade-White K, Williams K, Acland GM, Niemeyer G, et al. Mutations associated with neutropenia in dogs and humans disrupt intracellular transport of neutrophil elastase. Nat Genet 2003; 35:90-6; PMID:12897784; http://dx.doi.org/10.1038/ng1224
  • Hammond W, Boone T, Donahue R, Souza L, Dale D. A comparison of treatment of canine cyclic hematopoiesis with recombinant human granulocyte-macrophage colony-stimulating factor (GM- CSF), G-CSF interleukin-3, and canine G-CSF. Blood 1990; 76:523-32; PMID:1696146
  • Scheinberg P, Young NS. How I treat acquired aplastic anemia. Blood 2012; 120:1185-96; PMID:22517900; http://dx.doi.org/10.1182/blood-2011-12-274019
  • Tichelli A, Schrezenmeier H, Socié G, Marsh J, Bacigalupo A, Dührsen U, Franzke A, Hallek M, Thiel E, Wilhelm M, et al. A randomized controlled study in patients with newly diagnosed severe aplastic anemia receiving antithymocyte globulin (ATG), cyclosporine, with or without G-CSF: a study of the SAA Working Party of the European Group for Blood and Marrow Transplantation. Blood 2011; 117:4434-41; PMID:21233311; http://dx.doi.org/10.1182/blood-2010-08-304071
  • Jeng MR, Naidu PE, Rieman MD, Rodriguez-Galindo C, Nottage KA, Thornton DT, Li CS, Wiang WC. Granulocyte-macrophage colony stimulating factor and immunosuppression in the treatment of pediatric acquired severe aplastic anemia. Pediatr Blood Cancer 2005; 45:170-5; PMID:15593082; http://dx.doi.org/10.1002/pbc.20278
  • Forni GFH, Martino F, Hamaoka T, Jemma C, Caretto P, Giovarelli M. Helper strategy in tumor immunology: expansion of helper lymphocytes and utilization of helper lymphokines for experimental and clinical immunotherapy. Cancer Metastasis Rev 1988; 7:289-309; PMID:2974763; http://dx.doi.org/10.1007/BF00051371
  • Forni G, Giovarelli M, Santoni A. Lymphokine-activated tumor inhibition in vivo. I. The local administration of interleukin 2 triggers nonreactive lymphocytes from tumor-bearing mice to inhibit tumor growth. J Immunol 1985; 134:1305-11; PMID:3871210
  • Dranoff G. GM-CSF-based cancer vaccines. Immunol Rev 2002; 188:147-54; PMID:12445288; http://dx.doi.org/10.1034/j.1600-065X.2002.18813.x
  • Dranoff G. GM-CSF-secreting melanoma vaccines. Oncogene 2003; 22:3188-92; PMID:12789295; http://dx.doi.org/10.1038/sj.onc.1206459
  • Candido J, Hagemann T. Cancer-related inflammation. J Clin Immunol 2012; 33:79-84; PMID:23225204; http://dx.doi.org/10.1007/s10875-012-9847-0
  • Dranoff G, Jaffee E, Lazenby A, Golumbek P, Levitsky H, Brose K, Jackson V, Hamada H, Pardoll D, Mulligan RC. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci U S A 1993; 90:3539-43; PMID:8097319; http://dx.doi.org/10.1073/pnas.90.8.3539
  • Gridelli C, Rossi A, Maione P, Ferrara ML, Castaldo V, Sacco PC. Vaccines for the treatment of non-small cell lung cancer: A renewed anticancer strategy. Oncologist 2009; 14:909-20; PMID:19726457; http://dx.doi.org/10.1634/theoncologist.2009-0017
  • Yin W, He Q, Hu Z, Chen Z, Qifeng M, Zhichun S, Zhihui Q, Xiaoxia N, Li J, Gao J. A novel therapeutic vaccine of GM-CSF/TNFα surface-modified RM-1 cells against the orthotopic prostatic cancer. Vaccine 2010; 28:4937-44; PMID:20653081; http://dx.doi.org/10.1016/j.vaccine.2010.05.038
  • Laheru D, Lutz E, Burke J, Biedrzycki B, Solt S, Onners B, Tartakovsky I, Nemunaitis J, Le D, Sugar E, et al. Allogeneic GM-CSF secreting tumor immunotherapy (GVAX(®)) alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: A pilot study of safety, feasibility and immune activation. Clin Cancer Res 2008; 14:1455-63; PMID:18316569; http://dx.doi.org/10.1158/1078-0432.CCR-07-0371
  • Zilberberg J, Friedman TM, Dranoff G, Korngold R. Treatment with GM-CSF secreting myeloid leukemia cell vaccine prior to autologous-BMT improves the survival of leukemia challenged mice. Biol Blood Marrow Transplant 2011; 17:330-40; PMID:20946965; http://dx.doi.org/10.1016/j.bbmt.2010.09.020
  • Zhao F, Dou J, He XF, Wang J, Chu L, Hu W, Yu F, Hu K, Wu Y, Gu N. Enhancing therapy of B16F10 melanoma efficacy through tumor vaccine expressing GPI-anchored IL-21 and secreting GM-CSF in mouse model. Vaccine 2010; 28:2846-52; PMID:20153795; http://dx.doi.org/10.1016/j.vaccine.2010.01.057
  • Clavreul A, Piard N, Tanguy J-Y, Gamelin E, Rousselet M-C, Leynia P, Menei P. Autologous tumor cell vaccination plus infusion of GM-CSF by a programmable pump in the treatment of recurrent malignant gliomas. J Clin Neurosci 2010; 17:842-8; PMID:20466548; http://dx.doi.org/10.1016/j.jocn.2009.11.017
  • Zijlmans HJMAA, Fleuren GJ, Baelde HJ, Eilers PHC, Kenter GG, Gorter A. Role of tumor-derived proinflammatory cytokines GM-CSF, TNF-α, and IL-12 in the migration and differentiation of antigen-presenting cells in cervical carcinoma. Cancer 2007; 109:556-65; PMID:17177206; http://dx.doi.org/10.1002/cncr.22428
  • Driessens G, Hoffmann P, Pouwels M, Zlotta A, Schulman C, Velu T, Bruyns CA. Synergy between dendritic cells and GM–CSF-secreting tumor cells for the treatment of a murine renal cell carcinoma. J Immunother 2009; 32:140-4; PMID:19238012; http://dx.doi.org/10.1097/CJI.0b013e3181920275
  • Simons JW, Nelson W, Nemunaitis J. Phase II trials of a GM-CSF gene-transduced prostate cancer cell line vaccine (GVAX) in hormone refractory prostate cancer. Proc Am Soc Clin Oncol 2002; 21:729.
  • Tani K, Azuma M, Nakazaki Y, Oyaizu N, Hase H, Ohata J, Takahashi K, OiwaMonna M, Hanazawa K, Wakumoto Y, et al. Phase I study of autologous tumor vaccines transduced with the GM-CSF gene in four patients with stage IV renal cell cancer in Japan: Clinical and immunological findings. Mol Ther 2004; 10:799-816; PMID:15451464; http://dx.doi.org/10.1016/j.ymthe.2004.07.001
  • Drake CG. Prostate cancer as a model for tumour immunotherapy. Nature reviews Immunology 2010; 10:580-93; PMID:20651745; http://dx.doi.org/10.1038/nri2817
  • Hales RK, Banchereau J, Ribas A, Tarhini AA, Weber JS, Fox BA, Drake CG. Assessing oncologic benefit in clinical trials of immunotherapy agents. Ann Oncol 2010; 21:1944-51; PMID:20237004; http://dx.doi.org/10.1093/annonc/mdq048
  • Simons JW, Jaffee EM, Weber CE, Levitsky HI, Nelson WG, Carducci MA, Lazenby AJ, Cohen LK, Finn CC, Clift SM, et al. Bioactivity of autologous irradiated renal cell carcinoma vaccines generated by ex vivo granulocyte-macrophage colony-stimulating factor gene transfer. Cancer Res 1997; 57:1537-46; PMID:9108457
  • Le DT, Brockstedt DG, Nir-Paz R, Hampl J, Mathur S, Nemunaitis J, Sterman DH, Hassan R, Lutz E, Moyer B, et al. A live-attenuated listeria vaccine (ANZ-100) and a live-attenuated listeria vaccine expressing mesothelin (CRS-207) for advanced cancers: Phase 1 studies of safety and immune induction. Clin Cancer Res 2012; 18:858-68; PMID:22147941; http://dx.doi.org/10.1158/1078-0432.CCR-11-2121
  • Le DT, Wang-Gillam A, Picozzi V, Greten TF, Crocenzi T, Springett G, Morse M, Zeh H, Cohen D, Fine RL, et al. Safety and survival with GVAX pancreas prime and listeria monocytogenes–expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J Clin Oncol 2015; 33:1325-33; PMID:25584002; http://dx.doi.org/10.1200/JCO.2014.57.4244
  • Senzer N, Barve M, Kuhn J, Melnyk A, Beitsch P, Lazar M, Lifshitz S, Magee M, Oh J, Mill SW, et al. Phase I trial of “bi-shRNAi(furin)/GMCSF DNA/autologous tumor cell” vaccine (FANG) in advanced cancer. Mol Ther 2012; 20:679-86; PMID:22186789; http://dx.doi.org/10.1038/mt.2011.269
  • Phillip B, Maples P, Kumar P, Yu Y, Wang Z, PhD, Jay C, PhD, Pappen BO, Rao DD, PhD, Kuhn J, MD, Nemunaitis J, MD, et al. FANG Vaccine: Autologous tumor cell vaccine genetically modified to express GM-CSF and block production of furin. BioProc J (Winter 2009/2010); 8:4-14
  • Senzer N, Barve M, Nemunaitis J, Kuhn J, Melnyk A, Beitsch P, Magee M, Oh J, Bedell C, Kumar P, Rao DD, et al. Long term follow up: Phase I trial of“bi-shRNA furin/GMCSF DNA/autologous tumor cell” Immunotherapy (FANG™) in advanced cancer. J Vaccines Vaccination 2013; 4:209; http://dx.doi.org/10.4172/2157-7560.1000209
  • Ashley DM, Kong FM, Bigner DD, Hale LP. Endogenous expression of transforming growth factor β1 inhibits growth and tumorigenicity and enhances fas-mediated apoptosis in a murine high-grade glioma model. Cancer Res 1998; 58:302-9; PMID:9443409
  • Ashley DM, Sampson JH, Archer GE, Hale LP, Bigner DD. Local production of TGF β1 inhibits cerebral edema, enhances TNF-α induced apoptosis and improves survival in a murine glioma model. J Neuroimmunol 1998; 86:46-52; PMID:9655471; http://dx.doi.org/10.1016/S0165-5728(98)00017-4
  • Levy L, Hill CS. Alterations in components of the TGF-β superfamily signaling pathways in human cancer. Cytokine Growth Factor Rev 2006; 17:41-58; PMID:16310402; http://dx.doi.org/10.1016/j.cytogfr.2005.09.009
  • Bierie B, Moses HL. Tumour microenvironment: TGF[beta]: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer 2006; 6:506-20; PMID:16794634; http://dx.doi.org/10.1038/nrc1926
  • Thorens B, Mermod J-J, Vassalli P. Phagocytosis and inflammatory stimuli induce GM-CSF mRNA in macrophages through posttranscriptional regulation. Cell 1987; 48:671-9; PMID:3102073; http://dx.doi.org/10.1016/0092-8674(87)90245-5
  • Shannon MF, Gamble JR, Vadas MA. Nuclear proteins interacting with the promoter region of the human granulocyte/macrophage colony-stimulating factor gene. Proc Natl Acad Sci U S A 1988; 85:674-8; PMID:3257571; http://dx.doi.org/10.1073/pnas.85.3.674
  • Bickel M, Cohen RB, Pluznik DH. Post-transcriptional regulation of granulocyte-macrophage colony-stimulating factor synthesis in murine T cells. J Immunol 1990; 145:840-5; PMID:2197329
  • Gouy M, Gautier C. Codon usage in bacteria: correlation with gene expressivity. Nucleic Acids Res 1982; 10:7055-74; PMID:6760125; http://dx.doi.org/10.1093/nar/10.22.7055
  • Sharp PM. What can AIDS virus codon usage tell us? Nature 1986; 324:114-; PMID:3641061; http://dx.doi.org/10.1038/324114a0
  • Bennetzen JL, Hall BD. Codon selection in yeast. J Biol Chem 1982; 257:3026-31; PMID:7037777
  • Zolotukhin S, Potter M, Hauswirth WW, Guy J, Muzyczka N. A “humanized” green fluorescent protein cDNA adapted for high-level expression in mammalian cells. J Virol 1996; 70:4646-54; PMID:8676491
  • zur Megede J, Chen M-C, Doe B, Schaefer M, Greer CE, Selby M, Otten GR, Barnett SW. Increased expression and immunogenicity of sequence-modified human immunodeficiency virus type 1 gag gene. J Virol 2000; 74:2628-35; PMID:10684277; http://dx.doi.org/10.1128/JVI.74.6.2628-2635.2000
  • Qiu J-T, Chang T-C, Lin C-T, Chen Y-M, Li FQ, Soong Y-K, Lai CH. Novel codon-optimized GM-CSF gene as an adjuvant to enhance the immunity of a DNA vaccine against HIV-1 Gag. Vaccine 2007; 25:253-63; PMID:16971027; http://dx.doi.org/10.1016/j.vaccine.2006.07.034
  • Kotsopoulou E, Kim VN, Kingsman AJ, Kingsman SM, Mitrophanous KA. A Rev-independent human immunodeficiency virus type 1 (HIV-1)-based vector that exploits a codon-optimized HIV-1 gag-pol Gene. J Virol 2000; 74:4839-52; PMID:10775623; http://dx.doi.org/10.1128/JVI.74.10.4839-4852.2000
  • Haas J, Park E-C, Seed B. Codon usage limitation in the expression of HIV-1 envelope glycoprotein. Curr Biol 1996; 6:315-24; PMID:8805248; http://dx.doi.org/10.1016/S0960-9822(02)00482-7
  • Warren TL, Weiner GJ. Uses of granulocyte-macrophage colony-stimulating factor in vaccine development. Curr Opin Hematol 2000; 7:168-73; PMID:10786654; http://dx.doi.org/10.1097/00062752-200005000-00007
  • Kutzler MA, Robinson TM, Chattergoon MA, Choo DK, Choo AY, Choe PY, Ramanathan MP, Parkinson R, Kudchodkar S, Tamura Y, et al. Coimmunization with an optimized IL-15 plasmid results in enhanced function and longevity of CD8 T cells that are partially independent of CD4 T cell help. J Immunol 2005; 175:112-23; PMID:15972637; http://dx.doi.org/10.4049/jimmunol.175.1.112
  • Hsu C, Hughes MS, Zheng Z, Bray RB, Rosenberg SA, Morgan RA. Primary human T lymphocytes engineered with a codon-optimized IL-15 gene resist cytokine withdrawal-induced apoptosis and persist long-term in the absence of exogenous cytokine. J Immunol (Baltimore, Md : 1950) 2005; 175:7226-34; PMID:16301627; http://dx.doi.org/10.4049/jimmunol.175.11.7226
  • Lin C-C, Tsai C-C, Lee J-M, Fang C-H, Chang K-S, Wong K-K, Lin CT, Qiu JT. The efficacy of a novel vaccine approach using tumor cells that ectopically express a codon-optimized murine GM-CSF in a murine tumor model. Vaccine 2016; 34:134-41; PMID:26546261; http://dx.doi.org/10.1016/j.vaccine.2015.10.106
  • Lang RA, Metcalf D, Cuthbertson RA, Lyons I, Stanley E, Kelso A, Kannourakis G, Williamson DJ, Klintworth GK, Gonda TJ, et al. Transgenic mice expressing a hemopoietic growth factor gene (GM-CSF) develop accumulations of macrophages, blindness, and a fatal syndrome of tissue damage. Cell 1987; 51:675-86; PMID:3499986; http://dx.doi.org/10.1016/0092-8674(87)90136-X
  • Dranoff G, Jaffee E, Lazenby A, Golumbek P, Levitsky H, Brose K. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci USA 1993; 90:3539-43; PMID:8097319; http://dx.doi.org/10.1073/pnas.90.8.3539
  • Parker SE, Monteith D, Horton H, Hof R, Hernandez P, Vilalta A, Hartikka J, Hobart P, Bentley CE, Chang A, et al. Safety of a GM-CSF adjuvant-plasmid DNA malaria vaccine. Gene Ther 2001; 8:1011-23; PMID:11438836; http://dx.doi.org/10.1038/sj.gt.3301491
  • Barouch DH, Santra S, Tenner-Racz K, Racz P, Kuroda MJ, Schmitz JE, Jackson SS, Lifton MA, Freed DC, Perry HC, et al. Potent CD4+ T Cell Responses Elicited by a Bicistronic HIV-1 DNA Vaccine Expressing gp120 and GM-CSF. J Immunol 2002; 168:562-8; PMID:11777947; http://dx.doi.org/10.4049/jimmunol.168.2.562
  • Simons JW, Jaffee EM, Weber CE, Levitsky HI, Nelson WG, Carducci MA, Lazenby AJ, Cohen LK, Finn CC, Clift SM, et al. Bioactivity of autologous irradiated renal cell carcinoma vaccines generated by ex vivo granulocyte-macrophage colony-stimulating factor gene transfer. Cancer Res 1997; 57:1537-46; PMID:9108457
  • Mastrangelo MJ, Maguire HC, Jr., Eisenlohr LC, Laughlin CE, Monken CE, McCue PA, Kovatich AJ, Lattime EC. Intratumoral recombinant GM-CSF-encoding virus as gene therapy in patients with cutaneous melanoma. Cancer Gene Ther 1999; 6:409-22; PMID:10505851; http://dx.doi.org/10.1038/sj.cgt.7700066
  • Simons JW, Mikhak B, Chang J-F, DeMarzo AM, Carducci MA, Lim M, Weber CE, Baccala AA, Goemann MA, Clift SM, et al. Induction of immunity to prostate cancer antigens: Results of a clinical trial of vaccination with irradiated autologous prostate tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor using ex vivo gene transfer. Cancer Res 1999; 59:5160-8; PMID:10537292
  • Chang AE, Li Q, Bishop DK, Normolle DP, Redman BD, Nickoloff BJ. Immunogenetic therapy of human melanoma utilizing autologous tumor cells transduced to secrete granulocyte-macrophage colony-stimulating factor. Human Gene Ther 2000; 11:839-50; PMID:10779161; http://dx.doi.org/10.1089/10430340050015455
  • Salgia R, Lynch T, Skarin A, Lucca J, Lynch C, Jung K, Hodi FS, Jaklitsch M, Mentzer S, Swanson S, et al. Vaccination with irradiated autologous tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor augments antitumor immunity in some patients with metastatic non–small-cell lung carcinoma. J Clin Oncol 2003; 21:624-30; PMID:12586798; http://dx.doi.org/10.1200/JCO.2003.03.091
  • Soiffer R, Hodi FS, Haluska F, Jung K, Gillessen S, Singer S, Tanabe K, Duda R, Mentzer S, Jaklitsch M, et al. Vaccination with irradiated, autologous melanoma cells engineered to secrete granulocyte-macrophage colony-stimulating factor by adenoviral-mediated gene transfer augments antitumor immunity in patients with metastatic melanoma. J Clin Oncol 2003; 21:3343-50; PMID:12947071; http://dx.doi.org/10.1200/JCO.2003.07.005
  • Nemunaitis J, Sterman D, Jablons D, Smith JW, Fox B, Maples P, Hamilton S, Borellini F, Lin A, Morali S, et al. Granulocyte–macrophage colony-stimulating factor gene-modified autologous tumor vaccines in non–small-cell lung cancer. J Natl Cancer Inst 2004; 96:326-31; PMID:14970281; http://dx.doi.org/10.1093/jnci/djh028
  • Luiten RM, Kueter EWM, Mooi W, Gallee MPW, Rankin EM, Gerritsen WR, Clift SM, Nooijen WJ, Weder P, van de Kasteele WF, et al. Immunogenicity, including vitiligo, and feasibility of vaccination with autologous GM-CSF–transduced tumor cells in metastatic melanoma patients. J Clin Oncol 2005; 23:8978-91; PMID:16260696; http://dx.doi.org/10.1200/JCO.2005.01.6816
  • Simons JW, Carducci MA, Mikhak B, Lim M, Biedrzycki B, Borellini F, Clift SM, Hege KM, Ando DG, Piantadosi S, et al. Phase I/II Trial of an Allogeneic Cellular Immunotherapy in Hormone-Naïve Prostate Cancer. Clin Cancer Res 2006; 12(11):3394-3401; http://dx.doi.org/10.1158/1078-0432.CCR-06-0145
  • Laheru D, Lutz E, Burke J, Biedrzycki B, Solt S, Onners B, et al. Allogeneic Granulocyte Macrophage Colony-Stimulating Factor–Secreting Tumor Immunotherapy Alone or in Sequence with Cyclophosphamide for Metastatic Pancreatic Cancer: A Pilot Study of Safety, Feasibility, and Immune Activation. Clin Cancer Res 2008; 14(5):1455-1463; http://dx.doi.org/10.1158/1078-0432.CCR-07-0371
  • Emens LA, Asquith JM, Leatherman JM, Kobrin BJ, Petrik S, Laiko M, Levi J, Daphtary MM, Biedrzycki B, Wolff AC, et al. Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor–secreting breast tumor vaccine: A chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol 2009; 27:5911-8; PMID:19805669; http://dx.doi.org/10.1200/JCO.2009.23.3494
  • van den Eertwegh AJM, Versluis J, van den Berg HP, Santegoets SJAM, van Moorselaar RJA, van der Sluis TM, Gall HE, Harding TC, Jooss K, Lowy I, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012; 13:509-17; PMID:22326922; http://dx.doi.org/10.1016/S1470-2045(12)70007-4
  • Vuky J, Corman JM, Porter C, Olgac S, Auerbach E, Dahl K. Phase II trial of neoadjuvant docetaxel and CG1940/CG8711 followed by radical prostatectomy in patients with high-risk clinically localized prostate cancer. Oncologist 2013; 18:687-8; PMID:23740935; http://dx.doi.org/10.1634/theoncologist.2011-0234
  • Le DT, Wang-Gillam A, Picozzi V, Greten TF, Crocenzi T, Springett G, Morse M, Zeh H, Cohen D, Fine RL, et al. Safety and survival with GVAX pancreas prime and listeria monocytogenes–expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J Clin Oncol 2015; 33:1325-33; PMID:25584002; http://dx.doi.org/10.1200/JCO.2014.57.4244
  • Lipson EJ, Sharfman WH, Chen S, McMiller TL, Pritchard TS, Salas JT, Sartorius-Mergenthaler S, Freed I, Ravi S, Wang H, et al. Safety and immunologic correlates of Melanoma GVAX, a GM-CSF secreting allogeneic melanoma cell vaccine administered in the adjuvant setting. J Transl Med 2015; 13; PMID:26143264; http://dx.doi.org/10.1186/s12967-015-0572-3

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.