Skip to main content
F1000Research logoLink to F1000Research
. 2017 May 18;6:710. [Version 1] doi: 10.12688/f1000research.10795.1

Harnessing the immune response to target tumors

Luisa Manning 1, John Nemunaitis 1,2,a
PMCID: PMC5461900  PMID: 28620466

Abstract

Development of “immune-based targeted therapy” in oncology has limited experience with signal pathway modulation. However, as we have become better versed in understanding immune function related to anticancer response, “hints” of specific targets associated with sensitivity and resistance have been identified with targeted immune therapy. This brief review summarizes the relationship of several targeted immune therapeutics and activity associated clinical responsiveness.

Keywords: targeted immune therapy, anticancer response, checkpoint inhibitors, adoptive T-cell therapy, CAR-T cell therapy

Introduction

Although the immune system distinguishes self versus non-self antigens with the intent to eliminate cells expressing non-self antigens, cancer cells have developed mechanisms to escape or suppress the “non-self attack”, thereby enabling tumor proliferation and progression. Increasing numbers of innovative immunotherapies are being developed that address immune modulation of non-self targets to reverse cancer defenses.

Checkpoint inhibitors

Several targets have been associated with evidence of clinical benefit, resulting in a broad spectrum of investigational and approved immunotherapies. These include cellular therapies (adoptive T-cell and dendritic cell therapy, cytokine-induced killer cells, tumor vaccines, and autologous tumor cell therapy) and checkpoint inhibitors (PD-1/L-1 and CTLA-4 inhibitors). Several checkpoint inhibitors have shown superior clinical benefit over standard of care 13; however, a high percentage of patients do not show durable response rates in monotherapies or combination therapies with checkpoint blockade. Factors like tumor-infiltrating lymphocyte (TIL) infiltration and PD-L1 expression levels are associated with response 4. In addition, tumor mutation burden (TMB) appears to be a prognostic marker for immune response 5. During cancer cell proliferation, somatic mutations increase the expression of a variety of tumor-associated antigens and neoantigens. Studies show that high TMB correlates with the amount of immunogenic neoantigens ( P <0.0001), presented by major histocompatibility complex (MHC) molecules to immune effector cells, inducing higher durable immune responses (overall response rate of 63% versus 0%; P = 0.03) and progression-free survival prolongation (14.5 versus 3.7 months; P = 0.01) than in tumor types with lower mutation burden 6. Regardless of histology type, tumors with a mean mutational load of more than 10 somatic mutations per megabase of coding DNA appear more likely to be immunogenic to effector T cells eliciting antitumor immunity 7, 8. Other cancer types, such as colorectal cancer, and interestingly subgroups with a high number of somatic mutations and potential mutation-associated neoepitopes appear to correlate with higher responses to checkpoint inhibitors in mismatch repair-deficient tumors 9. Recent studies show that the overall response rate to PD-1/L-1 therapies in high TMB tumor types has been durable for years with delayed relapse or disease progression 10. On the other hand, signal pathways, such as those associated with interferon receptor expression related to loss of JAK 1 or JAK 2 function, result in unresponsiveness to interferon gamma, a common antiproliferative cytokine associated with oncolytic activity. This effect has been well demonstrated in a subset of PD-1/L1-refractory patients. Zaretsky et al. 10 identified inactivating mutations in JAK 1 and 2 that silence the CD8 T cell-induced interferon gamma signaling cascade, an adaptive antitumor response. Another mechanism such as beta 2-microglobulin inactivation results in loss of MHC1 expression. Moreover, mutations of death receptors—like Fas or tumor necrosis factor-related apoptosis-inducing ligand—are associated with insensitivities against granzymes or perforin or both, which also play major roles in immune escape and resistance.

Bu et al. 11 discussed PD-1 resistances and highlighted a pattern of upregulated genes first observed in patients with PD-1-resistant melanoma 12, termed innate PD-1 resistance effect. The analysis of somatic mutanomes and transcriptomes of pretreatment melanoma biopsies included the comparison of differentiated gene expression in PD-1 responders versus non-responders. Higher expressed genes in checkpoint non-responding tumors included mesenchymal transition genes ( AXL, ROR2, WNT5A, LOXL2, TWIST2, TAGLN, and FAP), immunosuppressive genes ( IL10, VEGFA, and VEGFC), and monocyte and macrophage chemotactic genes ( CCL2, CCL7, CCL8, and CCL13) 12, while immune responsive tumors also contained transforming growth factor beta (TGFβ) signaling defects.

To address and overcome resistant mechanisms, ongoing studies are extensively investigating combination approaches (that is, with checkpoint inhibitors). For example, experiments of targeted inhibition of mitogen-activated protein kinase show synergy with PD-1/L1 pathway inhibition and increases in CD8 T-cell number within the tumor environment in association with increased tumor response 13.

Adoptive T-cell therapies

Adoptive dendritic cell or T-cell therapies show clinically meaningful value in hematologic malignancies, and a small number of case reports support efficacy in solid tumors with demonstration of durable clinical responses 1416. For example, 20 to 25% of patients with metastatic melanoma showed durable responses to expanded TIL therapies 14, 17. This is most likely related to neoantigen signal identification. A remarkable case of response of adoptive T-cell therapy to a common neoantigen target was recently demonstrated to KRAS G12D mutation 16 and other, lesser-known mutations 14, 15. However, currently, the majority of cancer vaccines and adoptive T-cell approaches fall short of significant efficacy targeting pre-selected MHC-dependent (genetically modified T cells) or independent—chimeric antigen receptor-T (CAR-T)—antigens showing limited activity in solid tumors, possibly related to the lack of knowledge of relevant neoantigens ( Table 1). While CD19-targeting CAR-T cell therapies have demonstrated curative events in B-cell malignancies 18, 19, efficacy in solid tumors appears to be limited by heterogeneity, lack of relevant tumor-specific or -associated antigens and low immunogenicity 20, in balance with other immunosuppressive pathways not addressed within the tumor microenvironment.

Table 1. Cellular immunotherapies.

Target Tumor type(s) Reference/Clinical trial(s)
Chimeric antigen receptor–T cell therapies/targets
CD19 B-cell malignancies 18, 19; NCT02975687;
NCT02842138; NCT02813837
Mesothelin Mesothelioma, lung cancer, breast cancer 36, 37; NCT02930993; NCT02706782
L1-CAM Metastatic neuroblastoma 38; NCT02311621
GD2 Neuroblastoma 25, 39; NCT02107963; NCT02919046
Lewis Y Myeloid malignancies 24, 40; NCT01716364
EGFRvIII Brain tumor NCT01454596
HER2 Colon cancer, HER2-positive lung cancer, malignant
glioma, Her2-positive sarcoma
26, 41; NCT00889954;
NCT00902044; NCT01109095
CD20 Follicular and mantel cell lymphoma 42, 43; NCT00621452
CEA Stomach cancer, metastatic adenocarcinoma, breast
cancer
44; NCT00673829; NCT00673322
MUC-16/IL-12 Ovarian cancer 45; NCT02498912
WT1 Acute myeloid leukemia, NSCLC, breast, pancreatic,
ovarian, colorectal cancer, mesothelioma
27, 28; NCT02408016
CAIX Renal cell carcinoma 46, 47
FAP Malignant pleural mesothelioma 48
PSMA Prostate cancer NCT00664196; NCT01140373
Kappa light chain (klC) B-cell lymphoma, chronic lymphocytic leukemia,
multiple myeloma
NCT00881920
CD30 Hodgkin’s lymphoma, non-Hodgkin’s lymphomas NCT01316146
HLA-A1/MAGE1 Melanoma 49, 50
HLA-A2/NY-ESO-1 Sarcoma, melanoma 51
MUC1 Ovarian, breast, pancreas, colorectal, malignant
glioma, NSCLC, hepatocellular
52; NCT02587689; NCT02617134;
NCT02839954
VEGFR-2 Solid tumors 53, 54; NCT01218867
Adoptive cell therapies
Autologous tumor-infiltrating
lymphocyte therapy and IL-2
Metastatic melanoma 14
Dendritic cell vaccine and cytokine-
induced killer cell therapy
Hepatobiliary, pancreatic cancer 55
Adoptive T-cell transfer Metastatic melanoma 15
Dendritic cell-derived exosomes
(Dex)
NSCLC, melanoma, colorectal cancer 5659
Adoptive CD8 + T cells KRAS, G12D, colorectal 16
Autologous tumor cell therapy
Vigil EATC Ovarian cancer, Ewing’s sarcoma, NSCLC,
melanoma, triple-negative breast cancer, solid tumors
3235

IL, interleukin; NSCLC, non-small cell lung cancer.

Adoptive T-cell therapy “exhaustion” may also be influenced by upregulation of pathways such as PD-L1 expression on tumor cells. Strategies to convert the negative signal of PD-L1 to co-stimulatory receptors by PD1:28 chimer alteration showed encouraging results in activation of CD8 effector T cells 21. Tran et al. identified CD8 T-cell responses against mutant KRAS G12D and HLA-C*08:02 in a patient with colorectal cancer, receiving a single-dose infusion of 1.48 × 10 11 TILs (approximately 75% CD8 T cells) with durable regression of lung metastases with disease progression 9 months after treatment 16.

Tumor signaling/microenvironment modulation

Overcoming tumor-induced immunosuppression can also involve tumor signal modulation and microenvironment influence. Altered expressions of survival genes (Bcl-xL), increasing the expression of dominant negative TGFβ receptors to overcome inhibitory effects 22, regulatory T suppression, indoleamine 2,3-dioxygenase downregulation, and other signaling microenvironment therapeutics, such as WNT/β-catenin signaling pathway 23, are being tested to address benefit opportunity.

CAR-T: selective antigen targets

Targeting driver mutations or their de novo neoepitopes are very attractive and appear to be very promising in effective anticancer therapies. There are several cancer-associated or -specific antigen-loaded CAR-T cell therapies, selected by different algorithms, in clinical trials to investigate further efficacy in solid tumors ( Table 1). In vivo activity of gene-modified T cells was demonstrated in the delayed growth and prolonged survival of Lewis Y antigen CAR-T cell therapy in lymphoma with the report of two cases with stable disease 24. Louis et al. reported other responses, including three complete responses in patients with neuroblastoma treated with specific CAR-T cells targeting GD2 ganglioside 25. HER2-positive colon, lung cancer, and sarcomas are also under investigational therapy with HER2 CAR-T therapy, showing promising results with stable diseases for 12 weeks up to 14 months but no partial or complete responses in HER2-positive sarcomas 26. Other targets—that is, carcinoembryonic antigen (CEA) in colon cancer and WT1 in mesothelioma and ovarian cancer 27, 28—are being studied as well. Among the most challenging aspects of adoptive cell therapies and CAR-T engineering are the identification and use of antigens for focused immune effector cell activation to cancer targets only. Despite the large number of investigated tumor antigens with limited encouraging results, high rates of undesirable off-tumor effects, such as cytokine-release syndrome (CRS) or other immune-related adverse events, are widely seen in CAR-T cell therapies. Thus, new approaches with implications for suicide genes like inducible caspase9 or herpes simplex thymidine kinase are under investigation to enhance the safety of T-cell therapies 29, 30 along with novel regimens to directly address CRS (that is, interleukin-6 inhibitor) 31.

Interestingly, one investigational personalized cellular immunotherapy product with a mechanism directly associated with autologous DNA engineered tumor cells called Vigil 3235 shows evidence of enhanced tumor-specific antigen targeting via effector T-cell activation in correlation with clinical benefit in solid tumors. Autologous tumor cells include the full patient- and tumor-specific antigen repertoire. This is a unique aspect of the Vigil therapy.

Conclusions

The future is bright for combination immunotherapy, particularly as exact targets are identified with the tumor microenvironment, thereby enabling access to tumor “non-self” neoantigens.

Acknowledgments

The authors would like to acknowledge Michelle Richey and Brenda Marr for their capable and knowledgeable assistance in the preparation of the manuscript.

Editorial Note on the Review Process

F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).

The referees who approved this article are:

  • Lei Zheng, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA

  • David Fisher, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA

Funding Statement

The author(s) declared that no grants were involved in supporting this work.

[version 1; referees: 2 approved]

References

  • 1. Rizvi NA, Mazières J, Planchard D, et al. : Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 2015;16(3):257–65. 10.1016/S1470-2045(15)70054-9 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 2. Rosenberg JE, Hoffman-Censits J, Powles T, et al. : Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016;387(10031):1909–20. 10.1016/S0140-6736(16)00561-4 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 3. Hamanishi J, Mandai M, Ikeda T, et al. : Safety and Antitumor Activity of Anti-PD-1 Antibody, Nivolumab, in Patients With Platinum-Resistant Ovarian Cancer. J Clin Oncol. 2015;33(34):4015–22. 10.1200/JCO.2015.62.3397 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 4. Ameratunga M, Asadi K, Lin X, et al. : PD-L1 and Tumor Infiltrating Lymphocytes as Prognostic Markers in Resected NSCLC. PLoS One. 2016;11(4):e0153954. 10.1371/journal.pone.0153954 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 5. Burke JM, Lamm DL, Meng MV, et al. : A first in human phase 1 study of CG0070, a GM-CSF expressing oncolytic adenovirus, for the treatment of nonmuscle invasive bladder cancer. J Urol. 2012;188(6):2391–7. 10.1016/j.juro.2012.07.097 [DOI] [PubMed] [Google Scholar]
  • 6. Rizvi NA, Hellmann MD, Snyder A, et al. : Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–8. 10.1126/science.aaa1348 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 7. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. : Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415–21. 10.1038/nature12477 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 8. Schumacher TN, Schreiber RD: Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74. 10.1126/science.aaa4971 [DOI] [PubMed] [Google Scholar]
  • 9. Le DT, Uram JN, Wang H, et al. : PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;372(26):2509–20. 10.1056/NEJMoa1500596 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 10. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. : Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med. 2016;375(9):819–29. 10.1056/NEJMoa1604958 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 11. Bu X, Mahoney KM, Freeman GJ: Learning from PD-1 Resistance: New Combination Strategies. Trends Mol Med. 2016;22(6):448–51. 10.1016/j.molmed.2016.04.008 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 12. Hugo W, Zaretsky JM, Sun L, et al. : Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell. 2016;165(1):35–44. 10.1016/j.cell.2016.02.065 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 13. Ebert PJ, Cheung J, Yang Y, et al. : MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade. Immunity. 2016;44(3):609–21. 10.1016/j.immuni.2016.01.024 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 14. Rosenberg SA, Yang JC, Sherry RM, et al. : Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011;17(13):4550–7. 10.1158/1078-0432.CCR-11-0116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Verdegaal EM, de Miranda NF, Visser M, et al. : Neoantigen landscape dynamics during human melanoma-T cell interactions. Nature. 2016;536(7614):91–5. 10.1038/nature18945 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 16. Tran E, Robbins PF, Lu YC, et al. : T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. N Engl J Med. 2016;375(23):2255–62. 10.1056/NEJMoa1609279 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 17. Goff SL, Dudley ME, Citrin DE, et al. : Randomized, Prospective Evaluation Comparing Intensity of Lymphodepletion Before Adoptive Transfer of Tumor-Infiltrating Lymphocytes for Patients With Metastatic Melanoma. J Clin Oncol. 2016;34(20):2389–97. 10.1200/JCO.2016.66.7220 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 18. Park JH, Geyer MB, Brentjens RJ: CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. Blood. 2016;127(26):3312–20. 10.1182/blood-2016-02-629063 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 19. Brentjens RJ, Rivière I, Park JH, et al. : Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118(18):4817–28. 10.1182/blood-2011-04-348540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Martinet L, Garrido I, Filleron T, et al. : Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res. 2011;71(17):5678–87. 10.1158/0008-5472.CAN-11-0431 [DOI] [PubMed] [Google Scholar]
  • 21. Prosser ME, Brown CE, Shami AF, et al. : Tumor PD-L1 co-stimulates primary human CD8 + cytotoxic T cells modified to express a PD1:CD28 chimeric receptor. Mol Immunol. 2012;51(3–4):263–72. 10.1016/j.molimm.2012.03.023 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 22. Foster AE, Dotti G, Lu A, et al. : Antitumor activity of EBV-specific T lymphocytes transduced with a dominant negative TGF-beta receptor. J Immunother. 2008;31(5):500–5. 10.1097/CJI.0b013e318177092b [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Spranger S, Bao R, Gajewski TF: Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015;523(7559):231–5. 10.1038/nature14404 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 24. Peinert S, Prince HM, Guru PM, et al. : Gene-modified T cells as immunotherapy for multiple myeloma and acute myeloid leukemia expressing the Lewis Y antigen. Gene Ther. 2010;17(5):678–86. 10.1038/gt.2010.21 [DOI] [PubMed] [Google Scholar]
  • 25. Louis CU, Savoldo B, Dotti G, et al. : Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118(23):6050–6. 10.1182/blood-2011-05-354449 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Ahmed N, Brawley VS, Hegde M, et al. : Human Epidermal Growth Factor Receptor 2 (HER2) -Specific Chimeric Antigen Receptor-Modified T Cells for the Immunotherapy of HER2-Positive Sarcoma. J Clin Oncol. 2015;33(15):1688–96. 10.1200/JCO.2014.58.0225 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 27. Krug LM, Dao T, Brown AB, et al. : WT1 peptide vaccinations induce CD4 and CD8 T cell immune responses in patients with mesothelioma and non-small cell lung cancer. Cancer Immunol Immunother. 2010;59(10):1467–79. 10.1007/s00262-010-0871-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Dohi S, Ohno S, Ohno Y, et al. : WT1 peptide vaccine stabilized intractable ovarian cancer patient for one year: a case report. Anticancer Res. 2011;31(7):2441–5. [PubMed] [Google Scholar]
  • 29. Gargett T, Brown MP: The inducible caspase-9 suicide gene system as a “safety switch” to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front Pharmacol. 2014;5:235. 10.3389/fphar.2014.00235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Ciceri F, Bonini C, Stanghellini MT, et al. : Infusion of suicide-gene-engineered donor lymphocytes after family haploidentical haemopoietic stem-cell transplantation for leukaemia (the TK007 trial): a non-randomised phase I-II study. Lancet Oncol. 2009;10(5):489–500. 10.1016/S1470-2045(09)70074-9 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 31. Lee DW, Gardner R, Porter DL, et al. : Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124(2):188–95. 10.1182/blood-2014-05-552729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Senzer N, Barve M, Nemunaitis J, 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 Vaccin. 2013;4:209 10.4172/2157-7560.1000209 [DOI] [Google Scholar]
  • 33. Ghisoli M, Barve M, Mennel R, et al. : Three-year Follow up of GMCSF/bi-shRNA furin DNA-transfected Autologous Tumor Immunotherapy (Vigil) in Metastatic Advanced Ewing's Sarcoma. Mol Ther. 2016;24(8):1478–83. 10.1038/mt.2016.86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Oh J, Barve M, Matthews CM, et al. : Phase II study of Vigil® DNA engineered immunotherapy as maintenance in advanced stage ovarian cancer. Gynecol Oncol. 2016;143(3):504–10. 10.1016/j.ygyno.2016.09.018 [DOI] [PubMed] [Google Scholar]
  • 35. Barve M, Kuhn J, Lamont J, et al. : Follow-up of bi-shRNA furin / GM-CSF Engineered Autologous Tumor Cell (EATC) Immunotherapy Vigil ® in patients with advanced melanoma. Biomed Genet Genomics. 2016;1 10.15761/bgg.1000116 [DOI] [Google Scholar]
  • 36. Maus MV, Haas AR, Beatty GL, et al. : T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013;1(1):26–31. 10.1158/2326-6066.CIR-13-0006 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 37. Hasegawa K, Nakamura T, Harvey M, et al. : The use of a tropism-modified measles virus in folate receptor-targeted virotherapy of ovarian cancer. Clin Cancer Res. 2006;12(20 Pt 1):6170–8. 10.1158/1078-0432.CCR-06-0992 [DOI] [PubMed] [Google Scholar]
  • 38. Park JR, Digiusto DL, Slovak M, et al. : Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15(4):825–33. 10.1038/sj.mt.6300104 [DOI] [PubMed] [Google Scholar]
  • 39. Pule MA, Savoldo B, Myers GD, et al. : Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14(11):1264–70. 10.1038/nm.1882 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Westwood JA, Smyth MJ, Teng MW, et al. : Adoptive transfer of T cells modified with a humanized chimeric receptor gene inhibits growth of Lewis-Y-expressing tumors in mice. Proc Natl Acad Sci USA. 2005;102(52):19051–6. 10.1073/pnas.0504312102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Morgan RA, Yang JC, Kitano M, et al. : Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18(4):843–51. 10.1038/mt.2010.24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Zhao Y, Moon E, Carpenito C, et al. : Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer Res. 2010;70(22):9053–61. 10.1158/0008-5472.CAN-10-2880 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Till BG, Jensen MC, Wang J, et al. : CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results. Blood. 2012;119(17):3940–50. 10.1182/blood-2011-10-387969 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 44. Guest RD, Kirillova N, Mowbray S, et al. : Definition and application of good manufacturing process-compliant production of CEA-specific chimeric antigen receptor expressing T-cells for phase I/II clinical trial. Cancer Immunol Immunother. 2014;63(2):133–45. 10.1007/s00262-013-1492-9 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 45. Koneru M, Purdon TJ, Spriggs D, et al. : IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo. Oncoimmunology. 2015;4(3):e994446. 10.4161/2162402X.2014.994446 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 46. Lamers CH, Langeveld SC, Groot-van Ruijven CM, et al. : Gene-modified T cells for adoptive immunotherapy of renal cell cancer maintain transgene-specific immune functions in vivo. Cancer Immunol Immunother. 2007;56(12):1875–83. 10.1007/s00262-007-0330-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Weijtens ME, Willemsen RA, van Krimpen BA, et al. : Chimeric scFv/gamma receptor-mediated T-cell lysis of tumor cells is coregulated by adhesion and accessory molecules. Int J Cancer. 1998;77(2):181–7. [DOI] [PubMed] [Google Scholar]
  • 48. Petrausch U, Schuberth PC, Hagedorn C, et al. : Re-directed T cells for the treatment of fibroblast activation protein (FAP)-positive malignant pleural mesothelioma (FAPME-1). BMC Cancer. 2012;12:615. 10.1186/1471-2407-12-615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Willemsen RA, Debets R, Hart E, et al. : A phage display selected fab fragment with MHC class I-restricted specificity for MAGE-A1 allows for retargeting of primary human T lymphocytes. Gene Ther. 2001;8(21):1601–8. 10.1038/sj.gt.3301570 [DOI] [PubMed] [Google Scholar]
  • 50. Willemsen RA, Ronteltap C, Chames P, et al. : T cell retargeting with MHC class I-restricted antibodies: the CD28 costimulatory domain enhances antigen-specific cytotoxicity and cytokine production. J Immunol. 2005;174(12):7853–8. 10.4049/jimmunol.174.12.7853 [DOI] [PubMed] [Google Scholar]
  • 51. Schuberth PC, Jakka G, Jensen SM, et al. : Effector memory and central memory NY-ESO-1-specific re-directed T cells for treatment of multiple myeloma. Gene Ther. 2013;20(4):386–95. 10.1038/gt.2012.48 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 52. Wilkie S, Picco G, Foster J, et al. : Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptor. J Immunol. 2008;180(7):4901–9. 10.4049/jimmunol.180.7.4901 [DOI] [PubMed] [Google Scholar]
  • 53. Chinnasamy D, Yu Z, Theoret MR, et al. : Gene therapy using genetically modified lymphocytes targeting VEGFR-2 inhibits the growth of vascularized syngenic tumors in mice. J Clin Invest. 2010;120(11):3953–68. 10.1172/JCI43490 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 54. Niederman TMJ, Ghogawala Z, Carter BS, et al. : Antitumor activity of cytotoxic T lymphocytes engineered to target vascular endothelial growth factor receptors. Proc Natl Acad Sci USA. 2002;99(10):7009–14. 10.1073/pnas.092562399 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Zhang L, Zhu W, Li J, et al. : Clinical outcome of immunotherapy with dendritic cell vaccine and cytokine-induced killer cell therapy in hepatobiliary and pancreatic cancer. Mol Clin Oncol. 2016;4(1):129–33. 10.3892/mco.2015.660 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 56. Escudier B, Dorval T, Chaput N, et al. : Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of the first phase I clinical trial. J Transl Med. 2005;3(1):10. 10.1186/1479-5876-3-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Morse MA, Garst J, Osada T, et al. : A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J Transl Med. 2005;3(1):9. 10.1186/1479-5876-3-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Besse B, Charrier M, Lapierre V, et al. : Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2016;5(4):e1071008. 10.1080/2162402X.2015.1071008 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 59. Dai S, Wei D, Wu Z, et al. : Phase I clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Mol Ther. 2008;16(4):782–90. 10.1038/mt.2008.1 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from F1000Research are provided here courtesy of F1000 Research Ltd

RESOURCES