Talkin’ Toxins: From Coley’s to Modern Cancer Immunotherapy
Abstract
:1. Introduction
2. Pivotal Observations in Cancer Immunotherapy
2.1. The Story of Coley’s Toxins
2.2. Evidence the Immune System Targets Cancer
3. Immunomodulatory Agents
3.1. Cytokine Therapy
3.2. Immune Checkpoint Inhibitors
4. Vaccines
4.1. Tumor Antigens
4.2. Therapeutic Cancer Vaccines
4.3. Preventive Cancer Vaccines
4.4. Oncolytic Virotherapy
5. Adoptive Cell Therapy
5.1. Tumor-Infiltrating Lymphocytes and Engineered T-Cell Receptors
5.2. Chimeric Antigen Receptor T Cell (CAR-T Cell) Therapy
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Couzin-Frankel, J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013, 342, 1432–1433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Challis, G.B.; Stam, H.J. The spontaneous regression of cancer. A review of cases from 1900 to 1987. Acta Oncol. 1990, 29, 545–550. [Google Scholar] [CrossRef] [PubMed]
- Papac, R.J. Spontaneous regression of cancer. Cancer Treat. Rev. 1996, 22, 395–423. [Google Scholar] [CrossRef]
- Smith, J.L.; Stehlin, J.S. Spontaneous regression of primary malignant melanomas with regional metastases. Cancer 1965, 18, 1399–1415. [Google Scholar] [CrossRef]
- Haridy, Y.; Witzmann, F.; Asbach, P.; Schoch, R.R.; Fröbisch, N.; Rothschild, B.M. Triassic Cancer-Osteosarcoma in a 240-Million-Year-Old Stem-Turtle. JAMA Oncol. 2019, 5, 425–426. [Google Scholar] [CrossRef] [PubMed]
- Coley, W.B., II. contribution to the knowledge of sarcoma. Ann. Surg. 1891, 14, 199–220. [Google Scholar] [CrossRef] [PubMed]
- Coley, W.B. The treatment of malignant tumors by repeated inoculations of erysipelas: With a report of ten original cases. 1. Am. J. Med. Sci. 1893, 105, 487. [Google Scholar] [CrossRef]
- Stevens, D.L.; Bryant, A.E. Impetigo, erysipelas and cellulitis. In Streptococcus Pyogenes: Basic Biology to Clinical Manifestations; Ferretti, J.J., Stevens, D.L., Fischetti, V.A., Eds.; University of Oklahoma Health Sciences Center: Oklahoma City, OK, USA, 2016. [Google Scholar]
- Fehleisen, F. Die Aetiologie des Erysipels; Theodor Fischer: Berlin, Germany, 1883. [Google Scholar]
- Cann, S.A.H.; Van Netten, J.P.; Van Netten, C. Dr William Coley and tumour regression: A place in history or in the future. Postgrad. Med. 2003, 79, 672–680. [Google Scholar]
- McCarthy, E.F. The toxins of William B. Coley and the treatment of bone and soft-tissue sarcomas. Iowa Orthop. J. 2006, 26, 154–158. [Google Scholar]
- Coley, W.B. The therapeutic value of the mixed toxins of the streptococcus of erysipelas and bacillus prodigiosus in the treatment of one hundred and sixty cases. Am. J. Med. Sci. 1896, 112, 251. [Google Scholar] [CrossRef] [Green Version]
- Coley, W.B. The Treatment of Inoperable Sarcoma by Bacterial Toxins (the Mixed Toxins of the Streptococcus erysipelas and the Bacillus prodigiosus). Proc. R. Soc. Med. 1910, 3, 1–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johnston, B.J.; Novales, E.T. Clinical effect of Coley’s toxin. II. A seven-year study. Cancer Chemother. Rep. 1962, 21, 43–68. [Google Scholar] [PubMed]
- Ehrlich, P. Über den jetzigen Stand der Karzinomforschung. Ned Tijdschr Geneeskd. 1909, 5, 273–290. [Google Scholar]
- Burnet, F.M. The concept of immunological surveillance. Immunol. Asp. Neoplasia 1970, 13, 1–27. [Google Scholar]
- Thomas, L. On immunosurveillance in human cancer. Yale J. Biol. Med. 1982, 55, 329–333. [Google Scholar]
- Gross, L. Intradermal immunization of C3H mice against a sarcoma that originated in an animal of the same line. Cancer Res. 1943, 3, 326–333. [Google Scholar]
- Prehn, R.T.; Main, J.M. Immunity to methylcholanthrene-induced sarcomas. J. Natl. Cancer Inst. 1957, 18, 769–778. [Google Scholar]
- Shinkai, Y.; Rathbun, G.; Lam, K.P.; Oltz, E.M.; Stewart, V.; Mendelsohn, M.; Charron, J.; Datta, M.; Young, F.; Stall, A.M. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell 1992, 68, 855–867. [Google Scholar] [CrossRef]
- Shankaran, V.; Ikeda, H.; Bruce, A.T.; White, J.M.; Swanson, P.E.; Old, L.J.; Schreiber, R.D. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410, 1107–1111. [Google Scholar] [CrossRef]
- Cretney, E.; Takeda, K.; Yagita, H.; Glaccum, M.; Peschon, J.J.; Smyth, M.J. Increased susceptibility to tumor initiation and metastasis in TNF-related apoptosis-inducing ligand-deficient mice. J. Immunol. 2002, 168, 1356–1361. [Google Scholar] [CrossRef] [Green Version]
- van den Broek, M.E.; Kägi, D.; Ossendorp, F.; Toes, R.; Vamvakas, S.; Lutz, W.K.; Melief, C.J.; Zinkernagel, R.M.; Hengartner, H. Decreased tumor surveillance in perforin-deficient mice. J. Exp. Med. 1996, 184, 1781–1790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mayor, P.C.; Eng, K.H.; Singel, K.L.; Abrams, S.I.; Odunsi, K.; Moysich, K.B.; Fuleihan, R.; Garabedian, E.; Lugar, P.; Ochs, H.D.; et al. Cancer in primary immunodeficiency diseases: Cancer incidence in the United States Immune Deficiency Network Registry. J. Allergy Clin. Immunol. 2018, 141, 1028–1035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Engels, E.A.; Pfeiffer, R.M.; Fraumeni, J.F.; Kasiske, B.L.; Israni, A.K.; Snyder, J.J.; Wolfe, R.A.; Goodrich, N.P.; Bayakly, A.R.; Clarke, C.A.; et al. Spectrum of cancer risk among US solid organ transplant recipients. JAMA 2011, 306, 1891–1901. [Google Scholar] [CrossRef] [PubMed]
- Goedert, J.J.; Coté, T.R.; Virgo, P.; Scoppa, S.M.; Kingma, D.W.; Gail, M.H.; Jaffe, E.S.; Biggar, R.J. Spectrum of AIDS-associated malignant disorders. Lancet 1998, 351, 1833–1839. [Google Scholar] [CrossRef]
- Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koebel, C.M.; Vermi, W.; Swann, J.B.; Zerafa, N.; Rodig, S.J.; Old, L.J.; Smyth, M.J.; Schreiber, R.D. Adaptive immunity maintains occult cancer in an equilibrium state. Nature 2007, 450, 903–907. [Google Scholar] [CrossRef]
- DuPage, M.; Mazumdar, C.; Schmidt, L.M.; Cheung, A.F.; Jacks, T. Expression of tumour-specific antigens underlies cancer immunoediting. Nature 2012, 482, 405–409. [Google Scholar] [CrossRef] [Green Version]
- Miescher, S.; Whiteside, T.L.; Moretta, L.; von Fliedner, V. Clonal and frequency analyses of tumor-infiltrating T lymphocytes from human solid tumors. J. Immunol. 1987, 138, 4004–4011. [Google Scholar]
- Li, Y.; Patel, S.P.; Roszik, J.; Qin, Y. Hypoxia-Driven Immunosuppressive Metabolites in the Tumor Microenvironment: New Approaches for Combinational Immunotherapy. Front. Immunol. 2018, 9, 1591. [Google Scholar] [CrossRef] [Green Version]
- Barsoum, I.B.; Koti, M.; Siemens, D.R.; Graham, C.H. Mechanisms of hypoxia-mediated immune escape in cancer. Cancer Res. 2014, 74, 7185–7190. [Google Scholar] [CrossRef] [Green Version]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef] [PubMed]
- Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef] [PubMed]
- Butt, A.Q.; Mills, K.H.G. Immunosuppressive networks and checkpoints controlling antitumor immunity and their blockade in the development of cancer immunotherapeutics and vaccines. Oncogene 2014, 33, 4623–4631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sakaguchi, S.; Yamaguchi, T.; Nomura, T.; Ono, M. Regulatory T cells and immune tolerance. Cell 2008, 133, 775–787. [Google Scholar] [CrossRef] [Green Version]
- Gery, I.; Gershon, R.K.; Waksman, B.H. Potentiation of the T-lymphocyte response to mitogens. I. The responding cell. J. Exp. Med. 1972, 136, 128–142. [Google Scholar] [CrossRef] [Green Version]
- Mier, J.W.; Gallo, R.C. Purification and some characteristics of human T-cell growth factor from phytohemagglutinin-stimulated lymphocyte-conditioned media. Proc. Natl. Acad. Sci. USA 1980, 77, 6134–6138. [Google Scholar] [CrossRef] [Green Version]
- Welte, K.; Wang, C.Y.; Mertelsmann, R.; Venuta, S.; Feldman, S.P.; Moore, M.A. Purification of human interleukin 2 to apparent homogeneity and its molecular heterogeneity. J. Exp. Med. 1982, 156, 454–464. [Google Scholar] [CrossRef] [Green Version]
- Rosenberg, S.A.; Mulé, J.J.; Spiess, P.J.; Reichert, C.M.; Schwarz, S.L. Regression of established pulmonary metastases and subcutaneous tumor mediated by the systemic administration of high-dose recombinant interleukin 2. J. Exp. Med. 1985, 161, 1169–1188. [Google Scholar] [CrossRef]
- Eberlein, T.J.; Rosenstein, M.; Rosenberg, S.A. Regression of a disseminated syngeneic solid tumor by systemic transfer of lymphoid cells expanded in interleukin 2. J. Exp. Med. 1982, 156, 385–397. [Google Scholar] [CrossRef] [Green Version]
- Rosenberg, S.A.; Lotze, M.T.; Muul, L.M.; Leitman, S.; Chang, A.E.; Ettinghausen, S.E.; Matory, Y.L.; Skibber, J.M.; Shiloni, E.; Vetto, J.T. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N. Engl. J. Med. 1985, 313, 1485–1492. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Lotze, M.T.; Muul, L.M.; Chang, A.E.; Avis, F.P.; Leitman, S.; Linehan, W.M.; Robertson, C.N.; Lee, R.E.; Rubin, J.T. A progress report on the treatment of 157 patients with advanced cancer using lymphokine-activated killer cells and interleukin-2 or high-dose interleukin-2 alone. N. Engl. J. Med. 1987, 316, 889–897. [Google Scholar] [CrossRef] [PubMed]
- Schwartz, R.N.; Stover, L.; Dutcher, J. Managing toxicities of high-dose interleukin-2. Oncology 2002, 16, 11–20. [Google Scholar] [PubMed]
- Rosenberg, S.A.; Yang, J.C.; Topalian, S.L.; Schwartzentruber, D.J.; Weber, J.S.; Parkinson, D.R.; Seipp, C.A.; Einhorn, J.H.; White, D.E. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA 1994, 271, 907–913. [Google Scholar] [CrossRef] [PubMed]
- Dillman, R.O.; Church, C.; Oldham, R.K.; West, W.H.; Schwartzberg, L.; Birch, R. Inpatient continuous-infusion interleukin-2 in 788 patients with cancer. The National Biotherapy Study Group experience. Cancer 1993, 71, 2358–2370. [Google Scholar] [CrossRef]
- Lee, S.; Margolin, K. Cytokines in cancer immunotherapy. Cancers 2011, 3, 3856–3893. [Google Scholar] [CrossRef]
- Ku, C.C.; Murakami, M.; Sakamoto, A.; Kappler, J.; Marrack, P. Control of homeostasis of CD8+ memory T cells by opposing cytokines. Science 2000, 288, 675–678. [Google Scholar] [CrossRef]
- Evans, R.; Fuller, J.A.; Christianson, G.; Krupke, D.M. IL-15 mediates anti-tumor effects after cyclophosphamide injection of tumor-bearing mice and enhances adoptive immunotherapy: The potential role of NK cell. Cell. Immunol. 1997, 178, 197. [Google Scholar] [CrossRef]
- Kobayashi, H.; Dubois, S.; Sato, N.; Sabzevari, H.; Sakai, Y.; Waldmann, T.A.; Tagaya, Y. Role of trans-cellular IL-15 presentation in the activation of NK cell-mediated killing, which leads to enhanced tumor immunosurveillance. Blood 2005, 105, 721–727. [Google Scholar] [CrossRef] [Green Version]
- Waldmann, T.A. The biology of interleukin-2 and interleukin-15: Implications for cancer therapy and vaccine design. Nat. Rev. Immunol. 2006, 6, 595–601. [Google Scholar] [CrossRef]
- Bessard, A.; Solé, V.; Bouchaud, G.; Quéméner, A. High antitumor activity of RLI, an interleukin-15 (IL-15)–IL-15 receptor α fusion protein, in metastatic melanoma and colorectal cancer. Mol. Cancer Ther. 2009, 8, 2736–2745. [Google Scholar] [CrossRef] [Green Version]
- Howie, J.W. Experiments with interferon in man: A report to the medical research council from the scientific committee on interferon. Lancet 1965, 1, 505. [Google Scholar] [PubMed]
- Müller, U.; Steinhoff, U.; Reis, L.F.; Hemmi, S.; Pavlovic, J.; Zinkernagel, R.M.; Aguet, M. Functional role of type I and type II interferons in antiviral defense. Science 1994, 264, 1918–1921. [Google Scholar] [CrossRef]
- Waldmann, T.A. Cytokines in cancer immunotherapy. Cold Spring Harb. Perspect. Biol. 2017, 10. [Google Scholar] [CrossRef] [PubMed]
- Cantell, K.; Hirvonen, S.; Koistinen, V. [71] Partial purification of human leukocyte interferon on a large scale. In Methods in Enzymology; Academic Press: Cambridge, MA, USA, 1981. [Google Scholar]
- Merigan, T.C.; Sikora, K.; Breeden, J.H.; Levy, R.; Rosenberg, S.A. Preliminary observations on the effect of human leukocyte interferon in non-Hodgkin’s lymphoma. N. Engl. J. Med. 1978, 299, 1449–1453. [Google Scholar] [CrossRef] [PubMed]
- Quesada, J.R.; Hersh, E.M.; Gutterman, J.U. Hairy cell leukemia: Induction of remission with alpha interferon. Blood 1983, 62 (Suppl. 1), 207. [Google Scholar] [CrossRef] [PubMed]
- Quesada, J.R.; Swanson, D.A.; Trindade, A.; Gutterman, J.U. Renal cell carcinoma: Antitumor effects of leukocyte interferon. Cancer Res. 1983, 43, 940–947. [Google Scholar]
- Krown, S.E.; Burk, M.W.; Kirkwood, J.M.; Kerr, D. Human leukocyte (alpha) interferon in metastatic malignant melanoma. Am. Cancer Soc. Phase II Trial 1984, 68, 723–726. [Google Scholar]
- Kirkwood, J.M.; Ibrahim, J.G.; Sondak, V.K.; Richards, J.; Flaherty, L.E.; Ernstoff, M.S.; Smith, T.J.; Rao, U.; Steele, M.; Blum, R.H. High-and low-dose interferon alfa-2b in high-risk melanoma: First analysis of intergroup trial E1690/S9111/C9190. J. Clin. Oncol. 2000, 18, 2444–2458. [Google Scholar] [CrossRef]
- Hercus, T.R.; Thomas, D.; Guthridge, M.A.; Ekert, P.G.; King-Scott, J.; Parker, M.W.; Lopez, A.F. The granulocyte-macrophage colony-stimulating factor receptor: Linking its structure to cell signaling and its role in disease. Blood 2009, 114, 1289–1298. [Google Scholar] [CrossRef] [Green Version]
- Dranoff, G.; Jaffee, E.; Lazenby, A.; Golumbek, P.; Levitsky, H.; Brose, K.; Jackson, V.; Hamada, H.; Pardoll, D.; Mulligan, R.C. 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, 3539–3543. [Google Scholar] [CrossRef] [Green Version]
- Soiffer, R.; Lynch, T.; Mihm, M.; Jung, K.; Rhuda, C.; Schmollinger, J.C.; Hodi, F.S.; Liebster, L.; Lam, P.; Mentzer, S.; et al. Vaccination with irradiated autologous melanoma cells engineered to secrete human granulocyte-macrophage colony-stimulating factor generates potent antitumor immunity in patients with metastatic melanoma. Proc. Natl. Acad. Sci. USA 1998, 95, 13141–13146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Small, E.J.; Reese, D.M.; Um, B.; Whisenant, S.; Dixon, S.C.; Figg, W.D. Therapy of advanced prostate cancer with granulocyte macrophage colony-stimulating factor. Clin. Cancer Res. 1999, 5, 1738–1744. [Google Scholar] [PubMed]
- Lawson, D.H.; Lee, S.; Zhao, F.; Tarhini, A.A.; Margolin, K.A.; Ernstoff, M.S.; Atkins, M.B.; Cohen, G.I.; Whiteside, T.L.; Butterfield, L.H.; et al. Randomized, Placebo-Controlled, Phase III Trial of Yeast-Derived Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) Versus Peptide Vaccination Versus GM-CSF Plus Peptide Vaccination Versus Placebo in Patients With No Evidence of Disease After Complete Surgical Resection of Locally Advanced and/or Stage IV Melanoma: A Trial of the Eastern Cooperative Oncology Group-American College of Radiology Imaging Network Cancer Research Group (E4697). J. Clin. Oncol. 2015, 33, 4066–4076. [Google Scholar] [CrossRef] [PubMed]
- Hodi, F.S.; Lee, S.; McDermott, D.F.; Rao, U.N.; Butterfield, L.H.; Tarhini, A.A.; Leming, P.; Puzanov, I.; Shin, D.; Kirkwood, J.M. Ipilimumab plus sargramostim vs ipilimumab alone for treatment of metastatic melanoma: A randomized clinical trial. JAMA 2014, 312, 1744–1753. [Google Scholar] [CrossRef] [Green Version]
- O’Day, S.J.; Atkins, M.B.; Boasberg, P. Phase II multicenter trial of maintenance biotherapy after induction concurrent Biochemotherapy for patients with metastatic melanoma. J. Clin. Oncol. 2009, 27, 6207–6212. [Google Scholar] [CrossRef]
- Tang, H.; Qiao, J.; Fu, Y.-X. Immunotherapy and tumor microenvironment. Cancer Lett. 2016, 370, 85–90. [Google Scholar] [CrossRef]
- Mortarini, R.; Piris, A.; Maurichi, A.; Molla, A.; Bersani, I.; Bono, A.; Bartoli, C.; Santinami, M.; Lombardo, C.; Ravagnani, F.; et al. Lack of terminally differentiated tumor-specific CD8+ T cells at tumor site in spite of antitumor immunity to self-antigens in human metastatic melanoma. Cancer Res. 2003, 63, 2535–2545. [Google Scholar]
- Deng, L.; Zhang, H.; Luan, Y.; Zhang, J.; Xing, Q.; Dong, S.; Wu, X.; Liu, M.; Wang, S. Accumulation of foxp3+ T regulatory cells in draining lymph nodes correlates with disease progression and immune suppression in colorectal cancer patients. Clin. Cancer Res. 2010, 16, 4105–4112. [Google Scholar] [CrossRef] [Green Version]
- Gallimore, A.; Glithero, A.; Godkin, A.; Tissot, A.C.; Plückthun, A.; Elliott, T.; Hengartner, H.; Zinkernagel, R. Induction and exhaustion of lymphocytic choriomeningitis virus-specific cytotoxic T lymphocytes visualized using soluble tetrameric major histocompatibility complex class I-peptide complexes. J. Exp. Med. 1998, 187, 1383–1393. [Google Scholar] [CrossRef]
- Joffre, O.; Nolte, M.A.; Spörri, R.; Reis e Sousa, C. Inflammatory signals in dendritic cell activation and the induction of adaptive immunity. Immunol. Rev. 2009, 227, 234–247. [Google Scholar] [CrossRef]
- Shahinian, A.; Pfeffer, K.; Lee, K.P.; Kündig, T.M.; Kishihara, K.; Wakeham, A.; Kawai, K.; Ohashi, P.S.; Thompson, C.B.; Mak, T.W. Differential T cell costimulatory requirements in CD28-deficient mice. Science 1993, 261, 609–612. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Flies, D.B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 2013, 13, 227–242. [Google Scholar] [CrossRef] [PubMed]
- Hargadon, K.M.; Johnson, C.E.; Williams, C.J. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int. Immunopharmacol. 2018, 62, 29–39. [Google Scholar] [CrossRef] [PubMed]
- Brunet, J.F.; Denizot, F.; Luciani, M.F.; Roux-Dosseto, M.; Suzan, M.; Mattei, M.G.; Golstein, P. A new member of the immunoglobulin superfamily—CTLA-4. Nature 1987, 328, 267–270. [Google Scholar] [CrossRef]
- Walunas, T.L.; Lenschow, D.J.; Bakker, C.Y.; Linsley, P.S.; Freeman, G.J.; Green, J.M.; Thompson, C.B.; Bluestone, J.A. CTLA-4 can function as a negative regulator of T cell activation. Immunity 1994, 1, 405–413. [Google Scholar] [CrossRef]
- Qureshi, O.S.; Zheng, Y.; Nakamura, K.; Attridge, K.; Manzotti, C.; Schmidt, E.M.; Baker, J.; Jeffery, L.E.; Kaur, S.; Briggs, Z.; et al. Trans-endocytosis of CD80 and CD86: A molecular basis for the cell-extrinsic function of CTLA-4. Science 2011, 332, 600–603. [Google Scholar] [CrossRef] [Green Version]
- Krummel, M.F.; Allison, J.P. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J. Exp. Med. 1995, 182, 459–465. [Google Scholar] [CrossRef] [Green Version]
- Tivol, E.A.; Borriello, F.; Schweitzer, A.N.; Lynch, W.P.; Bluestone, J.A.; Sharpe, A.H. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 1995, 3, 541–547. [Google Scholar] [CrossRef] [Green Version]
- Waterhouse, P.; Penninger, J.M.; Timms, E.; Wakeham, A.; Shahinian, A.; Lee, K.P.; Thompson, C.B.; Griesser, H.; Mak, T.W. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 1995, 270, 985–988. [Google Scholar] [CrossRef]
- Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef] [Green Version]
- Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef] [PubMed]
- Robert, C.; Thomas, L.; Bondarenko, I.; O’Day, S.; Weber, J.; Garbe, C.; Lebbe, C.; Baurain, J.-F.; Testori, A.; Grob, J.-J.; et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 2011, 364, 2517–2526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schadendorf, D.; Hodi, F.S.; Robert, C.; Weber, J.S.; Margolin, K.; Hamid, O.; Patt, D.; Chen, T.-T.; Berman, D.M.; Wolchok, J.D. Pooled Analysis of Long-Term Survival Data From Phase II and Phase III Trials of Ipilimumab in Unresectable or Metastatic Melanoma. J. Clin. Oncol. 2015, 33, 1889–1894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992, 11, 3887–3895. [Google Scholar] [CrossRef] [PubMed]
- Nishimura, H.; Nose, M.; Hiai, H.; Minato, N.; Honjo, T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 1999, 11, 141–151. [Google Scholar] [CrossRef] [Green Version]
- Dong, H.; Zhu, G.; Tamada, K.; Chen, L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 1999, 5, 1365–1369. [Google Scholar] [CrossRef]
- Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C.; et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef] [Green Version]
- Iwai, Y.; Ishida, M.; Tanaka, Y.; Okazaki, T.; Honjo, T.; Minato, N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. USA 2002, 99, 12293–12297. [Google Scholar] [CrossRef] [Green Version]
- Curiel, T.J.; Wei, S.; Dong, H.; Alvarez, X.; Cheng, P.; Mottram, P.; Krzysiek, R.; Knutson, K.L.; Daniel, B.; Zimmermann, M.C.; et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat. Med. 2003, 9, 562–567. [Google Scholar] [CrossRef]
- Strome, S.E.; Dong, H.; Tamura, H.; Voss, S.G.; Flies, D.B.; Tamada, K.; Salomao, D.; Cheville, J.; Hirano, F.; Lin, W.; et al. B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res. 2003, 63, 6501–6505. [Google Scholar]
- Iwai, Y.; Terawaki, S.; Honjo, T. PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int. Immunol. 2005, 17, 133–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hamanishi, J.; Mandai, M.; Iwasaki, M.; Okazaki, T.; Tanaka, Y.; Yamaguchi, K.; Higuchi, T.; Yagi, H.; Takakura, K.; Minato, N.; et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc. Natl. Acad. Sci. USA 2007, 104, 3360–3365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.M.; Hwu, W.-J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
- Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. KEYNOTE-001 Investigators Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef]
- Weber, J.S.; D’Angelo, S.P.; Minor, D.; Hodi, F.S.; Gutzmer, R.; Neyns, B.; Hoeller, C.; Khushalani, N.I.; Miller, W.H.; Lao, C.D.; et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015, 16, 375–384. [Google Scholar] [CrossRef]
- Schachter, J.; Ribas, A.; Long, G.V.; Arance, A.; Grob, J.-J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus ipilimumab for advanced melanoma: Final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet 2017, 390, 1853–1862. [Google Scholar] [CrossRef]
- Powles, T.; Durán, I.; van der Heijden, M.S.; Loriot, Y.; Vogelzang, N.J.; De Giorgi, U.; Oudard, S.; Retz, M.M.; Castellano, D.; Bamias, A.; et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2018, 391, 748–757. [Google Scholar] [CrossRef]
- Rittmeyer, A.; Barlesi, F.; Waterkamp, D.; Park, K.; Ciardiello, F.; von Pawel, J.; Gadgeel, S.M.; Hida, T.; Kowalski, D.M.; Dols, M.C.; et al. OAK Study Group Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): A phase 3, open-label, multicentre randomised controlled trial. Lancet 2017, 389, 255–265. [Google Scholar] [CrossRef]
- Narayan, P.; Wahby, S.; Gao, J.J.; Amiri-Kordestani, L.; Ibrahim, A.; Bloomquist, E.; Tang, S.; Xu, Y.; Liu, J.; Fu, W.; et al. FDA Approval Summary: Atezolizumab plus paclitaxel protein-bound for the treatment of patients with advanced or metastatic TNBC whose tumors express PD-L1. Clin. Cancer Res. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaufman, H.L.; Russell, J.; Hamid, O.; Bhatia, S.; Terheyden, P.; D’Angelo, S.P.; Shih, K.C.; Lebbé, C.; Linette, G.P.; Milella, M.; et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: A multicentre, single-group, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 1374–1385. [Google Scholar] [CrossRef] [Green Version]
- Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Arén Frontera, O.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.; Barthélémy, P.; Porta, C.; George, S.; et al. CheckMate 214 Investigators Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2018, 378, 1277–1290. [Google Scholar] [CrossRef]
- Jenkins, R.W.; Barbie, D.A.; Flaherty, K.T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer 2018, 118, 9–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [Green Version]
- Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [Green Version]
- Hanna, M.G.; Peters, L.C. Specific immunotherapy of established visceral micrometastases by BCG-tumor cell vaccine alone or as an adjunct to surgery. Cancer 1978, 42, 2613–2625. [Google Scholar] [CrossRef]
- Murray, D.R.; Cassel, W.A.; Torbin, A.H.; Olkowski, Z.L.; Moore, M.E. Viral oncolysate in the management of malignant melanoma. II. Clinical studies. Cancer 1977, 40, 680–686. [Google Scholar] [CrossRef]
- McIllmurray, M.B.; Embleton, M.J.; Reeves, W.G.; Langman, M.J.; Deane, M. Controlled trial of active immunotherapy in management of stage IIB malignant melanoma. Br. Med. J. 1977, 1, 540–542. [Google Scholar] [CrossRef] [Green Version]
- van der Bruggen, P.; Traversari, C.; Chomez, P.; Lurquin, C.; De Plaen, E.; Van den Eynde, B.; Knuth, A.; Boon, T. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 1991, 254, 1643–1647. [Google Scholar] [CrossRef] [PubMed]
- Cheever, M.A.; Allison, J.P.; Ferris, A.S.; Finn, O.J.; Hastings, B.M.; Hecht, T.T.; Mellman, I.; Prindiville, S.A.; Viner, J.L.; Weiner, L.M.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thomas, R.; Al-Khadairi, G.; Roelands, J.; Hendrickx, W.; Dermime, S.; Bedognetti, D.; Decock, J. NY-ESO-1 Based Immunotherapy of Cancer: Current Perspectives. Front. Immunol. 2018, 9, 947. [Google Scholar] [CrossRef] [PubMed]
- Zajac, P.; Schultz-Thater, E.; Tornillo, L.; Sadowski, C.; Trella, E.; Mengus, C.; Iezzi, G.; Spagnoli, G.C. MAGE-A Antigens and Cancer Immunotherapy. Front. Med. 2017, 4, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turriziani, M.; Fantini, M.; Benvenuto, M. Carcinoembryonic antigen (CEA)-based cancer vaccines: Recent patents and antitumor effects from experimental models to clinical trials. Recent Pat. Anti-Cancer Drug Discov. 2012, 7, 265–296. [Google Scholar] [CrossRef]
- Evdokimova, V.N.; Butterfield, L.H. Alpha-fetoprotein and other tumour-associated antigens for immunotherapy of hepatocellular cancer. Expert Opin. Biol. Ther. 2008, 8, 325–336. [Google Scholar] [CrossRef] [PubMed]
- Westdorp, H.; Sköld, A.E.; Snijer, B.A.; Franik, S.; Mulder, S.F.; Major, P.P.; Foley, R.; Gerritsen, W.R.; de Vries, I.J.M. Immunotherapy for prostate cancer: Lessons from responses to tumor-associated antigens. Front. Immunol. 2014, 5, 191. [Google Scholar] [CrossRef] [Green Version]
- Scheuermann, R.H.; Racila, E. CD19 antigen in leukemia and lymphoma diagnosis and immunotherapy. Leuk. Lymphoma 1995, 18, 385–397. [Google Scholar] [CrossRef]
- Ayoub, N.M.; Al-Shami, K.M.; Yaghan, R.J. Immunotherapy for HER2-positive breast cancer: Recent advances and combination therapeutic approaches. Breast Cancer (Dove Med Press) 2019, 11, 53–69. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.-P.; Chen, W.; Schwarer, A.P.; Li, H. Telomerase in cancer immunotherapy. Biochim. Biophys. Acta 2010, 1805, 35–42. [Google Scholar] [CrossRef] [Green Version]
- Jiang, T.; Shi, T.; Zhang, H.; Hu, J.; Song, Y.; Wei, J.; Ren, S.; Zhou, C. Tumor neoantigens: From basic research to clinical applications. J. Hematol. Oncol. 2019, 12, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goydos, J.S.; Elder, E.; Whiteside, T.L.; Finn, O.J.; Lotze, M.T. A phase I trial of a synthetic mucin peptide vaccine. Induction of specific immune reactivity in patients with adenocarcinoma. J. Surg. Res. 1996, 63, 298–304. [Google Scholar] [CrossRef]
- Mukherji, B.; Chakraborty, N.G.; Yamasaki, S.; Okino, T.; Yamase, H.; Sporn, J.R.; Kurtzman, S.K.; Ergin, M.T.; Ozols, J.; Meehan, J. Induction of antigen-specific cytolytic T cells in situ in human melanoma by immunization with synthetic peptide-pulsed autologous antigen presenting cells. Proc. Natl. Acad. Sci. USA 1995, 92, 8078–8082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsu, F.J.; Benike, C.; Fagnoni, F.; Liles, T.M.; Czerwinski, D.; Taidi, B.; Engleman, E.G.; Levy, R. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat. Med. 1996, 2, 52–58. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.W.; Gulley, J.L. Poxviral vectors for cancer immunotherapy. Expert Opin. Biol. Ther. 2012, 12, 463–478. [Google Scholar] [CrossRef] [Green Version]
- Gallo, P.; Dharmapuri, S.; Cipriani, B.; Monaci, P. Adenovirus as vehicle for anticancer genetic immunotherapy. Gene Ther. 2005, 12 (Suppl. 1), S84–S91. [Google Scholar] [CrossRef] [Green Version]
- Roland, K.L.; Brenneman, K.E. Salmonella as a vaccine delivery vehicle. Expert Rev. Vaccines 2013, 12, 1033–1045. [Google Scholar] [CrossRef] [Green Version]
- Flickinger, J.C.; Rodeck, U.; Snook, A.E. Listeria monocytogenes as a Vector for Cancer Immunotherapy: Current Understanding and Progress. Vaccines 2018, 6, 48. [Google Scholar] [CrossRef] [Green Version]
- Jahanafrooz, Z.; Baradaran, B.; Mosafer, J.; Hashemzaei, M.; Rezaei, T.; Mokhtarzadeh, A.; Hamblin, M.R. Comparison of DNA and mRNA vaccines against cancer. Drug Discov. Today 2019. [Google Scholar] [CrossRef]
- Palucka, K.; Banchereau, J. Dendritic-cell-based therapeutic cancer vaccines. Immunity 2013, 39, 38–48. [Google Scholar] [CrossRef] [Green Version]
- Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. IMPACT Study Investigators Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 2010, 363, 411–422. [Google Scholar] [CrossRef] [Green Version]
- Peng, M.; Mo, Y.; Wang, Y.; Wu, P.; Zhang, Y.; Xiong, F.; Guo, C.; Wu, X.; Li, Y.; Li, X.; et al. Neoantigen vaccine: An emerging tumor immunotherapy. Mol. Cancer 2019, 18, 128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ott, P.A.; Hu, Z.; Keskin, D.B.; Shukla, S.A.; Sun, J.; Bozym, D.J.; Zhang, W.; Luoma, A.; Giobbie-Hurder, A.; Peter, L.; et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017, 547, 217–221. [Google Scholar] [CrossRef] [PubMed]
- Keskin, D.B.; Anandappa, A.J.; Sun, J.; Tirosh, I.; Mathewson, N.D.; Li, S.; Oliveira, G.; Giobbie-Hurder, A.; Felt, K.; Gjini, E.; et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature 2019, 565, 234–239. [Google Scholar] [CrossRef] [PubMed]
- Finn, O.J. The dawn of vaccines for cancer prevention. Nat. Rev. Immunol. 2018, 18, 183–194. [Google Scholar] [CrossRef]
- Chang, M.-H. Hepatitis B virus and cancer prevention. Recent Results Cancer Res. 2011, 188, 75–84. [Google Scholar] [CrossRef]
- Guo, F.; Cofie, L.E.; Berenson, A.B. Cervical cancer incidence in young U.S. females after human papillomavirus vaccine introduction. Am. J. Prev. Med. 2018, 55, 197–204. [Google Scholar] [CrossRef]
- Singh, R.; Bandyopadhyay, D. MUC1: A target molecule for cancer therapy. Cancer Biol. Ther. 2007, 6, 481–486. [Google Scholar] [CrossRef]
- Kimura, T.; McKolanis, J.R.; Dzubinski, L.A.; Islam, K.; Potter, D.M.; Salazar, A.M.; Schoen, R.E.; Finn, O.J. MUC1 vaccine for individuals with advanced adenoma of the colon: A cancer immunoprevention feasibility study. Cancer Prev. Res. 2013, 6, 18–26. [Google Scholar] [CrossRef] [Green Version]
- Lawler, S.E.; Speranza, M.-C.; Cho, C.-F.; Chiocca, E.A. Oncolytic viruses in cancer treatment: A review. JAMA Oncol. 2017, 3, 841–849. [Google Scholar] [CrossRef] [Green Version]
- Alvarez, C.A. NK cells impede glioblastoma virotherapy through NKp30 and NKp46 natural cytotoxicity receptors. Nat. Med. 2012, 18, 1827. [Google Scholar] [CrossRef] [Green Version]
- Yung, W.K.A.; Vence, L.M.; Gomez, C. Delta-24-RGD oncolytic adenovirus elicits anti-glioma immunity in an immunocompetent mouse model. PloS ONE 2014, 9, e97407. [Google Scholar]
- Nguyen, T.; Avci, N.G.; Shin, D.H.; Martinez, N. Tune up in situ autovaccination against solid tumors with oncolytic viruses. Cancers 2018, 10, 171. [Google Scholar] [CrossRef] [Green Version]
- Dock, G. The influence of complicating diseases upon leukaemia. Am. J. Med Sci. 1904, 127, 563. [Google Scholar] [CrossRef]
- Larson, C.; Oronsky, B.; Scicinski, J.; Fanger, G.R.; Stirn, M.; Oronsky, A.; Reid, T.R. Going viral: A review of replication-selective oncolytic adenoviruses. Oncotarget 2015, 6, 19976–19989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andtbacka, R.H.I.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Restifo, N.P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015, 348, 62–68. [Google Scholar] [CrossRef] [Green Version]
- Cohen, J.E.; Merims, S.; Frank, S.; Engelstein, R.; Peretz, T.; Lotem, M. Adoptive cell therapy: Past, present and future. Immunotherapy 2017, 9, 183–196. [Google Scholar] [CrossRef]
- Mitchison, N.A. Passive transfer of transplantation immunity. Nature 1953, 171, 267–268. [Google Scholar] [CrossRef] [PubMed]
- Delorme, E.J.; Alexander, P. Treatment of primary fibrosarcoma in the rat with immune lymphocytes. Lancet 1964, 2, 117–120. [Google Scholar] [CrossRef]
- Fefer, A. Immunotherapy and chemotherapy of Moloney sarcoma virus-induced tumors in mice. Cancer Res. 1969, 29, 2177–2183. [Google Scholar] [PubMed]
- Morgan, D.A.; Ruscetti, F.W.; Gallo, R. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science 1976, 193, 1007–1008. [Google Scholar] [CrossRef] [PubMed]
- Donohue, J.H.; Rosenstein, M.; Chang, A.E.; Lotze, M.T.; Robb, R.J.; Rosenberg, S.A. The systemic administration of purified interleukin 2 enhances the ability of sensitized murine lymphocytes to cure a disseminated syngeneic lymphoma. J. Immunol. 1984, 132, 2123–2128. [Google Scholar] [PubMed]
- Muul, L.M.; Spiess, P.J.; Director, E.P.; Rosenberg, S.A. Identification of specific cytolytic immune responses against autologous tumor in humans bearing malignant melanoma. J. Immunol. 1987, 138, 989–995. [Google Scholar] [PubMed]
- Rosenberg, S.A.; Spiess, P.; Lafreniere, R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 1986, 233, 1318–1321. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Packard, B.S.; Aebersold, P.M.; Solomon, D.; Topalian, S.L.; Toy, S.T.; Simon, P.; Lotze, M.T.; Yang, J.C.; Seipp, C.A. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N. Engl. J. Med. 1988, 319, 1676–1680. [Google Scholar] [CrossRef]
- Slavin, S.; Nagler, A.; Naparstek, E.; Kapelushnik, Y.; Aker, M.; Cividalli, G.; Varadi, G.; Kirschbaum, M.; Ackerstein, A.; Samuel, S.; et al. Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases. Blood 1998, 91, 756–763. [Google Scholar] [CrossRef] [Green Version]
- Dudley, M.E.; Wunderlich, J.R.; Yang, J.C. A phase I study of nonmyeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes in patients with metastatic melanoma. J. Immunother. 2002, 25, 243. [Google Scholar] [CrossRef] [Green Version]
- Dudley, M.E.; Wunderlich, J.R.; Robbins, P.F.; Yang, J.C.; Hwu, P.; Schwartzentruber, D.J.; Topalian, S.L.; Sherry, R.; Restifo, N.P.; Hubicki, A.M.; et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 2002, 298, 850–854. [Google Scholar] [CrossRef] [Green Version]
- Morgan, R.A.; Dudley, M.E.; Wunderlich, J.R.; Hughes, M.S.; Yang, J.C.; Sherry, R.M.; Royal, R.E.; Topalian, S.L.; Kammula, U.S.; Restifo, N.P.; et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006, 314, 126–129. [Google Scholar] [CrossRef] [Green Version]
- Johnson, L.A.; Morgan, R.A.; Dudley, M.E.; Cassard, L.; Yang, J.C.; Hughes, M.S.; Kammula, U.S.; Royal, R.E.; Sherry, R.M.; Wunderlich, J.R.; et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 2009, 114, 535–546. [Google Scholar] [CrossRef] [Green Version]
- Robbins, P.F.; Morgan, R.A.; Feldman, S.A.; Yang, J.C.; Sherry, R.M.; Dudley, M.E.; Wunderlich, J.R.; Nahvi, A.V.; Helman, L.J.; Mackall, C.L.; et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J. Clin. Oncol. 2011, 29, 917–924. [Google Scholar] [CrossRef]
- Pule, M.A.; Savoldo, B.; Myers, G.D.; Rossig, C.; Russell, H.V.; Dotti, G.; Huls, M.H.; Liu, E.; Gee, A.P.; Mei, Z.; et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: Persistence and antitumor activity in individuals with neuroblastoma. Nat. Med. 2008, 14, 1264–1270. [Google Scholar] [CrossRef]
- Parkhurst, M.R.; Yang, J.C.; Langan, R.C.; Dudley, M.E.; Nathan, D.-A.N.; Feldman, S.A.; Davis, J.L.; Morgan, R.A.; Merino, M.J.; Sherry, R.M.; et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol. Ther. 2011, 19, 620–626. [Google Scholar] [CrossRef]
- Robbins, P.F.; Lu, Y.-C.; El-Gamil, M.; Li, Y.F.; Gross, C.; Gartner, J.; Lin, J.C.; Teer, J.K.; Cliften, P.; Tycksen, E.; et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat. Med. 2013, 19, 747–752. [Google Scholar] [CrossRef]
- Tran, E.; Turcotte, S.; Gros, A.; Robbins, P.F.; Lu, Y.-C.; Dudley, M.E.; Wunderlich, J.R.; Somerville, R.P.; Hogan, K.; Hinrichs, C.S.; et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014, 344, 641–645. [Google Scholar] [CrossRef]
- Chandran, S.S.; Somerville, R.P.T.; Yang, J.C.; Sherry, R.M.; Klebanoff, C.A.; Goff, S.L.; Wunderlich, J.R.; Danforth, D.N.; Zlott, D.; Paria, B.C.; et al. Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: A single-centre, two-stage, single-arm, phase 2 study. Lancet Oncol. 2017, 18, 792–802. [Google Scholar] [CrossRef]
- Lo, W.; Parkhurst, M.; Robbins, P.F.; Tran, E.; Lu, Y.-C.; Jia, L.; Gartner, J.J.; Pasetto, A.; Deniger, D.; Malekzadeh, P.; et al. Immunologic Recognition of a Shared p53 Mutated Neoantigen in a Patient with Metastatic Colorectal Cancer. Cancer Immunol. Res. 2019, 7, 534–543. [Google Scholar] [CrossRef]
- Dudley, M.E.; Rosenberg, S.A. Adoptive cell transfer therapy. Semin. Oncol. 2007, 34, 524–531. [Google Scholar] [CrossRef] [Green Version]
- Hakomori, S. Aberrant glycosylation in tumors and tumor-associated carbohydrate antigens. Adv. Cancer Res. 1989, 52, 257–331. [Google Scholar]
- Werdelin, O.; Meldal, M.; Jensen, T. Processing of glycans on glycoprotein and glycopeptide antigens in antigen-presenting cells. Proc. Natl. Acad. Sci. USA 2002, 99, 9611–9613. [Google Scholar] [CrossRef] [Green Version]
- Garrido, F.; Aptsiauri, N.; Doorduijn, E.M.; Garcia Lora, A.M.; van Hall, T. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr. Opin. Immunol. 2016, 39, 44–51. [Google Scholar] [CrossRef] [Green Version]
- Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. USA 1989, 86, 10024–10028. [Google Scholar] [CrossRef] [Green Version]
- Stancovski, I.; Schindler, D.G.; Waks, T.; Yarden, Y.; Sela, M.; Eshhar, Z. Targeting of T lymphocytes to Neu/HER2-expressing cells using chimeric single chain Fv receptors. J. Immunol. 1993, 151, 6577–6582. [Google Scholar] [PubMed]
- Hwu, P.; Shafer, G.E.; Treisman, J.; Schindler, D.G.; Gross, G.; Cowherd, R.; Rosenberg, S.A.; Eshhar, Z. Lysis of ovarian cancer cells by human lymphocytes redirected with a chimeric gene composed of an antibody variable region and the Fc receptor gamma chain. J. Exp. Med. 1993, 178, 361–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krause, A.; Guo, H.F.; Latouche, J.B.; Tan, C.; Cheung, N.K.; Sadelain, M. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J. Exp. Med. 1998, 188, 619–626. [Google Scholar] [CrossRef] [Green Version]
- Finney, H.M.; Lawson, A.D.; Bebbington, C.R.; Weir, A.N. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J. Immunol. 1998, 161, 2791–2797. [Google Scholar] [PubMed]
- Haynes, N.M.; Trapani, J.A.; Teng, M.W.L.; Jackson, J.T.; Cerruti, L.; Jane, S.M.; Kershaw, M.H.; Smyth, M.J.; Darcy, P.K. Single-chain antigen recognition receptors that costimulate potent rejection of established experimental tumors. Blood 2002, 100, 3155–3163. [Google Scholar] [CrossRef] [Green Version]
- Lamers, C.H.J.; Sleijfer, S.; Vulto, A.G.; Kruit, W.H.J.; Kliffen, M.; Debets, R.; Gratama, J.W.; Stoter, G.; Oosterwijk, E. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: First clinical experience. J. Clin. Oncol. 2006, 24, e20-2. [Google Scholar] [CrossRef]
- Kershaw, M.H.; Westwood, J.A.; Parker, L.L.; Wang, G.; Eshhar, Z.; Mavroukakis, S.A.; White, D.E.; Wunderlich, J.R.; Canevari, S.; Rogers-Freezer, L.; et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin. Cancer Res. 2006, 12, 6106–6115. [Google Scholar] [CrossRef] [Green Version]
- Till, B.G.; Jensen, M.C.; Wang, J.; Chen, E.Y.; Wood, B.L.; Greisman, H.A.; Qian, X.; James, S.E.; Raubitschek, A.; Forman, S.J.; et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood 2008, 112, 2261–2271. [Google Scholar] [CrossRef] [Green Version]
- Hollyman, D.; Stefanski, J.; Przybylowski, M.; Bartido, S.; Borquez-Ojeda, O.; Taylor, C.; Yeh, R.; Capacio, V.; Olszewska, M.; Hosey, J.; et al. Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J. Immunother 2009, 32, 169–180. [Google Scholar] [CrossRef] [Green Version]
- Kochenderfer, J.N.; Wilson, W.H.; Janik, J.E.; Dudley, M.E.; Stetler-Stevenson, M.; Feldman, S.A.; Maric, I.; Raffeld, M.; Nathan, D.-A.N.; Lanier, B.J.; et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 2010, 116, 4099–4102. [Google Scholar] [CrossRef]
- Kalos, M.; Levine, B.L.; Porter, D.L.; Katz, S.; Grupp, S.A.; Bagg, A.; June, C.H. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 2011, 3, 95ra73. [Google Scholar] [CrossRef] [Green Version]
- Porter, D.L.; Levine, B.L.; Kalos, M.; Bagg, A.; June, C.H. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 2011, 365, 725–733. [Google Scholar] [CrossRef] [Green Version]
- Milone, M.C.; Fish, J.D.; Carpenito, C.; Carroll, R.G.; Binder, G.K.; Teachey, D.; Samanta, M.; Lakhal, M.; Gloss, B.; Danet-Desnoyers, G.; et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol. Ther. 2009, 17, 1453–1464. [Google Scholar] [CrossRef]
- Shimabukuro-Vornhagen, A.; Gödel, P.; Subklewe, M.; Stemmler, H.J.; Schlößer, H.A.; Schlaak, M.; Kochanek, M.; Böll, B.; von Bergwelt-Baildon, M.S. Cytokine release syndrome. J. Immunother. Cancer 2018, 6, 56. [Google Scholar] [CrossRef] [Green Version]
- Kochenderfer, J.N.; Dudley, M.E.; Feldman, S.A.; Wilson, W.H.; Spaner, D.E.; Maric, I.; Stetler-Stevenson, M.; Phan, G.Q.; Hughes, M.S.; Sherry, R.M.; et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 2012, 119, 2709–2720. [Google Scholar] [CrossRef]
- Kochenderfer, J.N.; Dudley, M.E.; Kassim, S.H.; Somerville, R.P.T.; Carpenter, R.O.; Stetler-Stevenson, M.; Yang, J.C.; Phan, G.Q.; Hughes, M.S.; Sherry, R.M.; et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J. Clin. Oncol. 2015, 33, 540–549. [Google Scholar] [CrossRef] [Green Version]
- Maus, M.V.; Grupp, S.A.; Porter, D.L.; June, C.H. Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood 2014, 123, 2625–2635. [Google Scholar] [CrossRef]
- Grupp, S.A.; Kalos, M.; Barrett, D.; Aplenc, R.; Porter, D.L.; Rheingold, S.R.; Teachey, D.T.; Chew, A.; Hauck, B.; Wright, J.F.; et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 2013, 368, 1509–1518. [Google Scholar] [CrossRef] [Green Version]
- Lee, D.W.; Gardner, R.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.; Grupp, S.A.; Mackall, C.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014, 124, 188–195. [Google Scholar] [CrossRef] [Green Version]
- Yeku, O.; Li, X.; Brentjens, R.J. Adoptive T-Cell Therapy for Solid Tumors. Am. Soc. Clin. Oncol. Educ. Book 2017, 37, 193–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baybutt, T.R.; Flickinger, J.C.; Caparosa, E.M.; Snook, A.E. Advances in Chimeric Antigen Receptor T-Cell Therapies for Solid Tumors. Clin. Pharmacol. Ther. 2019, 105, 71–78. [Google Scholar] [CrossRef] [PubMed]
- Mukherjee, S. Genomics-Guided Immunotherapy for Precision Medicine in Cancer. Cancer Biother. Radiopharm. 2019. [Google Scholar] [CrossRef]
Antigen Target | Cancer Type | Phase | ClinicalTrials.gov Designation |
---|---|---|---|
CD117 | Osteoid Sarcoma, Ewing Sarcoma | I/II | NCT03356782 |
CD133 | Liver, Pancreatic, Brain, Breas, Ovarian, Colorectal, Acute Myeloid and Lymphoid Leukemias | I/II | NCT02541370 |
Osteoid Sarcoma, Ewing Sarcoma | I/II | NCT03356782 | |
CD171 | Neuroblastoma, Ganglioneuroblastoma, | I | NCT02311621 |
CEA | Colorectal | I/II | NCT02959151 |
Lung, Colorectal, Gastric, Breast, Pancreatic | I | NCT02349724 | |
EGFR | Colorectal | I/II | NCT03152435 |
EGFRvIII | Glioma, Glioblastoma, Gliosarcoma | I/II | NCT01454596 |
EpCAM | Colon, Esophageal, Pancreatic, Prostate, Gastric, Hepatic | I/II | NCT03013712 |
EphA2 | Glioma | I/II | NCT02575261 |
ErbB | Head and Neck | I/II | NCT01818323 |
FRα | Urothelial Bladder | I/II | NCT03185468 |
GD2 | Glioma | I/II | NCT03252171 |
Neuroblastoma | I/II | NCT03373097 | |
I | NCT01822652 | ||
II | NCT02765243 | ||
Cervical | I/II | NCT03356795 | |
Osteoid Sarcoma, Ewing Sarcoma | I/II | NCT03356782 | |
Sarcoma, Osteosarcoma, Neuroblastoma, Melanoma | I | NCT02107963 | |
GPC3 | Hepatocellular Carcinoma | I/II | NCT02723942 |
NCT02959151 | |||
NCT03084380 | |||
HER2 | Breast | I/II | NCT02547961 |
Sarcoma | I | NCT00902044 | |
IL-13Rα2 | Glioma, Glioblastoma | I | NCT02208362 |
Mesothelin | Pancreatic | I/II | NCT02959151 |
Cervical | I/II | NCT03356795 | |
Advanced Solid Tumors | I/II | NCT03615313 | |
Pancreatic, Ovarian, Mesothelioma | I | NCT02159716 | |
Malignant Pleural Disease, Mesothelioma, Lung, Breast | I | NCT02414269 | |
MUC1 | Cervical | I/II | NCT03356795 |
Esophageal | I/II | NCT03706326 | |
Non-Small Cell Lung | I/II | NCT03525782 | |
Osteoid Sarcoma, Ewing Sarcoma | I/II | NCT03356782 | |
Intrahepatic Cholangiocarcinoma | I/II | NCT03633773 | |
MUC-16 | Ovarian | I | NCT02498912 |
PSMA | Urothelial Bladder | I/II | NCT03185468 |
Cervical | I/II | NCT03356795 | |
Prostate | I | NCT01140373 |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Carlson, R.D.; Flickinger, J.C., Jr.; Snook, A.E. Talkin’ Toxins: From Coley’s to Modern Cancer Immunotherapy. Toxins 2020, 12, 241. https://doi.org/10.3390/toxins12040241
Carlson RD, Flickinger JC Jr., Snook AE. Talkin’ Toxins: From Coley’s to Modern Cancer Immunotherapy. Toxins. 2020; 12(4):241. https://doi.org/10.3390/toxins12040241
Chicago/Turabian StyleCarlson, Robert D., John C. Flickinger, Jr., and Adam E. Snook. 2020. "Talkin’ Toxins: From Coley’s to Modern Cancer Immunotherapy" Toxins 12, no. 4: 241. https://doi.org/10.3390/toxins12040241
APA StyleCarlson, R. D., Flickinger, J. C., Jr., & Snook, A. E. (2020). Talkin’ Toxins: From Coley’s to Modern Cancer Immunotherapy. Toxins, 12(4), 241. https://doi.org/10.3390/toxins12040241