Assessing the Future of Solid Tumor Immunotherapy
Abstract
:1. Introduction
2. Standard of Care Therapies
3. Basics of Immunotherapy
4. Solid Tumor Immunotherapy Barrier Overview
4.1. Tumor Microenvironment (TME) General Features and Soluble Mediators
4.2. T Cell Exhaustion in the TME
4.3. Novel Delivery Technologies
5. Immunotherapy Types for Solid Tumors
5.1. Checkpoint Inhibitors
5.2. Bi-Specific Antibodies
5.3. Oncolytic Viruses (OV)
5.4. Cancer Vaccines
5.4.1. Preventative vs. Therapeutic Vaccines
5.4.2. Clinical Trials
5.5. Adoptive Cell Therapy (ACT)
5.5.1. Autologous Cell Therapy
5.5.2. Allogeneic Cell Therapy
5.5.3. Combination Therapy or Other Strategies of Regional Delivery
5.5.4. Side Effects
6. Summary
7. Conclusions and Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
ACT | adoptive cell therapy |
ADCs | antibody-drug conjugated |
AP-1 | activator protein 1 |
bFGF | basic fibroblast growth factor |
BsAb | bispecific monoclonal antibodies |
BCMA | B-cell maturation antigen |
BsAb | bispecific antibody |
BiTEs | bispecific T cell engagers |
BTLA | band T lymphocyte attenuator |
CAR-NK | chimeric antigen receptor NK cells |
CAR-T | chimeric antigen receptor T cells |
CRS | cytokine release syndrome |
CTLA-4 | cytotoxic T lymphocyte antigen 4 |
FDA | food and drug administration |
GvHD | graft-versus-host disease |
HBV | hepatitis B virus |
HPV | human papillomavirus |
HLA | human leukocyte antigen |
iCasp9 | inducible Caspase 9 |
ICI | immune checkpoint inhibitors |
IFN | interferon |
IL | interleukin |
iPSC | induced pluripotent stem cells |
LAG-3 | lymphocyte activation gene 3 protein |
LPD | lipid-protamine-DNA |
mAb | monoclonal antibody |
MDSCs | myeloid derived suppressor cells |
NF-κB | nuclear factor kappa B |
mRNA | messenger RNA |
OTOTT | on-target-off-tumor-toxicity |
OV | oncolytic virus |
PD-1 | programmed cell death protein |
PD-L1 | programmed cell death protein and its ligand |
scFv | single chain variable fragment |
STAT3 | signal transducer and activator of transcription 3 |
TAM | tumor associated macrophages |
TGF-β | transforming growth factor beta |
TIL’s | tumor-infiltrating lymphocytes |
TIM-3 | T-cell immunoglobulin domain and mucin domain protein 3 |
TME | tumor microenvironment |
Tregs | regulatory T cells |
TIGIT | T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain |
TNF | tumor necrosis factor |
TRAIL | TNF related apoptosis inducing ligand |
VEGF | vascular endothelial growth factor |
References
- Kucerova, P.; Cervinkova, M. Spontaneous regression of tumour and the role of microbial infection—Possibilities for cancer treatment. Anti-Cancer Drugs 2016, 27, 269–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dobosz, P.; Dzieciątkowski, T. The Intriguing History of Cancer Immunotherapy. Front. Immunol. 2019, 10, 2965. [Google Scholar] [CrossRef] [Green Version]
- Mellman, I.; Coukos, G.; Dranoff, G. Cancer immunotherapy comes of age. Nature 2011, 480, 480–489. [Google Scholar] [CrossRef] [PubMed]
- Burga, R.A.; Thorn, M.; Point, G.R.; Guha, P.; Nguyen, C.T.; Licata, L.A.; DeMatteo, R.P.; Ayala, A.; Espat, N.J.; Junghans, R.P.; et al. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol. Immunother. 2015, 64, 817–829. [Google Scholar] [CrossRef] [Green Version]
- Swanson, G.P.; Rynearson, K.; Symmonds, R. Significance of margins of excision on breast cancer recurrence. Am. J. Clin. Oncol. 2002, 25, 438–441. [Google Scholar] [CrossRef]
- DeVita, V.T., Jr.; Chu, E. A History of Cancer Chemotherapy. Cancer Res. 2008, 68, 8643–8653. [Google Scholar] [CrossRef] [Green Version]
- De Angelis, C. Side Effects Related to Systemic Cancer Treatment: Are We Changing the Promethean Experience with Molecularly Targeted Therapies? Curr. Oncol. 2008, 15, 198–199. [Google Scholar] [CrossRef] [PubMed]
- Baskar, R.; Lee, K.A.; Yeo, R.; Yeoh, K.-W. Cancer and Radiation Therapy: Current Advances and Future Directions. Int. J. Med. Sci. 2012, 9, 193–199. [Google Scholar] [CrossRef] [Green Version]
- Cheng, H.; Wang, Z.; Fu, L.; Xu, T. Macrophage Polarization in the Development and Progression of Ovarian Cancers: An Overview. Front. Oncol. 2019, 9, 421. [Google Scholar] [CrossRef] [Green Version]
- Jiang, T.; Zhou, C. The past, present and future of immunotherapy against tumor. Transl. Lung Cancer Res. 2015, 4, 253–264. [Google Scholar] [CrossRef]
- Ventola, C.L. Cancer Immunotherapy, Part 1: Current Strategies and Agents. Pharm. Ther. 2017, 42, 375–383. [Google Scholar]
- Filin, I.Y.; Solovyeva, V.V.; Kitaeva, K.V.; Rutland, C.S.; Rizvanov, A.A. Current Trends in Cancer Immunotherapy. Biomedicines 2020, 8, 621. [Google Scholar] [CrossRef] [PubMed]
- Maeda, H.; Khatami, M. Analyses of repeated failures in cancer therapy for solid tumors: Poor tumor-selective drug delivery, low therapeutic efficacy and unsustainable costs. Clin. Transl. Med. 2018, 7, 11. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Goding Sauer, A.; Fedewa, S.A.; Butterly, L.F.; Anderson, J.C.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
- Hou, B.; Tang, Y.; Li, W.; Zeng, Q.; Chang, D. Efficiency of CAR-T Therapy for Treatment of Solid Tumor in Clinical Trials: A Meta-Analysis. Dis. Markers 2019, 2019, 3425291. [Google Scholar] [CrossRef] [Green Version]
- Duffy, A.G.; Greten, T.F. Immunological off-target effects of standard treatments in gastrointestinal cancers. Ann. Oncol. 2013, 25, 24–32. [Google Scholar] [CrossRef]
- Giampieri, R.; Del Prete, M.; Prochilo, T.; Puzzoni, M.; Pusceddu, V.; Pani, F.; Maccaroni, E.; Mascia, R.; Baleani, M.G.; Meletani, T.; et al. Off-target effects and clinical outcome in metastatic colorectal cancer patients receiving regorafenib: The TRIBUTE analysis. Sci. Rep. 2017, 7, 45703. [Google Scholar] [CrossRef] [Green Version]
- Brown, J.M.; Giaccia, A.J. The unique physiology of solid tumors: Opportunities (and problems) for cancer therapy. Cancer Res. 1998, 58, 1408–1416. [Google Scholar]
- Khawar, I.A.; Kim, J.H.; Kuh, H.-J. Improving drug delivery to solid tumors: Priming the tumor microenvironment. J. Control Release 2015, 201, 78–89. [Google Scholar] [CrossRef]
- Vaupel, P.; Thews, O.; Hoeckel, M. Treatment resistance of solid tumors: Role of hypoxia and anemia. Med. Oncol. 2001, 18, 243–259. [Google Scholar] [CrossRef]
- Trédan, O.; Galmarini, C.M.; Patel, K.; Tannock, I.F. Drug Resistance and the Solid Tumor Microenvironment. J. Natl. Cancer Inst. 2007, 99, 1441–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chai, L.F.; Prince, E.; Pillarisetty, V.G.; Katz, S.C. Challenges in assessing solid tumor responses to immunotherapy. Cancer Gene Ther. 2019, 27, 528–538. [Google Scholar] [CrossRef] [PubMed]
- Whiteside, T.L. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008, 27, 5904–5912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poggi, A.; Musso, A.; Dapino, I.; Zocchi, M.R. Mechanisms of tumor escape from immune system: Role of mesenchymal stromal cells. Immunol. Lett. 2014, 159, 55–72. [Google Scholar] [CrossRef] [PubMed]
- Lindau, D.; Gielen, P.; Kroesen, M.; Wesseling, P.; Adema, G.J. The immunosuppressive tumour network: Myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 2012, 138, 105–115. [Google Scholar] [CrossRef] [PubMed]
- Guha, P.; Gardell, J.; Rabinowitz, B.; Lopes, M.; DaSilva, N.A.; Rowley, D.; Katz, S.C. Monocytic and granulocytic myeloid-derived suppressor cell plasticity and differentiation are organ-specific. Oncogene 2020, 40, 693–704. [Google Scholar] [CrossRef]
- Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, H.; Pardoll, D.; Jove, R. STATs in cancer inflammation and immunity: A leading role for STAT3. Nat. Cancer 2009, 9, 798–809. [Google Scholar] [CrossRef]
- Grivennikov, S.; Karin, E.; Terzic, J.; Mucida, D.; Yu, G.Y.; Vallabhapurapu, S.; Scheller, J.; Rose-John, S.; Cheroutre, H.; Eckmann, L.; et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell 2009, 15, 103–113. [Google Scholar] [CrossRef] [Green Version]
- Becker, C.; Fantini, M.C.; Schramm, C.; Lehr, H.A.; Wirtz, S.; Becker, A.; Burg, J.; Strand, S.; Kiesslich, R.; Huber, S.; et al. TGF-beta suppresses tumor progression in colon cancer by inhibition of IL–6 trans-signaling. Immunity 2004, 42, P139. [Google Scholar] [CrossRef]
- Bollrath, J.; Phesse, T.; von Burstin, V.A.; Putoczki, T.; Bennecke, M.; Bateman, T.; Nebelsiek, T.; Lundgren-May, T.; Canli, Ö.; Schwitalla, S.; et al. gp130-Mediated Stat3 Activation in Enterocytes Regulates Cell Survival and Cell-Cycle Progression during Colitis-Associated Tumorigenesis. Cancer Cell 2009, 15, 91–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Popivanova, B.K.; Kitamura, K.; Wu, Y.; Kondo, T.; Kagaya, T.; Kaneko, S.; Oshima, M.; Fujii, C.; Mukaida, N. Blocking TNF-α in mice reduces colorectal carcinogenesis associated with chronic colitis. J. Clin. Investig. 2008, 118, 560–570. [Google Scholar] [CrossRef] [PubMed]
- Wang, D.J.; Ratnam, N.M.; Byrd, J.C.; Guttridge, D.C. NF-kappaB functions in tumor initiation by suppressing the surveillance of both innate and adaptive immune cells. Cell Rep. 2014, 9, 90–103. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Shen, Y.; Wang, S.; Shen, Q.; Zhou, X. The role of STAT3 in leading the crosstalk between human cancers and the immune system. Cancer Lett. 2018, 415, 117–128. [Google Scholar] [CrossRef] [PubMed]
- Van Horssen, R.; Hagen, T.L.M.T.; Eggermont, A.M.M. TNF-α in Cancer Treatment: Molecular Insights, Antitumor Effects, and Clinical Utility. Oncologist 2006, 11, 397–408. [Google Scholar] [CrossRef]
- Zhao, L.; Ching, L.-M.; Kestell, P.; Baguley, B.C. The antitumour activity of 5,6-dimethylxanthenone-4-acetic acid (DMXAA) in TNF receptor-1 knockout mice. Br. J. Cancer 2002, 87, 465–470. [Google Scholar] [CrossRef] [Green Version]
- Bertrand, F.; Montfort, A.; Marcheteau, E.; Imbert, C.; Gilhodes, J.; Filleron, T.; Rochaix, P.; Andrieu-Abadie, N.; Levade, T.; Meyer, N.; et al. TNFalpha blockade overcomes resistance to anti-PD-1 in experimental melanoma. Nat. Commun. 2017, 8, 2256. [Google Scholar] [CrossRef] [Green Version]
- Massagué, J. TGFβ in cancer. Cell 2008, 134, 215–230. [Google Scholar] [CrossRef] [Green Version]
- Berraondo, P.; Sanmamed, M.F.; Ochoa, M.C.; Etxeberria, I.; Aznar, M.A.; Pérez-Gracia, J.L.; Rodriguez-Ruiz, M.E.; Ponz-Sarvise, M.; Castañón, E.; Melero, I. Cytokines in clinical cancer immunotherapy. Br. J. Cancer 2019, 120, 6–15. [Google Scholar] [CrossRef] [Green Version]
- Jiang, Y.; Li, Y.; Zhu, B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 2015, 6, e1792. [Google Scholar] [CrossRef] [Green Version]
- Zarour, H.M. Reversing T-cell Dysfunction and Exhaustion in Cancer. Clin. Cancer Res. 2016, 22, 1856–1864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ribas, A.; Medina, T.; Kummar, S.; Amin, A.; Kalbasi, A.; Drabick, J.J.; Barve, M.; Daniels, G.A.; Wong, D.J.; Schmidt, E.V.; et al. SD-101 in Combination with Pembrolizumab in Advanced Melanoma: Results of a Phase Ib, Multicenter Study. Cancer Discov. 2018, 8, 1250–1257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, W.; Shen, L.; Wang, Y.; Liu, Q.; Goodwin, T.J.; Li, J.; Dorosheva, O.; Liu, T.; Liu, R.; Huang, L. Synergistic and low adverse effect cancer immunotherapy by immunogenic chemotherapy and locally expressed PD-L1 trap. Nat. Commun. 2018, 9, 2237. [Google Scholar] [CrossRef] [PubMed]
- Wong, C.; Stylianopoulos, T.; Cui, J.; Martin, J.; Chauhan, V.P.; Jiang, W.; Popović, Z.; Jain, R.K.; Bawendi, M.G.; Fukumura, D. Multistage nanoparticle delivery system for deep penetration into tumor tissue. Proc. Natl. Acad. Sci. USA 2011, 108, 2426–2431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Katz, S.C.; Hardaway, J.; Prince, E.; Guha, P.; Cunetta, M.; Moody, A.; Wang, L.J.; Armenio, V.; Espat, N.J.; Junghans, R.P. HITM-SIR: Phase Ib trial of intraarterial chimeric antigen receptor T-cell therapy and selective internal radiation therapy for CEA+ liver metastases. Cancer Gene Ther. 2019, 27, 341–355. [Google Scholar] [CrossRef] [PubMed]
- Katz, S.C.; Moody, A.E.; Guha, P.; Hardaway, J.C.; Prince, E.; Laporte, J.; Stancu, M.; Slansky, J.E.; Jordan, K.R.; Schulick, R.D.; et al. HITM-SURE: Hepatic immunotherapy for metastases phase Ib anti-CEA CAR-T study utilizing pressure enabled drug delivery. J. Immunother. Cancer 2020, 8, e001097. [Google Scholar] [CrossRef]
- Narayanan, J.S.S.; Vicente, D.A.; Ray, P.; Chai, L.F.; Erdem, S.; Carr, M.J.; Capacio, B.A.; Cox, B.F.; Jaroch, D.B.; Katz, S.C.; et al. Pressure-enabled delivery of gemcitabine in an orthotopic pancreatic cancer mouse model. Surgery 2020, 168, 448–456. [Google Scholar] [CrossRef]
- Labani-Motlagh, A.; Ashja-Mahdavi, M.; Loskog, A. The Tumor Microenvironment: A Milieu Hindering and Obstructing Antitumor Immune Responses. Front. Immunol. 2020, 11, 940. [Google Scholar] [CrossRef]
- Pento, J.T. Monoclonal Antibodies for the Treatment of Cancer. Anticancer Res. 2017, 37, 5935–5939. [Google Scholar] [CrossRef] [Green Version]
- Graziani, G.; Tentori, L.; Navarra, P. Ipilimumab: A novel immunostimulatory monoclonal antibody for the treatment of cancer. Pharmacol. Res. 2012, 65, 9–22. [Google Scholar] [CrossRef]
- Lipson, E.J.; Drake, C.G. Ipilimumab: An Anti-CTLA-4 Antibody for Metastatic Melanoma. Clin. Cancer Res. 2011, 17, 6958–6962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robert, C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat. Commun. 2020, 11, 3801. [Google Scholar] [CrossRef] [PubMed]
- Andrews, L.P.; Yano, H.; Vignali, D.A.A. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: Breakthroughs or backups. Nat. Immunol. 2019, 20, 1425–1434. [Google Scholar] [CrossRef]
- Quezada, S.; Peggs, K.S. Exploiting CTLA-4, PD-1 and PD-L1 to reactivate the host immune response against cancer. Br. J. Cancer 2013, 108, 1560–1565. [Google Scholar] [CrossRef] [PubMed]
- Meyers, D.E.; Bryan, P.M.; Banerji, S.; Morris, D.G. Targeting the PD-1/PD-L1 axis for the treatment of non-small-cell lung cancer. Curr. Oncol. 2018, 25, e324–e334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thorn, M.; Guha, P.; Cunetta, M.; Espat, N.J.; Miller, G.; Junghans, R.P.; Katz, S.C. Tumor-associated GM-CSF overexpression induces immunoinhibitory molecules via STAT3 in myeloid-suppressor cells infiltrating liver metastases. Cancer Gene Ther. 2016, 23, 188–198. [Google Scholar] [CrossRef] [PubMed]
- Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rotte, A. Combination of CTLA-4 and PD-1 blockers for treatment of cancer. J. Exp. Clin. Cancer Res. 2019, 38, 255. [Google Scholar] [CrossRef]
- Postow, M.A.; Chesney, J.; Pavlick, A.C.; Robert, C.; Grossmann, K.; McDermott, D.; Linette, G.P.; Meyer, N.; Giguere, J.K.; Agarwala, S.S.; et al. Nivolumab and Ipilimumab versus Ipilimumab in Untreated Melanoma. N. Engl. J. Med. 2015, 372, 2006–2017. [Google Scholar] [CrossRef] [Green Version]
- Hodi, F.S.; Chesney, J.; Pavlick, A.C.; Robert, C.; Grossmann, K.F.; McDermott, D.F.; Linette, G.P.; Meyer, N.; Giguere, J.K.; Agarwala, S.S.; et al. Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial. Lancet Oncol. 2016, 17, 1558–1568. [Google Scholar] [CrossRef] [Green Version]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 1270–1271. [Google Scholar] [CrossRef] [Green Version]
- Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
- Hodi, F.S.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1480–1492. [Google Scholar] [CrossRef]
- Johnson, D.B.; Chandra, S.; Sosman, J.A. Immune Checkpoint Inhibitor Toxicity in 2018. JAMA 2018, 320, 1702–1703. [Google Scholar] [CrossRef] [PubMed]
- Duffy, M.J.; Crown, J. Biomarkers for Predicting Response to Immunotherapy with Immune Checkpoint Inhibitors in Cancer Patients. Clin. Chem. 2019, 65, 1228–1238. [Google Scholar] [CrossRef] [PubMed]
- Uboha, N.V.; Milhem, M.M.; Kovacs, C.; Amin, A.; Magley, A.; Das Purkayastha, D.; Piha-Paul, S.A. Phase II study of spartalizumab (PDR001) and LAG525 in advanced solid tumors and hematologic malignancies. J. Clin. Oncol. 2019, 37, 2553. [Google Scholar] [CrossRef]
- Lakhani, N.; Spreafico, A.; Tolcher, A.; Rodon, J.; Janku, F.; Chandana, S.; Oliva, M.; Sharma, M.; Abdul-Karim, R.; Hansen, U.; et al. 1019O Phase I studies of Sym021, an anti-PD-1 antibody, alone and in combination with Sym022 (anti-LAG-3) or Sym023 (anti-TIM-3). Ann. Oncol. 2020, 31, S704. [Google Scholar] [CrossRef]
- Labrijn, A.F.; Janmaat, M.L.; Reichert, J.M.; Parren, P.W.H.I. Bispecific antibodies: A mechanistic review of the pipeline. Nat. Rev. Drug Discov. 2019, 18, 585–608. [Google Scholar] [CrossRef] [PubMed]
- Brischwein, K.; Parr, L.; Pflanz, S.; Volkland, J.; Lumsden, J.; Klinger, M.; Locher, M.; Hammond, S.A.; Kiener, P.; Kufer, P.; et al. Strictly Target Cell-dependent Activation of T Cells by Bispecific Single-chain Antibody Constructs of the BiTE Class. J. Immunother. 2007, 30, 798–807. [Google Scholar] [CrossRef]
- Brischwein, K.; Schlereth, B.; Guller, B.; Steiger, C.; Wolf, A.; Lutterbuese, R.; Offner, S.; Locher, M.; Urbig, T.; Raum, T.; et al. MT110: A novel bispecific single-chain antibody construct with high efficacy in eradicating established tumors. Mol. Immunol. 2006, 43, 1129–1143. [Google Scholar] [CrossRef]
- Lutterbuese, R.; Raum, T.; Kischel, R.; Hoffmann, P.; Mangold, S.; Rattel, B.; Friedrich, M.; Thomas, O.; Lorenczewski, G.; Rau, D.; et al. T cell-engaging BiTE antibodies specific for EGFR potently eliminate KRAS- and BRAF-mutated colorectal cancer cells. Proc. Natl. Acad. Sci. USA 2010, 107, 12605–12610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Torisu-Itakura, H.; Schoellhammer, H.F.; Sim, M.-S.; Irie, R.F.; Hausmann, S.; Raum, T.; Baeuerle, P.A.; Morton, D.L. Redirected Lysis of Human Melanoma Cells by a MCSP/CD3-bispecific BiTE Antibody That Engages Patient-derived T Cells. J. Immunother. 2011, 34, 597–605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klinger, M.; Brandl, C.; Zugmaier, G.; Hijazi, Y.; Bargou, R.C.; Topp, M.S.; Gökbuget, N.; Neumann, S.; Goebeler, M.; Viardot, A.; et al. Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood 2012, 119, 6226–6233. [Google Scholar] [CrossRef] [PubMed]
- Goebeler, M.-E.; Knop, S.; Viardot, A.; Kufer, P.; Topp, M.S.; Einsele, H.; Noppeney, R.; Hess, G.; Kallert, S.; Mackensen, A.; et al. Bispecific T-Cell Engager (BiTE) Antibody Construct Blinatumomab for the Treatment of Patients with Relapsed/Refractory Non-Hodgkin Lymphoma: Final Results From a Phase I Study. J. Clin. Oncol. 2016, 34, 1104–1111. [Google Scholar] [CrossRef] [PubMed]
- Donnelly, O.; Harrington, K.; Melcher, A.; Pandha, H. Live viruses to treat cancer. J. R. Soc. Med. 2013, 106, 310–314. [Google Scholar] [CrossRef] [Green Version]
- Kelly, E.; Russell, S.J. History of Oncolytic Viruses: Genesis to Genetic Engineering. Mol. Ther. 2007, 15, 651–659. [Google Scholar] [CrossRef]
- Zou, Y.; Luo, Y.; Zhang, J.; Xia, N.; Tan, G.; Huang, C. Bibliometric analysis of oncolytic virus research, 2000 to 2018. Medicine 2019, 98, e16817. [Google Scholar] [CrossRef]
- Fukuhara, H.; Ino, Y.; Todo, T. Oncolytic virus therapy: A new era of cancer treatment at dawn. Off. J. Jpn. Cancer Assoc. 2016, 107, 1373–1379. [Google Scholar] [CrossRef]
- Bommareddy, P.K.; Patel, A.; Hossain, S.; Kaufman, H.L. Talimogene Laherparepvec (T-VEC) and Other Oncolytic Viruses for the Treatment of Melanoma. Am. J. Clin. Dermatol. 2016, 18, 1–15. [Google Scholar] [CrossRef]
- Pylayeva-Gupta, Y.; Lee, K.E.; Hajdu, C.H.; Miller, G.; Bar-Sagi, D. Oncogenic Kras-Induced GM-CSF Production Promotes the Development of Pancreatic Neoplasia. Cancer Cell 2012, 21, 836–847. [Google Scholar] [CrossRef] [Green Version]
- Jou, J.; Harrington, K.J.; Zocca, M.-B.; Ehrnrooth, E.; Cohen, E.E. The Changing Landscape of Therapeutic Cancer Vaccines—Novel Platforms and Neoantigen Identification. Clin. Cancer Res. 2020, 27, 689–703. [Google Scholar] [CrossRef] [PubMed]
- Lollini, P.L.; Cavallo, F.; Nanni, P.; Forni, G. Vaccines for tumour prevention. Nat. Cancer 2006, 6, 204–216. [Google Scholar] [CrossRef]
- Morse, M.A.; Gwin, W.R., III; Mitchell, D.A. Vaccine Therapies for Cancer: Then and Now. Target. Oncol. 2021, 16, 121–152. [Google Scholar] [CrossRef]
- DeMaria, P.J.; Bilusic, M. Cancer Vaccines. Hematol. Oncol. Clin. N. Am. 2019, 33, 199–214. [Google Scholar] [CrossRef]
- Lamm, D.L.; Blumenstein, B.A.; Crawford, E.D.; Montie, J.E.; Scardino, P.; Grossman, H.B.; Stanisic, T.H.; Smith, J.A.; Sullivan, J.; Sarosdy, M.F.; et al. A Randomized Trial of Intravesical Doxorubicin and Immunotherapy with Bacille Calmette–Guérin for Transitional-Cell Carcinoma of the Bladder. N. Engl. J. Med. 1991, 325, 1205–1209. [Google Scholar] [CrossRef]
- Laheru, D.; Lutz, E.; Burke, J.; Biedrzycki, B.; Solt, S.; Onners, B.; Tartakovsky, I.; Nemunaitis, J.; Le, D.; Sugar, E.; 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, 1455–1463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lipson, E.J.; Sharfman, W.H.; Chen, S.; McMiller, T.L.; Pritchard, T.S.; Salas, J.T.; 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, 214. [Google Scholar] [CrossRef] [Green Version]
- Salgia, R.; Lynch, T.; Skarin, A.; Lucca, J.; Lynch, C.; Jung, K.; Hodi, F.S.; 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, 624–630. [Google Scholar] [CrossRef] [PubMed]
- Small, E.J.; Sacks, N.; Nemunaitis, J.; Urba, W.J.; Dula, E.; Centeno, A.S.; Nelson, W.G.; Ando, D.; Howard, C.; Borellini, F.; et al. Granulocyte Macrophage Colony-Stimulating Factor–Secreting Allogeneic Cellular Immunotherapy for Hormone-Refractory Prostate Cancer. Clin. Cancer Res. 2007, 13, 3883–3891. [Google Scholar] [CrossRef] [Green Version]
- Rosenberg, S.A.; Restifo, N.P.; Yang, J.C.; Morgan, R.A.; Dudley, M.E. Adoptive cell transfer: A clinical path to effective cancer immunotherapy. Nat. Cancer 2008, 8, 299–308. [Google Scholar] [CrossRef]
- Dudley, M.E.; Rosenberg, S.A. Adoptive-cell-transfer therapy for the treatment of patients with cancer. Nat. Rev. Cancer 2003, 3, 666–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Itzhaki, O.; Hovav, E.; Ziporen, Y.; Levy, D.; Kubi, A.; Zikich, D.; Hershkovitz, L.; Treves, A.J.; Shalmon, B.; Zippel, D.; et al. Establishment and Large-scale Expansion of Minimally cultured “Young” Tumor Infiltrating Lymphocytes for Adoptive Transfer Therapy. J. Immunother. 2011, 34, 212–220. [Google Scholar] [CrossRef]
- Muranski, P.; Boni, A.; Wrzesinski, C.; Citrin, D.; Rosenberg, S.A.; Childs, R.W.; Restifo, N.P. Increased intensity lymphodepletion and adoptive immunotherapy—How far can we go? Nat. Clin. Pract. Oncol. 2006, 3, 668–681. [Google Scholar] [CrossRef] [Green Version]
- 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.; et al. 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] [PubMed]
- Besser, M.J.; Shapira-Frommer, R.; Itzhaki, O.; Treves, A.J.; Zippel, D.B.; Levy, D.; Kubi, A.; Shoshani, N.; Zikich, D.; Ohayon, Y.; et al. Adoptive Transfer of Tumor-Infiltrating Lymphocytes in Patients with Metastatic Melanoma: Intent-to-Treat Analysis and Efficacy after Failure to Prior Immunotherapies. Clin. Cancer Res. 2013, 19, 4792–4800. [Google Scholar] [CrossRef] [PubMed] [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] [PubMed] [Green Version]
- Hartmann, J.; Schüßler-Lenz, M.; Bondanza, A.; Buchholz, C.J. Clinical development of CAR T cells—Challenges and opportunities in translating innovative treatment concepts. EMBO Mol. Med. 2017, 9, 1183–1197. [Google Scholar] [CrossRef]
- Chmielewski, M.; Abken, H. TRUCKs: The fourth generation of CARs. Exp. Opin. Biol. Ther. 2015, 15, 1145–1154. [Google Scholar] [CrossRef]
- Gargett, T.; Brown, M.P. 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. [Google Scholar] [CrossRef]
- Lorenzo-Herrero, S.; López-Soto, A.; Sordo-Bahamonde, C.; Gonzalez-Rodriguez, S.; Vitale, M.; Gonzalez, S. NK Cell-Based Immunotherapy in Cancer Metastasis. Cancers 2018, 11, 29. [Google Scholar] [CrossRef] [Green Version]
- Hermanson, D.L.; Kaufman, D.S. Utilizing Chimeric Antigen Receptors to Direct Natural Killer Cell Activity. Front. Immunol. 2015, 6, 195. [Google Scholar] [CrossRef] [Green Version]
- Miller, J.S.; Soignier, Y.; Panoskaltsis-Mortari, A.; McNearney, S.A.; Yun, G.H.; Fautsch, S.K.; McKenna, D.; Le, C.; DeFor, T.E.; Burns, L.J.; et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005, 105, 3051–3057. [Google Scholar] [CrossRef] [Green Version]
- Geller, M.A.; Cooley, S.; Judson, P.L.; Ghebre, R.; Carson, L.F.; Argenta, P.A.; Jonson, A.L.; Panoskaltsis-Mortari, A.; Curtsinger, J.; McKenna, D.; et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy 2011, 13, 98–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Veluchamy, J.P.; Kok, N.; van der Vliet, H.J.; Verheul, H.M.W.; De Gruijl, T.D.; Spanholtz, J. The Rise of Allogeneic Natural Killer Cells as a Platform for Cancer Immunotherapy: Recent Innovations and Future Developments. Front. Immunol. 2017, 8, 631. [Google Scholar] [CrossRef] [PubMed]
- Cichocki, F.; Bjordahl, R.; Gaidarova, S.; Mahmood, S.; Abujarour, R.; Wang, H.; Tuininga, K.; Felices, M.; Davis, Z.B.; Bendzick, L.; et al. iPSC-derived NK cells maintain high cytotoxicity and enhance in vivo tumor control in concert with T cells and anti-PD-1 therapy. Sci. Transl. Med. 2020, 12, eaaz5618. [Google Scholar] [CrossRef]
- Li, Y.; Hermanson, D.L.; Moriarity, B.S.; Kaufman, D.S. Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-tumor Activity. Cell Stem Cell 2018, 23, 181–192.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilhelm, M.; Smetak, M.; Schaefer-Eckart, K.; Kimmel, B.; Birkmann, J.; Einsele, H.; Kunzmann, V. Successful adoptive transfer and in vivo expansion of haploidentical gammadelta T cells. J. Transl. Med. 2014, 12, 45. [Google Scholar] [CrossRef]
- Khairallah, C.; Chu, T.H.; Sheridan, B.S. Tissue Adaptations of Memory and Tissue-Resident Gamma Delta T Cells. Front. Immunol. 2018, 9, 2636. [Google Scholar] [CrossRef]
- Deniger, D.C.; Switzer, K.; Mi, T.; Maiti, S.; Hurton, L.; Singh, H.; Huls, H.; Olivares, S.; Lee, D.A.; Champlin, R.E.; et al. Bispecific T-cells expressing polyclonal repertoire of endogenous gammadelta T-cell receptors and introduced CD19-specific chimeric antigen receptor. Mol. Ther. 2013, 21, 638–647. [Google Scholar] [CrossRef] [Green Version]
- Capsomidis, A.; Benthall, G.; Van Acker, H.H.; Fisher, J.; Kramer, A.M.; Abeln, Z.; Majani, Y.; Gileadi, T.; Wallace, R.; Gustafsson, K.; et al. Chimeric Antigen Receptor-Engineered Human Gamma Delta T Cells: Enhanced Cytotoxicity with Retention of Cross Presentation. Mol. Ther. 2018, 26, 354–365. [Google Scholar] [CrossRef] [Green Version]
- Lamers, C.H.; Klaver, Y.; Gratama, J.W.; Sleijfer, S.; Debets, R. Treatment of metastatic renal cell carcinoma (mRCC) with CAIX CAR-engineered T-cells-a completed study overview. Biochem. Soc. Trans. 2016, 44, 951–959. [Google Scholar] [CrossRef] [PubMed]
- Lamers, C.H.; Sleijfer, S.; van Steenbergen, S.; van Elzakker, P.; van Krimpen, B.; Groot, C.; Vulto, A.; Bakker, M.D.; Oosterwijk, E.; Debets, R.; et al. Treatment of Metastatic Renal Cell Carcinoma With CAIX CAR-engineered T cells: Clinical Evaluation and Management of On-target Toxicity. Mol. Ther. 2013, 21, 904–912. [Google Scholar] [CrossRef] [PubMed]
- Vierboom, M.P.; Bos, G.M.; Ooms, M.; Offringa, R.; Melief, C.J. Cyclophosphamide enhances anti-tumor effect of wild-type p53-specific CTL. Int. J. Cancer 2000, 87, 253–260. [Google Scholar] [CrossRef]
- Reits, E.A.; Hodge, J.W.; Herberts, C.A.; Groothuis, T.A.; Chakraborty, M.; Wansley, E.K.; Camphausen, K.; Luiten, R.M.; De Ru, A.H.; Neijssen, J.; et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 2006, 203, 1259–1271. [Google Scholar] [CrossRef]
- Ganss, R.; Ryschich, E.; Klar, E.; Arnold, B.; Hämmerling, G.J. Combination of T-cell therapy and trigger of inflammation induces remodeling of the vasculature and tumor eradication. Cancer Res. 2002, 62, 1462–1470. [Google Scholar]
- Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.-D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef] [Green Version]
- Domschke, C.; Schneeweiss, A.; Stefanovic, S.; Wallwiener, M.; Heil, J.; Rom, J.; Sohn, C.; Beckhove, P.; Schuetz, F. Cellular Immune Responses and Immune Escape Mechanisms in Breast Cancer: Determinants of Immunotherapy. Breast Care 2016, 11, 102–107. [Google Scholar] [CrossRef] [Green Version]
- Neagu, M.R.; Reardon, D.A. An Update on the Role of Immunotherapy and Vaccine Strategies for Primary Brain Tumors. Curr. Treat. Options Oncol. 2015, 16, 54. [Google Scholar] [CrossRef]
- Ajina, A.; Maher, J. Prospects for combined use of oncolytic viruses and CAR T-cells. J. Immunother. Cancer 2017, 5, 90. [Google Scholar] [CrossRef] [Green Version]
- Nishio, N.; Diaconu, I.; Liu, H.; Cerullo, V.; Caruana, I.; Hoyos, V.; Bouchier-Hayes, L.; Savoldo, B.; Dotti, G. Armed Oncolytic Virus Enhances Immune Functions of Chimeric Antigen Receptor–Modified T Cells in Solid Tumors. Cancer Res. 2014, 74, 5195–5205. [Google Scholar] [CrossRef] [Green Version]
- Bonifant, C.; Jackson, H.J.; Brentjens, R.J.; Curran, K.J. Toxicity and management in CAR T-cell therapy. Mol. Ther. Oncolytics 2016, 3, 16011. [Google Scholar] [CrossRef] [PubMed]
- Curran, K.J.; Pegram, H.J.; Brentjens, R.J. Chimeric antigen receptors for T cell immunotherapy: Current understanding and future directions. J. Gene Med. 2012, 14, 405–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lamers, C.H.; Sleijfer, S.; Vulto, A.G.; Kruit, W.H.; 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] [PubMed]
- 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] [PubMed]
- 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 Pt 1, 6106–6115. [Google Scholar] [CrossRef] [Green Version]
Antigen | Cancer | Phase | Trial ID # |
---|---|---|---|
CD20 | Melanoma, Non Hodgkin Lymphoma, Mantle cell Lymphoma | Phase 1/2 | NCT04160195, NCT04186520 |
CD171 | Neuroblastoma | Phase 1 | NCT02311621 |
CEA | Lung, colorectal, gastric, breast, pancreatic, peritoneal, liver | Phase 1 | NCT03682744, NCT01373047, |
NCT03818165, NCT02416466, | |||
NCT02850536 | |||
Claudin 18.2 | Gastric, pancreatic | Phase 1 | NCT04404595 |
EGFRIII | Glioblastoma, gliosarcoma and brain tumor | Phase 1/2 | NCT01454596, NCT03283631 |
EGFR806 | Central nervous system tumor, pediatric glioma | Phase 1 | NCT03638167, NCT03618381 |
GD2 | Glioma | Phase 1 | NCT04099797 |
Glypican-3 | Liver | Phase 1 | NCT02932956, NCT02905188 |
NCT04377932 | |||
HER2 | Central nervous system tumor, pediatric glioma, breast | Phase 1/2 | NCT03500991, NCT03696030 |
NCT02442297, NCT03740256 | |||
NCT04483778, NCT03618381 | |||
NCT00924287, NCT04650451 | |||
NCT01109095 | |||
HLA-A2 | Ependymoma | Phase 1 | NCT01795313 |
IL-13Rα2 | Glioblastoma, cutaneous melanoma | Phase 1 | NCT02208362, NCT04119024 |
NCT04003649 | |||
KK-LC-1 | Epithelial | Phase 1 | NCT05035407 |
Mesothelin | Ovarian, cervical, pancreatic, lung, breast, mesothelioma | Phase 1/2 | NCT01583686, NCT04577326 |
NCT03054298, NCT02159716 | |||
NCT02414269 | |||
PSCA | Prostate cancer, metastatic pancreatic | Phase 1/2 | NCT03873805, NCT02744287 |
TAA-T | Hematopoietic malignancies, acute myeloid leukemia, MDS, Hodgkin lymphoma, B cell lymphoma | Phase 1 | NCT02203903, NCT03843294 |
VEGFR2 | Metastatic melanoma, renal | Phase 1/2 | NCT01218867 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Guha, P.; Heatherton, K.R.; O’Connell, K.P.; Alexander, I.S.; Katz, S.C. Assessing the Future of Solid Tumor Immunotherapy. Biomedicines 2022, 10, 655. https://doi.org/10.3390/biomedicines10030655
Guha P, Heatherton KR, O’Connell KP, Alexander IS, Katz SC. Assessing the Future of Solid Tumor Immunotherapy. Biomedicines. 2022; 10(3):655. https://doi.org/10.3390/biomedicines10030655
Chicago/Turabian StyleGuha, Prajna, Kara R. Heatherton, Kyle P. O’Connell, Ian S. Alexander, and Steven C. Katz. 2022. "Assessing the Future of Solid Tumor Immunotherapy" Biomedicines 10, no. 3: 655. https://doi.org/10.3390/biomedicines10030655
APA StyleGuha, P., Heatherton, K. R., O’Connell, K. P., Alexander, I. S., & Katz, S. C. (2022). Assessing the Future of Solid Tumor Immunotherapy. Biomedicines, 10(3), 655. https://doi.org/10.3390/biomedicines10030655