Skip to main content
Top
Published in: Cancer Cell International 1/2021

Open Access 01-12-2021 | Colorectal Cancer | Review

Monoclonal antibodies and chimeric antigen receptor (CAR) T cells in the treatment of colorectal cancer

Authors: Ke-Tao Jin, Bo Chen, Yu-Yao Liu, H uan-Rong Lan, Jie-Ping Yan

Published in: Cancer Cell International | Issue 1/2021

Login to get access

Abstract

Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer deaths worldwide. Besides common therapeutic approaches, such as surgery, chemotherapy, and radiotherapy, novel therapeutic approaches, including immunotherapy, have been an advent in CRC treatment. The immunotherapy approaches try to elicit patients` immune responses against tumor cells to eradicate the tumor. Monoclonal antibodies (mAbs) and chimeric antigen receptor (CAR) T cells are two branches of cancer immunotherapy. MAbs demonstrate the great ability to completely recognize cancer cell-surface receptors and blockade proliferative or inhibitory pathways. On the other hand, T cell activation by genetically engineered CAR receptor via the TCR/CD3 and costimulatory domains can induce potent immune responses against specific tumor-associated antigens (TAAs). Both of these approaches have beneficial anti-tumor effects on CRC. Herein, we review the different mAbs against various pathways and their applications in clinical trials, the different types of CAR-T cells, various specific CAR-T cells against TAAs, and their clinical use in CRC treatment.
Literature
1.
go back to reference Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRefPubMed Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRefPubMed
2.
go back to reference Veisi Malekshahi Z, Hashemi Goradel N, Shakouri Khomartash M, Maleksabet A, Kadkhodazadeh M, Kardar GA, et al. CEA plasmid as therapeutic DNA vaccination against colorectal cancer. Iran J Immunol. 2019;16:235–45.PubMed Veisi Malekshahi Z, Hashemi Goradel N, Shakouri Khomartash M, Maleksabet A, Kadkhodazadeh M, Kardar GA, et al. CEA plasmid as therapeutic DNA vaccination against colorectal cancer. Iran J Immunol. 2019;16:235–45.PubMed
3.
go back to reference De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 2010;11:753–62.PubMedCrossRef De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 2010;11:753–62.PubMedCrossRef
5.
go back to reference Martin FL, Martinez EZ, Stopper H, Garcia SB, Uyemura SA, Kannen V. Increased exposure to pesticides and colon cancer: Early evidence in Brazil. Chemosphere. 2018;209:623–31.PubMedCrossRef Martin FL, Martinez EZ, Stopper H, Garcia SB, Uyemura SA, Kannen V. Increased exposure to pesticides and colon cancer: Early evidence in Brazil. Chemosphere. 2018;209:623–31.PubMedCrossRef
6.
go back to reference Hashemi Goradel N, Heidarzadeh S, Jahangiri S, Farhood B, Mortezaee K, Khanlarkhani N, et al. Fusobacterium nucleatum and colorectal cancer: a mechanistic overview. J Cell Physiol. 2019;234:2337–44.PubMedCrossRef Hashemi Goradel N, Heidarzadeh S, Jahangiri S, Farhood B, Mortezaee K, Khanlarkhani N, et al. Fusobacterium nucleatum and colorectal cancer: a mechanistic overview. J Cell Physiol. 2019;234:2337–44.PubMedCrossRef
7.
go back to reference Khiavi MA, Safary A, Somi MH. Recent advances in targeted therapy of colorectal cancer: impacts of monoclonal antibodies nanoconjugates. BioImpacts BI. 2019;9:123.CrossRef Khiavi MA, Safary A, Somi MH. Recent advances in targeted therapy of colorectal cancer: impacts of monoclonal antibodies nanoconjugates. BioImpacts BI. 2019;9:123.CrossRef
8.
go back to reference Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27:1386–422.PubMedCrossRef Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27:1386–422.PubMedCrossRef
9.
go back to reference Goulart A, Ferreira C, Rodrigues A, Coimbra B, Sousa N, Leão P. The correlation between serum vascular endothelial growth factor (VEGF) and tumor VEGF receptor 3 in colorectal cancer. Ann Surg Treat Res. 2019;97:15–20.PubMedPubMedCentralCrossRef Goulart A, Ferreira C, Rodrigues A, Coimbra B, Sousa N, Leão P. The correlation between serum vascular endothelial growth factor (VEGF) and tumor VEGF receptor 3 in colorectal cancer. Ann Surg Treat Res. 2019;97:15–20.PubMedPubMedCentralCrossRef
10.
go back to reference Mokhtari M, Ardestani MM, Movahedipour M. An immunohistochemical study of EGFR expression in colorectal cancer and its correlation with lymph nodes status and tumor grade. J Res Med Sci Off J Isfahan Univ Med Sci. 2012;17:741. Mokhtari M, Ardestani MM, Movahedipour M. An immunohistochemical study of EGFR expression in colorectal cancer and its correlation with lymph nodes status and tumor grade. J Res Med Sci Off J Isfahan Univ Med Sci. 2012;17:741.
11.
go back to reference Shiratsuchi I, Akagi Y, Kawahara A, Kinugasa T, Romeo K, Yoshida T, et al. Expression of IGF-1 and IGF-1R and their relation to clinicopathological factors in colorectal cancer. Anticancer Res. 2011;31:2541–5.PubMed Shiratsuchi I, Akagi Y, Kawahara A, Kinugasa T, Romeo K, Yoshida T, et al. Expression of IGF-1 and IGF-1R and their relation to clinicopathological factors in colorectal cancer. Anticancer Res. 2011;31:2541–5.PubMed
12.
go back to reference Wang X-Y, Zheng Z-X, Sun Y, Bai Y-H, Shi Y-F, Zhou L-X, et al. Significance of HER2 protein expression and HER2 gene amplification in colorectal adenocarcinomas. World J Gastrointest Oncol. 2019;11:335.PubMedPubMedCentralCrossRef Wang X-Y, Zheng Z-X, Sun Y, Bai Y-H, Shi Y-F, Zhou L-X, et al. Significance of HER2 protein expression and HER2 gene amplification in colorectal adenocarcinomas. World J Gastrointest Oncol. 2019;11:335.PubMedPubMedCentralCrossRef
13.
go back to reference Vonlaufen A, Wiedle G, Borisch B, Birrer S, Luder P, Imhof BA. Integrin α v β 3 expression in colon carcinoma correlates with survival. Mod Pathol. 2001;14:1126–32.PubMedCrossRef Vonlaufen A, Wiedle G, Borisch B, Birrer S, Luder P, Imhof BA. Integrin α v β 3 expression in colon carcinoma correlates with survival. Mod Pathol. 2001;14:1126–32.PubMedCrossRef
14.
go back to reference Pothuraju R, Rachagani S, Krishn SR, Chaudhary S, Nimmakayala RK, Siddiqui JA, et al. Molecular implications of MUC5AC-CD44 axis in colorectal cancer progression and chemoresistance. Mol Cancer. 2020;19:1–14.CrossRef Pothuraju R, Rachagani S, Krishn SR, Chaudhary S, Nimmakayala RK, Siddiqui JA, et al. Molecular implications of MUC5AC-CD44 axis in colorectal cancer progression and chemoresistance. Mol Cancer. 2020;19:1–14.CrossRef
15.
go back to reference Devetzi M, Kosmidou V, Vlassi M, Perysinakis I, Aggeli C, Choreftaki T, et al. Death receptor 5 (DR5) and a 5-gene apoptotic biomarker panel with significant differential diagnostic potential in colorectal cancer. Sci Rep. 2016;6:36532.PubMedPubMedCentralCrossRef Devetzi M, Kosmidou V, Vlassi M, Perysinakis I, Aggeli C, Choreftaki T, et al. Death receptor 5 (DR5) and a 5-gene apoptotic biomarker panel with significant differential diagnostic potential in colorectal cancer. Sci Rep. 2016;6:36532.PubMedPubMedCentralCrossRef
16.
go back to reference Omura Y, Toiyama Y, Okugawa Y, Yin C, Shigemori T, Kusunoki K, et al. Prognostic impacts of tumoral expression and serum levels of PD-L1 and CTLA-4 in colorectal cancer patients. Cancer Immunol Immunother. 2020;69:2533–46.PubMedCrossRef Omura Y, Toiyama Y, Okugawa Y, Yin C, Shigemori T, Kusunoki K, et al. Prognostic impacts of tumoral expression and serum levels of PD-L1 and CTLA-4 in colorectal cancer patients. Cancer Immunol Immunother. 2020;69:2533–46.PubMedCrossRef
17.
go back to reference Markman JL, Shiao SL. Impact of the immune system and immunotherapy in colorectal cancer. J Gastrointest Oncol. 2015;6:208.PubMedPubMedCentral Markman JL, Shiao SL. Impact of the immune system and immunotherapy in colorectal cancer. J Gastrointest Oncol. 2015;6:208.PubMedPubMedCentral
18.
go back to reference Ge P, Wang W, Li L, Zhang G, Gao Z, Tang Z, et al. Profiles of immune cell infiltration and immune-related genes in the tumor microenvironment of colorectal cancer. Biomed Pharmacother. 2019;118:109228.PubMedCrossRef Ge P, Wang W, Li L, Zhang G, Gao Z, Tang Z, et al. Profiles of immune cell infiltration and immune-related genes in the tumor microenvironment of colorectal cancer. Biomed Pharmacother. 2019;118:109228.PubMedCrossRef
19.
go back to reference Roelands J, Kuppen PJK, Vermeulen L, Maccalli C, Decock J, Wang E, et al. Immunogenomic classification of colorectal cancer and therapeutic implications. Int J Mol Sci. 2017;18:2229.PubMedCentralCrossRef Roelands J, Kuppen PJK, Vermeulen L, Maccalli C, Decock J, Wang E, et al. Immunogenomic classification of colorectal cancer and therapeutic implications. Int J Mol Sci. 2017;18:2229.PubMedCentralCrossRef
20.
go back to reference Fleming V, Hu X, Weber R, Nagibin V, Groth C, Altevogt P, et al. Targeting myeloid-derived suppressor cells to bypass tumor-induced immunosuppression. Front Immunol. 2018;9:398.PubMedPubMedCentralCrossRef Fleming V, Hu X, Weber R, Nagibin V, Groth C, Altevogt P, et al. Targeting myeloid-derived suppressor cells to bypass tumor-induced immunosuppression. Front Immunol. 2018;9:398.PubMedPubMedCentralCrossRef
21.
go back to reference Hu G, Wu P, Cheng P, Zhang Z, Wang Z, Yu X, et al. Tumor-infiltrating CD39+ γ δ Tregs are novel immunosuppressive T cells in human colorectal cancer. Oncoimmunology. 2017;6:e1277305.PubMedPubMedCentralCrossRef Hu G, Wu P, Cheng P, Zhang Z, Wang Z, Yu X, et al. Tumor-infiltrating CD39+ γ δ Tregs are novel immunosuppressive T cells in human colorectal cancer. Oncoimmunology. 2017;6:e1277305.PubMedPubMedCentralCrossRef
22.
go back to reference Wei C, Yang C, Wang S, Shi D, Zhang C, Lin X, et al. M2 macrophages confer resistance to 5-fluorouracil in colorectal cancer through the activation of CCL22/PI3K/AKT signaling. Onco Targets Ther. 2019;12:3051.PubMedPubMedCentralCrossRef Wei C, Yang C, Wang S, Shi D, Zhang C, Lin X, et al. M2 macrophages confer resistance to 5-fluorouracil in colorectal cancer through the activation of CCL22/PI3K/AKT signaling. Onco Targets Ther. 2019;12:3051.PubMedPubMedCentralCrossRef
23.
go back to reference Lan J, Sun L, Xu F, Liu L, Hu F, Song D, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res. 2019;79:146–58.PubMedCrossRef Lan J, Sun L, Xu F, Liu L, Hu F, Song D, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res. 2019;79:146–58.PubMedCrossRef
24.
go back to reference Marech I, Ammendola M, Sacco R, Sammarco G, Zuccalà V, Zizzo N, et al. Tumour-associated macrophages correlate with microvascular bed extension in colorectal cancer patients. J Cell Mol Med. 2016;20:1373–80.PubMedPubMedCentralCrossRef Marech I, Ammendola M, Sacco R, Sammarco G, Zuccalà V, Zizzo N, et al. Tumour-associated macrophages correlate with microvascular bed extension in colorectal cancer patients. J Cell Mol Med. 2016;20:1373–80.PubMedPubMedCentralCrossRef
25.
go back to reference Nosho K, Sukawa Y, Adachi Y, Ito M, Mitsuhashi K, Kurihara H, et al. Association of Fusobacterium nucleatum with immunity and molecular alterations in colorectal cancer. World J Gastroenterol. 2016;22:557.PubMedPubMedCentralCrossRef Nosho K, Sukawa Y, Adachi Y, Ito M, Mitsuhashi K, Kurihara H, et al. Association of Fusobacterium nucleatum with immunity and molecular alterations in colorectal cancer. World J Gastroenterol. 2016;22:557.PubMedPubMedCentralCrossRef
26.
go back to reference Köhler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–7.PubMedCrossRef Köhler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–7.PubMedCrossRef
27.
go back to reference Buss NAPS, Henderson SJ, McFarlane M, Shenton JM, De Haan L. Monoclonal antibody therapeutics: history and future. Curr Opin Pharmacol. 2012;12:615–22.PubMedCrossRef Buss NAPS, Henderson SJ, McFarlane M, Shenton JM, De Haan L. Monoclonal antibody therapeutics: history and future. Curr Opin Pharmacol. 2012;12:615–22.PubMedCrossRef
28.
go back to reference Presta LG. Engineering of therapeutic antibodies to minimize immunogenicity and optimize function. Adv Drug Deliv Rev. 2006;58:640–56.PubMedCrossRef Presta LG. Engineering of therapeutic antibodies to minimize immunogenicity and optimize function. Adv Drug Deliv Rev. 2006;58:640–56.PubMedCrossRef
29.
go back to reference Harding FA, Stickler MM, Razo J, DuBridge R. The immunogenicity of humanized and fully human antibodies: residual immunogenicity resides in the CDR regions. MAbs. 2010;2:256–65.PubMedPubMedCentralCrossRef Harding FA, Stickler MM, Razo J, DuBridge R. The immunogenicity of humanized and fully human antibodies: residual immunogenicity resides in the CDR regions. MAbs. 2010;2:256–65.PubMedPubMedCentralCrossRef
30.
go back to reference Mallbris L, Davies J, Glasebrook A, Tang Y, Glaesner W, Nickoloff BJ. Molecular insights into fully human and humanized monoclonal antibodies: What are the differences and should dermatologists care? J Clin Aesthet Dermatol. 2016;9:13.PubMedPubMedCentral Mallbris L, Davies J, Glasebrook A, Tang Y, Glaesner W, Nickoloff BJ. Molecular insights into fully human and humanized monoclonal antibodies: What are the differences and should dermatologists care? J Clin Aesthet Dermatol. 2016;9:13.PubMedPubMedCentral
31.
32.
go back to reference Lu R-M, Hwang Y-C, Liu I-J, Lee C-C, Tsai H-Z, Li H-J, et al. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci. 2020;27:1–30.PubMedPubMedCentralCrossRef Lu R-M, Hwang Y-C, Liu I-J, Lee C-C, Tsai H-Z, Li H-J, et al. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci. 2020;27:1–30.PubMedPubMedCentralCrossRef
33.
go back to reference Tsumoto K, Isozaki Y, Yagami H, Tomita M. Future perspectives of therapeutic monoclonal antibodies. Immunotherapy. 2019;11:119–27.PubMedCrossRef Tsumoto K, Isozaki Y, Yagami H, Tomita M. Future perspectives of therapeutic monoclonal antibodies. Immunotherapy. 2019;11:119–27.PubMedCrossRef
34.
go back to reference Douillard J-Y, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, et al. Panitumumab–FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023–34.PubMedCrossRef Douillard J-Y, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, et al. Panitumumab–FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023–34.PubMedCrossRef
35.
go back to reference Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen Y-J, Ciombor KK, et al. NCCN guidelines insights: colon cancer, version 2.2018. J Natl Compr Cancer Netw. 2018;16:359–69.CrossRef Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen Y-J, Ciombor KK, et al. NCCN guidelines insights: colon cancer, version 2.2018. J Natl Compr Cancer Netw. 2018;16:359–69.CrossRef
36.
go back to reference Hashemi Goradel N, Ghiyami-Hour F, Jahangiri S, Negahdari B, Sahebkar A, Masoudifar A, et al. Nanoparticles as new tools for inhibition of cancer angiogenesis. J Cell Physiol. 2018;233:2902–10.PubMedCrossRef Hashemi Goradel N, Ghiyami-Hour F, Jahangiri S, Negahdari B, Sahebkar A, Masoudifar A, et al. Nanoparticles as new tools for inhibition of cancer angiogenesis. J Cell Physiol. 2018;233:2902–10.PubMedCrossRef
37.
go back to reference Goradel NH, Asghari MH, Moloudizargari M, Negahdari B, Haghi-Aminjan H, Abdollahi M. Melatonin as an angiogenesis inhibitor to combat cancer: Mechanistic evidence. Toxicol Appl Pharmacol. 2017;335:56–63.PubMedCrossRef Goradel NH, Asghari MH, Moloudizargari M, Negahdari B, Haghi-Aminjan H, Abdollahi M. Melatonin as an angiogenesis inhibitor to combat cancer: Mechanistic evidence. Toxicol Appl Pharmacol. 2017;335:56–63.PubMedCrossRef
38.
go back to reference Goradel NH, Mohammadi N, Haghi-Aminjan H, Farhood B, Negahdari B, Sahebkar A. Regulation of tumor angiogenesis by microRNAs: State of the art. J Cell Physiol. 2019;234:1099–110.PubMedCrossRef Goradel NH, Mohammadi N, Haghi-Aminjan H, Farhood B, Negahdari B, Sahebkar A. Regulation of tumor angiogenesis by microRNAs: State of the art. J Cell Physiol. 2019;234:1099–110.PubMedCrossRef
39.
go back to reference Costache MI, Ioana M, Iordache S, Ene D, Costache CA, Săftoiu A. VEGF expression in pancreatic cancer and other malignancies: a review of the literature. Rom J Intern Med. 2015;53:199–208.PubMed Costache MI, Ioana M, Iordache S, Ene D, Costache CA, Săftoiu A. VEGF expression in pancreatic cancer and other malignancies: a review of the literature. Rom J Intern Med. 2015;53:199–208.PubMed
40.
go back to reference Bendardaf R, Buhmeida A, Hilska M, Laato M, Syrjänen S, Syrjänen K, et al. VEGF-1 expression in colorectal cancer is associated with disease localization, stage, and long-term disease-specific survival. Anticancer Res. 2008;28:3865–70.PubMed Bendardaf R, Buhmeida A, Hilska M, Laato M, Syrjänen S, Syrjänen K, et al. VEGF-1 expression in colorectal cancer is associated with disease localization, stage, and long-term disease-specific survival. Anticancer Res. 2008;28:3865–70.PubMed
41.
go back to reference Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R, et al. Bevacizumab (Avastin®) in cancer treatment: a review of 15 years of clinical experience and future outlook. Cancer Treat Rev. 2020;86:102017.PubMedCrossRef Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R, et al. Bevacizumab (Avastin®) in cancer treatment: a review of 15 years of clinical experience and future outlook. Cancer Treat Rev. 2020;86:102017.PubMedCrossRef
42.
43.
go back to reference Hey SP, Gyawali B, D’Andrea E, Kanagaraj M, Franklin JM, Kesselheim AS. A systematic review and meta-analysis of bevacizumab in first-line metastatic breast cancer: lessons for research and regulatory Enterprises. JNCI J Natl Cancer Inst. 2020;112:335–42.PubMedCrossRef Hey SP, Gyawali B, D’Andrea E, Kanagaraj M, Franklin JM, Kesselheim AS. A systematic review and meta-analysis of bevacizumab in first-line metastatic breast cancer: lessons for research and regulatory Enterprises. JNCI J Natl Cancer Inst. 2020;112:335–42.PubMedCrossRef
44.
go back to reference Marchetti C, Muzii L, Romito A, Panici PB. First-line treatment of women with advanced ovarian cancer: focus on bevacizumab. Onco Targets Ther. 2019;12:1095.PubMedPubMedCentralCrossRef Marchetti C, Muzii L, Romito A, Panici PB. First-line treatment of women with advanced ovarian cancer: focus on bevacizumab. Onco Targets Ther. 2019;12:1095.PubMedPubMedCentralCrossRef
45.
go back to reference Seto T, Azuma K, Yamanaka T, Sugawara S, Yoshioka H, Wakuda K, et al. Randomized phase III study of continuation maintenance bevacizumab with or without pemetrexed in advanced nonsquamous non–small-cell lung cancer: COMPASS (WJOG5610L). J Clin Oncol. 2020;38:793–803.PubMedCrossRef Seto T, Azuma K, Yamanaka T, Sugawara S, Yoshioka H, Wakuda K, et al. Randomized phase III study of continuation maintenance bevacizumab with or without pemetrexed in advanced nonsquamous non–small-cell lung cancer: COMPASS (WJOG5610L). J Clin Oncol. 2020;38:793–803.PubMedCrossRef
46.
go back to reference Kabbinavar F, Hurwitz HI, Fehrenbacher L, Meropol NJ, Novotny WF, Lieberman G, et al. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer. J Clin Oncol. 2003;21:60–5.PubMedCrossRef Kabbinavar F, Hurwitz HI, Fehrenbacher L, Meropol NJ, Novotny WF, Lieberman G, et al. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer. J Clin Oncol. 2003;21:60–5.PubMedCrossRef
47.
go back to reference Saltz LB, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol. 2008;26:2013–9.PubMedCrossRef Saltz LB, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol. 2008;26:2013–9.PubMedCrossRef
48.
go back to reference Bennouna J, Sastre J, Arnold D, Österlund P, Greil R, Van Cutsem E, et al. Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): a randomised phase 3 trial. Lancet Oncol. 2013;14:29–37.PubMedCrossRef Bennouna J, Sastre J, Arnold D, Österlund P, Greil R, Van Cutsem E, et al. Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): a randomised phase 3 trial. Lancet Oncol. 2013;14:29–37.PubMedCrossRef
49.
go back to reference Baraniskin A, Buchberger B, Pox C, Graeven U, Holch JW, Schmiegel W, et al. Efficacy of bevacizumab in first-line treatment of metastatic colorectal cancer: a systematic review and meta-analysis. Eur J Cancer. 2019;106:37–44.PubMedCrossRef Baraniskin A, Buchberger B, Pox C, Graeven U, Holch JW, Schmiegel W, et al. Efficacy of bevacizumab in first-line treatment of metastatic colorectal cancer: a systematic review and meta-analysis. Eur J Cancer. 2019;106:37–44.PubMedCrossRef
50.
go back to reference Verdaguer H, Tabernero J, Macarulla T. Ramucirumab in metastatic colorectal cancer: evidence to date and place in therapy. Ther Adv Med Oncol. 2016;8:230–42.PubMedPubMedCentralCrossRef Verdaguer H, Tabernero J, Macarulla T. Ramucirumab in metastatic colorectal cancer: evidence to date and place in therapy. Ther Adv Med Oncol. 2016;8:230–42.PubMedPubMedCentralCrossRef
51.
go back to reference Chiorean EG, Hurwitz HI, Cohen RB, Schwartz JD, Dalal RP, Fox FE, et al. Phase I study of every 2-or 3-week dosing of ramucirumab, a human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2 in patients with advanced solid tumors. Ann Oncol. 2015;26:1230–7.PubMedCrossRef Chiorean EG, Hurwitz HI, Cohen RB, Schwartz JD, Dalal RP, Fox FE, et al. Phase I study of every 2-or 3-week dosing of ramucirumab, a human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2 in patients with advanced solid tumors. Ann Oncol. 2015;26:1230–7.PubMedCrossRef
52.
go back to reference Spratlin JL, Cohen RB, Eadens M, Gore L, Camidge DR, Diab S, et al. Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J Clin Oncol. 2010;28:780.PubMedPubMedCentralCrossRef Spratlin JL, Cohen RB, Eadens M, Gore L, Camidge DR, Diab S, et al. Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J Clin Oncol. 2010;28:780.PubMedPubMedCentralCrossRef
53.
go back to reference Garcia-Carbonero R, Rivera F, Maurel J, Ayoub J-PM, Moore MJ, Cervantes A, et al. An open-label phase II study evaluating the safety and efficacy of ramucirumab combined with mFOLFOX-6 as first-line therapy for metastatic colorectal cancer. Oncologist. 2014;19:350.PubMedPubMedCentralCrossRef Garcia-Carbonero R, Rivera F, Maurel J, Ayoub J-PM, Moore MJ, Cervantes A, et al. An open-label phase II study evaluating the safety and efficacy of ramucirumab combined with mFOLFOX-6 as first-line therapy for metastatic colorectal cancer. Oncologist. 2014;19:350.PubMedPubMedCentralCrossRef
54.
go back to reference Tabernero J, Yoshino T, Cohn AL, Obermannova R, Bodoky G, Garcia-Carbonero R, et al. Ramucirumab versus placebo in combination with second-line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, double-blin. Lancet Oncol. 2015;16:499–508.PubMedCrossRef Tabernero J, Yoshino T, Cohn AL, Obermannova R, Bodoky G, Garcia-Carbonero R, et al. Ramucirumab versus placebo in combination with second-line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, double-blin. Lancet Oncol. 2015;16:499–508.PubMedCrossRef
56.
go back to reference Kim ES, Serur A, Huang J, Manley CA, McCrudden KW, Frischer JS, et al. Potent VEGF blockade causes regression of coopted vessels in a model of neuroblastoma. Proc Natl Acad Sci. 2002;99:11399–404.PubMedCrossRefPubMedCentral Kim ES, Serur A, Huang J, Manley CA, McCrudden KW, Frischer JS, et al. Potent VEGF blockade causes regression of coopted vessels in a model of neuroblastoma. Proc Natl Acad Sci. 2002;99:11399–404.PubMedCrossRefPubMedCentral
57.
go back to reference Khayat D, Tejpar S, Spano J-P, Verslype C, Bloch J, Vandecaveye V, et al. Intravenous aflibercept administered in combination with irinotecan, 5-fluorouracil and leucovorin in patients with advanced solid tumours: results from the expansion cohort of a phase I study. Eur J Cancer. 2013;49:790–7.PubMedCrossRef Khayat D, Tejpar S, Spano J-P, Verslype C, Bloch J, Vandecaveye V, et al. Intravenous aflibercept administered in combination with irinotecan, 5-fluorouracil and leucovorin in patients with advanced solid tumours: results from the expansion cohort of a phase I study. Eur J Cancer. 2013;49:790–7.PubMedCrossRef
58.
go back to reference Lockhart AC, Rothenberg ML, Dupont J, Cooper W, Chevalier P, Sternas L, et al. Phase I study of intravenous vascular endothelial growth factor trap, aflibercept, in patients with advanced solid tumors. J Clin Oncol. 2010;28:207.PubMedCrossRef Lockhart AC, Rothenberg ML, Dupont J, Cooper W, Chevalier P, Sternas L, et al. Phase I study of intravenous vascular endothelial growth factor trap, aflibercept, in patients with advanced solid tumors. J Clin Oncol. 2010;28:207.PubMedCrossRef
59.
go back to reference Townsley CA, Siu LL, San Pedro-Salcedo M, Liu L, Wakelee HA. A phase I study of aflibercept, pemetrexed (P), and cisplatin (C) in patients with advanced solid tumors. J Clin Oncol. 2010;28:2536.CrossRef Townsley CA, Siu LL, San Pedro-Salcedo M, Liu L, Wakelee HA. A phase I study of aflibercept, pemetrexed (P), and cisplatin (C) in patients with advanced solid tumors. J Clin Oncol. 2010;28:2536.CrossRef
60.
go back to reference Denda T, Sakai D, Hamaguchi T, Sugimoto N, Ura T, Yamazaki K, et al. Phase II trial of aflibercept with FOLFIRI as a second-line treatment for Japanese patients with metastatic colorectal cancer. Cancer Sci. 2019;110:1032–43.PubMedPubMedCentralCrossRef Denda T, Sakai D, Hamaguchi T, Sugimoto N, Ura T, Yamazaki K, et al. Phase II trial of aflibercept with FOLFIRI as a second-line treatment for Japanese patients with metastatic colorectal cancer. Cancer Sci. 2019;110:1032–43.PubMedPubMedCentralCrossRef
61.
go back to reference Van Cutsem E, Tabernero J, Lakomy R, Prenen H, Prausová J, Macarulla T, et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol. 2012;30:3499–506.PubMedCrossRef Van Cutsem E, Tabernero J, Lakomy R, Prenen H, Prausová J, Macarulla T, et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol. 2012;30:3499–506.PubMedCrossRef
62.
go back to reference Lee SH. Tanibirumab (TTAC-0001): a fully human monoclonal antibody targets vascular endothelial growth factor receptor 2 (VEGFR-2). Arch Pharm Res. 2011;34:1223.PubMedCrossRef Lee SH. Tanibirumab (TTAC-0001): a fully human monoclonal antibody targets vascular endothelial growth factor receptor 2 (VEGFR-2). Arch Pharm Res. 2011;34:1223.PubMedCrossRef
63.
go back to reference Lee SJ, Lee SY, Lee WS, San Yoo J, Sun J-M, Lee J, et al. Phase I trial and pharmacokinetic study of tanibirumab, a fully human monoclonal antibody to vascular endothelial growth factor receptor 2, in patients with refractory solid tumors. Invest New Drugs. 2017;35:782–90.PubMedPubMedCentralCrossRef Lee SJ, Lee SY, Lee WS, San Yoo J, Sun J-M, Lee J, et al. Phase I trial and pharmacokinetic study of tanibirumab, a fully human monoclonal antibody to vascular endothelial growth factor receptor 2, in patients with refractory solid tumors. Invest New Drugs. 2017;35:782–90.PubMedPubMedCentralCrossRef
64.
go back to reference Kienast Y, Klein C, Scheuer W, Raemsch R, Lorenzon E, Bernicke D, et al. Ang-2-VEGF-A CrossMab, a novel bispecific human IgG1 antibody blocking VEGF-A and Ang-2 functions simultaneously, mediates potent antitumor, antiangiogenic, and antimetastatic efficacy. Clin Cancer Res. 2013;19:6730–40.PubMedCrossRef Kienast Y, Klein C, Scheuer W, Raemsch R, Lorenzon E, Bernicke D, et al. Ang-2-VEGF-A CrossMab, a novel bispecific human IgG1 antibody blocking VEGF-A and Ang-2 functions simultaneously, mediates potent antitumor, antiangiogenic, and antimetastatic efficacy. Clin Cancer Res. 2013;19:6730–40.PubMedCrossRef
65.
go back to reference Schaefer W, Regula JT, Bähner M, Schanzer J, Croasdale R, Dürr H, et al. Immunoglobulin domain crossover as a generic approach for the production of bispecific IgG antibodies. Proc Natl Acad Sci. 2011;108:11187–92.PubMedCrossRefPubMedCentral Schaefer W, Regula JT, Bähner M, Schanzer J, Croasdale R, Dürr H, et al. Immunoglobulin domain crossover as a generic approach for the production of bispecific IgG antibodies. Proc Natl Acad Sci. 2011;108:11187–92.PubMedCrossRefPubMedCentral
66.
go back to reference Hidalgo M, Martinez-Garcia M, Le Tourneau C, Massard C, Garralda E, Boni V, et al. First-in-human phase I study of single-agent vanucizumab, a first-in-class bispecific anti-angiopoietin-2/anti-VEGF-A antibody, in adult patients with advanced solid tumors. Clin Cancer Res. 2018;24:1536–45.PubMedCrossRef Hidalgo M, Martinez-Garcia M, Le Tourneau C, Massard C, Garralda E, Boni V, et al. First-in-human phase I study of single-agent vanucizumab, a first-in-class bispecific anti-angiopoietin-2/anti-VEGF-A antibody, in adult patients with advanced solid tumors. Clin Cancer Res. 2018;24:1536–45.PubMedCrossRef
67.
go back to reference Bendell JC, Sauri T, Gracián AC, Alvarez R, López-López C, García-Alfonso P, et al. The McCAVE trial: VANUCIZUMAB plus mFOLFOX-6 Versus Bevacizumab plus mFOLFOX-6 in patients with previously untreated metastatic colorectal carcinoma (mCRC). Oncologist. 2020;25:e451.PubMedCrossRef Bendell JC, Sauri T, Gracián AC, Alvarez R, López-López C, García-Alfonso P, et al. The McCAVE trial: VANUCIZUMAB plus mFOLFOX-6 Versus Bevacizumab plus mFOLFOX-6 in patients with previously untreated metastatic colorectal carcinoma (mCRC). Oncologist. 2020;25:e451.PubMedCrossRef
68.
go back to reference Costa R, Shah AN, Santa-Maria CA, Cruz MR, Mahalingam D, Carneiro BA, et al. Targeting epidermal growth factor receptor in triple negative breast cancer: new discoveries and practical insights for drug development. Cancer Treat Rev. 2017;53:111–9.PubMedCrossRef Costa R, Shah AN, Santa-Maria CA, Cruz MR, Mahalingam D, Carneiro BA, et al. Targeting epidermal growth factor receptor in triple negative breast cancer: new discoveries and practical insights for drug development. Cancer Treat Rev. 2017;53:111–9.PubMedCrossRef
69.
go back to reference Sforza V, Martinelli E, Ciardiello F, Gambardella V, Napolitano S, Martini G, et al. Mechanisms of resistance to anti-epidermal growth factor receptor inhibitors in metastatic colorectal cancer. World J Gastroenterol. 2016;22:6345.PubMedPubMedCentralCrossRef Sforza V, Martinelli E, Ciardiello F, Gambardella V, Napolitano S, Martini G, et al. Mechanisms of resistance to anti-epidermal growth factor receptor inhibitors in metastatic colorectal cancer. World J Gastroenterol. 2016;22:6345.PubMedPubMedCentralCrossRef
70.
go back to reference Krasinskas AM. EGFR signaling in colorectal carcinoma. Patholog Res Int 2011;2011. Krasinskas AM. EGFR signaling in colorectal carcinoma. Patholog Res Int 2011;2011.
71.
go back to reference Li N, Bu X, Wu P, Wu P, Huang P. The, “HER2–PI3K/Akt–FASN Axis” regulated malignant phenotype of colorectal cancer cells. Lipids. 2012;47:403–11.PubMedCrossRef Li N, Bu X, Wu P, Wu P, Huang P. The, “HER2–PI3K/Akt–FASN Axis” regulated malignant phenotype of colorectal cancer cells. Lipids. 2012;47:403–11.PubMedCrossRef
72.
go back to reference Huang C-W, Tsai H-L, Chen Y-T, Huang C-M, Ma C-J, Lu C-Y, et al. The prognostic values of EGFR expression and KRAS mutation in patients with synchronous or metachronous metastatic colorectal cancer. BMC Cancer. 2013;13:599.PubMedPubMedCentralCrossRef Huang C-W, Tsai H-L, Chen Y-T, Huang C-M, Ma C-J, Lu C-Y, et al. The prognostic values of EGFR expression and KRAS mutation in patients with synchronous or metachronous metastatic colorectal cancer. BMC Cancer. 2013;13:599.PubMedPubMedCentralCrossRef
73.
go back to reference Gomez D, De Rosa A, Addison A, Brooks A, Malik HZ, Cameron IC. Cetuximab therapy in the treatment of metastatic colorectal cancer: the future frontier? Int J Surg. 2013;11:507–13.PubMedCrossRef Gomez D, De Rosa A, Addison A, Brooks A, Malik HZ, Cameron IC. Cetuximab therapy in the treatment of metastatic colorectal cancer: the future frontier? Int J Surg. 2013;11:507–13.PubMedCrossRef
74.
go back to reference Gridelli C, Maione P, Ferrara ML, Rossi A. Cetuximab and other anti-epidermal growth factor receptor monoclonal antibodies in the treatment of non-small cell lung cancer. Oncologist. 2009;14:601–11.PubMedCrossRef Gridelli C, Maione P, Ferrara ML, Rossi A. Cetuximab and other anti-epidermal growth factor receptor monoclonal antibodies in the treatment of non-small cell lung cancer. Oncologist. 2009;14:601–11.PubMedCrossRef
75.
go back to reference Bokemeyer C, Bondarenko I, Hartmann JT, De Braud F, Schuch G, Zubel A, et al. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: the OPUS study. Ann Oncol. 2011;22:1535–46.PubMedCrossRef Bokemeyer C, Bondarenko I, Hartmann JT, De Braud F, Schuch G, Zubel A, et al. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: the OPUS study. Ann Oncol. 2011;22:1535–46.PubMedCrossRef
76.
go back to reference Saltz LB, Meropol NJ, Loehrer PJ Sr, Needle MN, Kopit J, Mayer RJ. Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. J Clin Oncol. 2004;22:1201–8.PubMedCrossRef Saltz LB, Meropol NJ, Loehrer PJ Sr, Needle MN, Kopit J, Mayer RJ. Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. J Clin Oncol. 2004;22:1201–8.PubMedCrossRef
77.
go back to reference Matsuda A, Yamada T, Jamjittrong S, Shinji S, Ohta R, Sonoda H, et al. Comparison between biweekly and weekly cetuximab in patients with metastatic colorectal cancer: a meta-analysis. Anticancer Res. 2020;40:3469–76.PubMedCrossRef Matsuda A, Yamada T, Jamjittrong S, Shinji S, Ohta R, Sonoda H, et al. Comparison between biweekly and weekly cetuximab in patients with metastatic colorectal cancer: a meta-analysis. Anticancer Res. 2020;40:3469–76.PubMedCrossRef
78.
go back to reference Battaglin F, Puccini A, Djaballah SA, Lenz H-J. The impact of panitumumab treatment on survival and quality of life in patients with RAS wild-type metastatic colorectal cancer. Cancer Manag Res. 2019;11:5911.PubMedPubMedCentralCrossRef Battaglin F, Puccini A, Djaballah SA, Lenz H-J. The impact of panitumumab treatment on survival and quality of life in patients with RAS wild-type metastatic colorectal cancer. Cancer Manag Res. 2019;11:5911.PubMedPubMedCentralCrossRef
79.
go back to reference Wainberg Z, Hecht JR. Panitumumab in colon cancer: a review and summary of ongoing trials. Expert Opin Biol Ther. 2006;6:1229–35.PubMedCrossRef Wainberg Z, Hecht JR. Panitumumab in colon cancer: a review and summary of ongoing trials. Expert Opin Biol Ther. 2006;6:1229–35.PubMedCrossRef
80.
go back to reference Veronese ML, O’Dwyer PJ. Monoclonal antibodies in the treatment of colorectal cancer. Eur J Cancer. 2004;40:1292–301.PubMedCrossRef Veronese ML, O’Dwyer PJ. Monoclonal antibodies in the treatment of colorectal cancer. Eur J Cancer. 2004;40:1292–301.PubMedCrossRef
81.
go back to reference Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, et al. Final results from PRIME: randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer. Ann Oncol. 2014;25:1346–55.PubMedCrossRef Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, et al. Final results from PRIME: randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer. Ann Oncol. 2014;25:1346–55.PubMedCrossRef
82.
go back to reference Lievre A, Bachet J-B, Le Corre D, Boige V, Landi B, Emile J-F, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006;66:3992–5.PubMedCrossRef Lievre A, Bachet J-B, Le Corre D, Boige V, Landi B, Emile J-F, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006;66:3992–5.PubMedCrossRef
83.
go back to reference Prewett M, Tonra JR, Rajiv B, Hooper AT, Makhoul G, Finnerty B, et al. Antitumor activity of a novel, human anti-epidermal growth factor receptor (EGFR) monoclonal antibody (IMC-11F8) in human tumor xenograft models. AACR. 2004;64:1235. Prewett M, Tonra JR, Rajiv B, Hooper AT, Makhoul G, Finnerty B, et al. Antitumor activity of a novel, human anti-epidermal growth factor receptor (EGFR) monoclonal antibody (IMC-11F8) in human tumor xenograft models. AACR. 2004;64:1235.
84.
go back to reference Elez E, Hendlisz A, Delaunoit T, Sastre J, Cervantes A, Varea R, et al. Phase II study of necitumumab plus modified FOLFOX6 as first-line treatment in patients with locally advanced or metastatic colorectal cancer. Br J Cancer. 2016;114:372–80.PubMedPubMedCentralCrossRef Elez E, Hendlisz A, Delaunoit T, Sastre J, Cervantes A, Varea R, et al. Phase II study of necitumumab plus modified FOLFOX6 as first-line treatment in patients with locally advanced or metastatic colorectal cancer. Br J Cancer. 2016;114:372–80.PubMedPubMedCentralCrossRef
85.
go back to reference Lewis AL, Chaft J, Girotra M, Fischer GW. Immune checkpoint inhibitors: a narrative review of considerations for the anaesthesiologist. Br J Anaesth. 2020;124:251–60.PubMedCrossRefPubMedCentral Lewis AL, Chaft J, Girotra M, Fischer GW. Immune checkpoint inhibitors: a narrative review of considerations for the anaesthesiologist. Br J Anaesth. 2020;124:251–60.PubMedCrossRefPubMedCentral
86.
go back to reference Huyghe N, Baldin P, Van den Eynde M. Immunotherapy with immune checkpoint inhibitors in colorectal cancer: what is the future beyond deficient mismatch-repair tumours? Gastroenterol Rep. 2020;8:11–24.CrossRef Huyghe N, Baldin P, Van den Eynde M. Immunotherapy with immune checkpoint inhibitors in colorectal cancer: what is the future beyond deficient mismatch-repair tumours? Gastroenterol Rep. 2020;8:11–24.CrossRef
87.
go back to reference Jácome AA, Eng C. Role of immune checkpoint inhibitors in the treatment of colorectal cancer: focus on nivolumab. Expert Opin Biol Ther. 2019;19:1247–63.PubMedCrossRef Jácome AA, Eng C. Role of immune checkpoint inhibitors in the treatment of colorectal cancer: focus on nivolumab. Expert Opin Biol Ther. 2019;19:1247–63.PubMedCrossRef
88.
go back to reference Swaika A, Hammond WA, Joseph RW. Current state of anti-PD-L1 and anti-PD-1 agents in cancer therapy. Mol Immunol. 2015;67:4–17.PubMedCrossRef Swaika A, Hammond WA, Joseph RW. Current state of anti-PD-L1 and anti-PD-1 agents in cancer therapy. Mol Immunol. 2015;67:4–17.PubMedCrossRef
89.
go back to reference O’Neil BH, Wallmark JM, Lorente D, Elez E, Raimbourg J, Gomez-Roca C, et al. Safety and antitumor activity of the anti–PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma. PLoS ONE. 2017;12:e0189848.PubMedPubMedCentralCrossRef O’Neil BH, Wallmark JM, Lorente D, Elez E, Raimbourg J, Gomez-Roca C, et al. Safety and antitumor activity of the anti–PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma. PLoS ONE. 2017;12:e0189848.PubMedPubMedCentralCrossRef
90.
go back to reference Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.PubMedPubMedCentralCrossRef Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.PubMedPubMedCentralCrossRef
91.
go back to reference Le DT, Kavan P, Kim TW, Burge ME, Van Cutsem E, Hara H, et al. KEYNOTE-164: pembrolizumab for patients with advanced microsatellite instability high (MSI-H) colorectal cancer. Am Soc Clin Oncol. 2018;36:3514.CrossRef Le DT, Kavan P, Kim TW, Burge ME, Van Cutsem E, Hara H, et al. KEYNOTE-164: pembrolizumab for patients with advanced microsatellite instability high (MSI-H) colorectal cancer. Am Soc Clin Oncol. 2018;36:3514.CrossRef
92.
go back to reference Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5:43–51.PubMedCrossRef Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5:43–51.PubMedCrossRef
93.
go back to reference Chaudhari PB. Nivolumab-Pearls of evidence. Indian J Med Paediatr Oncol Off J Indian Soc Med Paediatr Oncol. 2017;38:520.CrossRef Chaudhari PB. Nivolumab-Pearls of evidence. Indian J Med Paediatr Oncol Off J Indian Soc Med Paediatr Oncol. 2017;38:520.CrossRef
94.
go back to reference Yamamoto N, Nokihara H, Yamada Y, Shibata T, Tamura Y, Seki Y, et al. Phase I study of Nivolumab, an anti-PD-1 antibody, in patients with malignant solid tumors. Invest New Drugs. 2017;35:207–16.PubMedCrossRef Yamamoto N, Nokihara H, Yamada Y, Shibata T, Tamura Y, Seki Y, et al. Phase I study of Nivolumab, an anti-PD-1 antibody, in patients with malignant solid tumors. Invest New Drugs. 2017;35:207–16.PubMedCrossRef
95.
go back to reference Overman MJ, Lonardi S, Wong KYM, Lenz H-J, Gelsomino F, Aglietta M, et al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol. 2018;36:773–9.PubMedCrossRef Overman MJ, Lonardi S, Wong KYM, Lenz H-J, Gelsomino F, Aglietta M, et al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol. 2018;36:773–9.PubMedCrossRef
96.
go back to reference Inman BA, Longo TA, Ramalingam S, Harrison MR. Atezolizumab: a PD-L1–blocking antibody for bladder cancer. Clin Cancer Res. 2017;23:1886–90.PubMedCrossRef Inman BA, Longo TA, Ramalingam S, Harrison MR. Atezolizumab: a PD-L1–blocking antibody for bladder cancer. Clin Cancer Res. 2017;23:1886–90.PubMedCrossRef
97.
go back to reference Antoniotti C, Borelli B, Rossini D, Pietrantonio F, Morano F, Salvatore L, et al. AtezoTRIBE: a randomised phase II study of FOLFOXIRI plus bevacizumab alone or in combination with atezolizumab as initial therapy for patients with unresectable metastatic colorectal cancer. BMC Cancer. 2020;20:1–9.CrossRef Antoniotti C, Borelli B, Rossini D, Pietrantonio F, Morano F, Salvatore L, et al. AtezoTRIBE: a randomised phase II study of FOLFOXIRI plus bevacizumab alone or in combination with atezolizumab as initial therapy for patients with unresectable metastatic colorectal cancer. BMC Cancer. 2020;20:1–9.CrossRef
98.
go back to reference Eng C, Kim TW, Bendell J, Argilés G, Tebbutt NC, Di Bartolomeo M, et al. Atezolizumab with or without cobimetinib versus regorafenib in previously treated metastatic colorectal cancer (IMblaze370): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2019;20:849–61.PubMedCrossRef Eng C, Kim TW, Bendell J, Argilés G, Tebbutt NC, Di Bartolomeo M, et al. Atezolizumab with or without cobimetinib versus regorafenib in previously treated metastatic colorectal cancer (IMblaze370): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2019;20:849–61.PubMedCrossRef
99.
go back to reference Zhao Y, Yang W, Huang Y, Cui R, Li X, Li B. Evolving roles for targeting CTLA-4 in cancer immunotherapy. Cell Physiol Biochem. 2018;47:721–34.PubMedCrossRef Zhao Y, Yang W, Huang Y, Cui R, Li X, Li B. Evolving roles for targeting CTLA-4 in cancer immunotherapy. Cell Physiol Biochem. 2018;47:721–34.PubMedCrossRef
100.
go back to reference Kooshkaki O, Derakhshani A, Hosseinkhani N, Torabi M, Safaei S, Brunetti O, et al. Combination of ipilimumab and nivolumab in cancers: from clinical practice to ongoing clinical trials. Int J Mol Sci. 2020;21:4427.PubMedCentralCrossRef Kooshkaki O, Derakhshani A, Hosseinkhani N, Torabi M, Safaei S, Brunetti O, et al. Combination of ipilimumab and nivolumab in cancers: from clinical practice to ongoing clinical trials. Int J Mol Sci. 2020;21:4427.PubMedCentralCrossRef
101.
go back to reference Morse MA, Overman MJ, Hartman L, Khoukaz T, Brutcher E, Lenz H, et al. Safety of nivolumab plus low-dose ipilimumab in previously treated microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer. Oncologist. 2019;24:1453.PubMedPubMedCentralCrossRef Morse MA, Overman MJ, Hartman L, Khoukaz T, Brutcher E, Lenz H, et al. Safety of nivolumab plus low-dose ipilimumab in previously treated microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer. Oncologist. 2019;24:1453.PubMedPubMedCentralCrossRef
102.
go back to reference Ribas A, Hanson DC, Noe DA, Millham R, Guyot DJ, Bernstein SH, et al. Tremelimumab (CP-675,206), a cytotoxic T lymphocyte–associated antigen 4 blocking monoclonal antibody in clinical development for patients with cancer. Oncologist. 2007;12:873–83.PubMedCrossRef Ribas A, Hanson DC, Noe DA, Millham R, Guyot DJ, Bernstein SH, et al. Tremelimumab (CP-675,206), a cytotoxic T lymphocyte–associated antigen 4 blocking monoclonal antibody in clinical development for patients with cancer. Oncologist. 2007;12:873–83.PubMedCrossRef
103.
go back to reference Chung KY, Gore I, Fong L, Venook A, Beck SB, Dorazio P, et al. Phase II study of the anti-cytotoxic T-lymphocyte–associated antigen 4 monoclonal antibody, tremelimumab, in patients with refractory metastatic colorectal cancer. J Clin Oncol. 2010;28:3485–90.CrossRefPubMed Chung KY, Gore I, Fong L, Venook A, Beck SB, Dorazio P, et al. Phase II study of the anti-cytotoxic T-lymphocyte–associated antigen 4 monoclonal antibody, tremelimumab, in patients with refractory metastatic colorectal cancer. J Clin Oncol. 2010;28:3485–90.CrossRefPubMed
104.
go back to reference Rocha Lima CM, Bayraktar S, Flores AM, MacIntyre J, Montero A, Baranda JC, et al. Phase Ib study of drozitumab combined with first-line mFOLFOX6 plus bevacizumab in patients with metastatic colorectal cancer. Cancer Invest. 2012;30:727–31.PubMedCrossRef Rocha Lima CM, Bayraktar S, Flores AM, MacIntyre J, Montero A, Baranda JC, et al. Phase Ib study of drozitumab combined with first-line mFOLFOX6 plus bevacizumab in patients with metastatic colorectal cancer. Cancer Invest. 2012;30:727–31.PubMedCrossRef
105.
go back to reference Karp DD, Paz-Ares LG, Novello S, Haluska P, Garland L, Cardenal F, et al. Phase II study of the anti–insulin-like growth factor type 1 receptor antibody CP-751,871 in combination with paclitaxel and carboplatin in previously untreated, locally advanced, or metastatic non–small-cell lung cancer. J Clin Oncol. 2009;27:2516–22.PubMedCrossRef Karp DD, Paz-Ares LG, Novello S, Haluska P, Garland L, Cardenal F, et al. Phase II study of the anti–insulin-like growth factor type 1 receptor antibody CP-751,871 in combination with paclitaxel and carboplatin in previously untreated, locally advanced, or metastatic non–small-cell lung cancer. J Clin Oncol. 2009;27:2516–22.PubMedCrossRef
106.
go back to reference Françoso A, Simioni PU. Immunotherapy for the treatment of colorectal tumors: focus on approved and in-clinical-trial monoclonal antibodies. Drug Des Devel Ther. 2017;11:177.PubMedPubMedCentralCrossRef Françoso A, Simioni PU. Immunotherapy for the treatment of colorectal tumors: focus on approved and in-clinical-trial monoclonal antibodies. Drug Des Devel Ther. 2017;11:177.PubMedPubMedCentralCrossRef
107.
go back to reference Le DT, Uram JN, Wang H, Bartlett B, Kemberling H, Eyring A, et al. Programmed death-1 blockade in mismatch repair deficient colorectal cancer. J Clin Oncol. 2016;34:103.CrossRef Le DT, Uram JN, Wang H, Bartlett B, Kemberling H, Eyring A, et al. Programmed death-1 blockade in mismatch repair deficient colorectal cancer. J Clin Oncol. 2016;34:103.CrossRef
108.
go back to reference Mathijssen RHJ, De Jong FA, Loos WJ, Van Der Bol JM, Verweij J, Sparreboom A. Flat-fixed dosing versus body surface area-based dosing of anticancer drugs in adults: does it make a difference? Oncologist. 2007;12:913.PubMedCrossRef Mathijssen RHJ, De Jong FA, Loos WJ, Van Der Bol JM, Verweij J, Sparreboom A. Flat-fixed dosing versus body surface area-based dosing of anticancer drugs in adults: does it make a difference? Oncologist. 2007;12:913.PubMedCrossRef
109.
go back to reference Kreisl TN, Zhang W, Odia Y, Shih JH, Butman JA, Hammoud D, et al. A phase II trial of single-agent bevacizumab in patients with recurrent anaplastic glioma. Neuro Oncol. 2011;13:1143–50.PubMedPubMedCentralCrossRef Kreisl TN, Zhang W, Odia Y, Shih JH, Butman JA, Hammoud D, et al. A phase II trial of single-agent bevacizumab in patients with recurrent anaplastic glioma. Neuro Oncol. 2011;13:1143–50.PubMedPubMedCentralCrossRef
110.
go back to reference Lafayette RA, McCall B, Li N, Chu L, Werner P, Das A, et al. Incidence and relevance of proteinuria in bevacizumab-treated patients: pooled analysis from randomized controlled trials. Am J Nephrol. 2014;40:75–83.PubMedCrossRef Lafayette RA, McCall B, Li N, Chu L, Werner P, Das A, et al. Incidence and relevance of proteinuria in bevacizumab-treated patients: pooled analysis from randomized controlled trials. Am J Nephrol. 2014;40:75–83.PubMedCrossRef
111.
go back to reference Almagro JC, Daniels-Wells TR, Perez-Tapia SM, Penichet ML. Progress and challenges in the design and clinical development of antibodies for cancer therapy. Front Immunol. 2018;8:1751.PubMedPubMedCentralCrossRef Almagro JC, Daniels-Wells TR, Perez-Tapia SM, Penichet ML. Progress and challenges in the design and clinical development of antibodies for cancer therapy. Front Immunol. 2018;8:1751.PubMedPubMedCentralCrossRef
112.
go back to reference Ramos-de-la-Peña AM, González-Valdez J, Aguilar O. Protein A chromatography: challenges and progress in the purification of monoclonal antibodies. J Sep Sci. 2019;42:1816–27.PubMedCrossRef Ramos-de-la-Peña AM, González-Valdez J, Aguilar O. Protein A chromatography: challenges and progress in the purification of monoclonal antibodies. J Sep Sci. 2019;42:1816–27.PubMedCrossRef
113.
go back to reference Ulmer N, Vogg S, Müller-Späth T, Morbidelli M. Purification of human monoclonal antibodies and their fragments. In: Steinitz M, editor. Human monoclonal antibodies. Methods in Molecular Biology; vol. 1904, 2019. pp. 63–88. Ulmer N, Vogg S, Müller-Späth T, Morbidelli M. Purification of human monoclonal antibodies and their fragments. In: Steinitz M, editor. Human monoclonal antibodies. Methods in Molecular Biology; vol. 1904, 2019. pp. 63–88.
114.
go back to reference Arosio P, Barolo G, Müller-Späth T, Wu H, Morbidelli M. Aggregation stability of a monoclonal antibody during downstream processing. Pharm Res. 2011;28:1884–94.PubMedCrossRef Arosio P, Barolo G, Müller-Späth T, Wu H, Morbidelli M. Aggregation stability of a monoclonal antibody during downstream processing. Pharm Res. 2011;28:1884–94.PubMedCrossRef
115.
go back to reference Sifniotis V, Cruz E, Eroglu B, Kayser V. Current advancements in addressing key challenges of therapeutic antibody design, manufacture, and formulation. Antibodies. 2019;8:36.PubMedCentralCrossRef Sifniotis V, Cruz E, Eroglu B, Kayser V. Current advancements in addressing key challenges of therapeutic antibody design, manufacture, and formulation. Antibodies. 2019;8:36.PubMedCentralCrossRef
116.
go back to reference Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science (80-). 1986;233:1318–21.CrossRef Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science (80-). 1986;233:1318–21.CrossRef
118.
go back to reference Hayes C. Cellular immunotherapies for cancer. Irish J Med Sci 2020;1–17. Hayes C. Cellular immunotherapies for cancer. Irish J Med Sci 2020;1–17.
119.
go back to reference Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–9.CrossRefPubMed Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–9.CrossRefPubMed
120.
go back to reference Comoli P, Chabannon C, Koehl U, Lanza F, Urbano-Ispizua A, Hudecek M, et al. Development of adaptive immune effector therapies in solid tumors. Ann Oncol. 2019;30:1740–50.PubMedCrossRef Comoli P, Chabannon C, Koehl U, Lanza F, Urbano-Ispizua A, Hudecek M, et al. Development of adaptive immune effector therapies in solid tumors. Ann Oncol. 2019;30:1740–50.PubMedCrossRef
123.
go back to reference Su X, Vale R. Mechanisms of chimeric antigen receptor (CAR) signaling during T cell activation. Biophys J. 2018;114:107a–8a.CrossRef Su X, Vale R. Mechanisms of chimeric antigen receptor (CAR) signaling during T cell activation. Biophys J. 2018;114:107a–8a.CrossRef
124.
go back to reference Smith AJ, Oertle J, Warren D, Prato D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: summary and perspective. J Cell Immunother. 2016;2:59–68.CrossRef Smith AJ, Oertle J, Warren D, Prato D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: summary and perspective. J Cell Immunother. 2016;2:59–68.CrossRef
125.
go back to reference Subklewe M, von Bergwelt-Baildon M, Humpe A. Chimeric antigen receptor T cells: a race to revolutionize cancer therapy. Transfus Med Hemotherapy. 2019;46:15–24.CrossRef Subklewe M, von Bergwelt-Baildon M, Humpe A. Chimeric antigen receptor T cells: a race to revolutionize cancer therapy. Transfus Med Hemotherapy. 2019;46:15–24.CrossRef
126.
go back to reference Huang R, Li X, He Y, Zhu W, Gao L, Liu Y, et al. Recent advances in CAR-T cell engineering. J Hematol Oncol. 2020;13:1–19.CrossRef Huang R, Li X, He Y, Zhu W, Gao L, Liu Y, et al. Recent advances in CAR-T cell engineering. J Hematol Oncol. 2020;13:1–19.CrossRef
127.
go back to reference Tokarew N, Ogonek J, Endres S, von Bergwelt-Baildon M, Kobold S. Teaching an old dog new tricks: next-generation CAR T cells. Br J Cancer. 2019;120:26–37.PubMedCrossRef Tokarew N, Ogonek J, Endres S, von Bergwelt-Baildon M, Kobold S. Teaching an old dog new tricks: next-generation CAR T cells. Br J Cancer. 2019;120:26–37.PubMedCrossRef
128.
go back to reference Rodrigues JG, Balmaña M, Macedo JA, Poças J, Fernandes Â, De-Freitas-Junior JCM, et al. Glycosylation in cancer: Selected roles in tumour progression, immune modulation and metastasis. Cell Immunol. 2018;333:46–57.PubMedCrossRef Rodrigues JG, Balmaña M, Macedo JA, Poças J, Fernandes Â, De-Freitas-Junior JCM, et al. Glycosylation in cancer: Selected roles in tumour progression, immune modulation and metastasis. Cell Immunol. 2018;333:46–57.PubMedCrossRef
129.
go back to reference Tong G, Xu W, Zhang G, Liu J, Zheng Z, Chen Y, et al. The role of tissue and serum carcinoembryonic antigen in stages I to III of colorectal cancer—a retrospective cohort study. Cancer Med. 2018;7:5327–38.PubMedPubMedCentralCrossRef Tong G, Xu W, Zhang G, Liu J, Zheng Z, Chen Y, et al. The role of tissue and serum carcinoembryonic antigen in stages I to III of colorectal cancer—a retrospective cohort study. Cancer Med. 2018;7:5327–38.PubMedPubMedCentralCrossRef
130.
go back to reference Chi X, Yang P, Zhang E, Gu J, Xu H, Li M, et al. Significantly increased anti-tumor activity of carcinoembryonic antigen-specific chimeric antigen receptor T cells in combination with recombinant human IL-12. Cancer Med. 2019;8:4753–65.PubMedPubMedCentralCrossRef Chi X, Yang P, Zhang E, Gu J, Xu H, Li M, et al. Significantly increased anti-tumor activity of carcinoembryonic antigen-specific chimeric antigen receptor T cells in combination with recombinant human IL-12. Cancer Med. 2019;8:4753–65.PubMedPubMedCentralCrossRef
131.
go back to reference Blat D, Zigmond E, Alteber Z, Waks T, Eshhar Z. Suppression of murine colitis and its associated cancer by carcinoembryonic antigen-specific regulatory T cells. Mol Ther. 2014;22:1018–28.PubMedPubMedCentralCrossRef Blat D, Zigmond E, Alteber Z, Waks T, Eshhar Z. Suppression of murine colitis and its associated cancer by carcinoembryonic antigen-specific regulatory T cells. Mol Ther. 2014;22:1018–28.PubMedPubMedCentralCrossRef
132.
go back to reference Zhang C, Wang Z, Yang Z, Wang M, Li S, Li Y, et al. Phase I escalating-dose trial of CAR-T therapy targeting CEA+ metastatic colorectal cancers. Mol Ther. 2017;25:1248–58.PubMedPubMedCentralCrossRef Zhang C, Wang Z, Yang Z, Wang M, Li S, Li Y, et al. Phase I escalating-dose trial of CAR-T therapy targeting CEA+ metastatic colorectal cancers. Mol Ther. 2017;25:1248–58.PubMedPubMedCentralCrossRef
133.
go back to reference Valentino MA, Lin JE, Snook AE, Li P, Kim GW, Marszalowicz G, et al. A uroguanylin-GUCY2C endocrine axis regulates feeding in mice. J Clin Invest. 2011;121:3578–88.PubMedPubMedCentralCrossRef Valentino MA, Lin JE, Snook AE, Li P, Kim GW, Marszalowicz G, et al. A uroguanylin-GUCY2C endocrine axis regulates feeding in mice. J Clin Invest. 2011;121:3578–88.PubMedPubMedCentralCrossRef
134.
go back to reference Aka AA, Rappaport JA, Pattison AM, Sato T, Snook AE, Waldman SA. Guanylate cyclase C as a target for prevention, detection, and therapy in colorectal cancer. Expert Rev Clin Pharmacol. 2017;10:549–57.PubMedPubMedCentralCrossRef Aka AA, Rappaport JA, Pattison AM, Sato T, Snook AE, Waldman SA. Guanylate cyclase C as a target for prevention, detection, and therapy in colorectal cancer. Expert Rev Clin Pharmacol. 2017;10:549–57.PubMedPubMedCentralCrossRef
135.
go back to reference Frick GS, Pitari GM, Weinberg DS, Hyslop T, Schulz S, Waldman SA. Guanylyl cyclase C: a molecular marker for staging and postoperative surveillance of patients with colorectal cancer. Expert Rev Mol Diagn. 2005;5:701–13.PubMedCrossRef Frick GS, Pitari GM, Weinberg DS, Hyslop T, Schulz S, Waldman SA. Guanylyl cyclase C: a molecular marker for staging and postoperative surveillance of patients with colorectal cancer. Expert Rev Mol Diagn. 2005;5:701–13.PubMedCrossRef
136.
go back to reference Carrithers SL, Barber MT, Biswas S, Parkinson SJ, Park PK, Goldstein SD, et al. Guanylyl cyclase C is a selective marker for metastatic colorectal tumors in human extraintestinal tissues. Proc Natl Acad Sci. 1996;93:14827–32.PubMedCrossRefPubMedCentral Carrithers SL, Barber MT, Biswas S, Parkinson SJ, Park PK, Goldstein SD, et al. Guanylyl cyclase C is a selective marker for metastatic colorectal tumors in human extraintestinal tissues. Proc Natl Acad Sci. 1996;93:14827–32.PubMedCrossRefPubMedCentral
137.
go back to reference Magee MS, Kraft CL, Abraham TS, Baybutt TR, Marszalowicz GP, Li P, et al. GUCY2C-directed CAR-T cells oppose colorectal cancer metastases without autoimmunity. Oncoimmunology. 2016;5:e1227897.PubMedPubMedCentralCrossRef Magee MS, Kraft CL, Abraham TS, Baybutt TR, Marszalowicz GP, Li P, et al. GUCY2C-directed CAR-T cells oppose colorectal cancer metastases without autoimmunity. Oncoimmunology. 2016;5:e1227897.PubMedPubMedCentralCrossRef
138.
go back to reference Magee MS, Abraham TS, Baybutt TR, Flickinger JC, Ridge NA, Marszalowicz GP, et al. Human GUCY2C-targeted chimeric antigen receptor (CAR)-expressing T cells eliminate colorectal cancer metastases. Cancer Immunol Res. 2018;6:509–16.PubMedPubMedCentralCrossRef Magee MS, Abraham TS, Baybutt TR, Flickinger JC, Ridge NA, Marszalowicz GP, et al. Human GUCY2C-targeted chimeric antigen receptor (CAR)-expressing T cells eliminate colorectal cancer metastases. Cancer Immunol Res. 2018;6:509–16.PubMedPubMedCentralCrossRef
140.
141.
go back to reference Antonangeli F, Soriani A, Cerboni C, Sciumè G, Santoni A. How mucosal epithelia deal with stress: role of NKG2D/NKG2D ligands during inflammation. Front Immunol. 2017;8:1583.PubMedPubMedCentralCrossRef Antonangeli F, Soriani A, Cerboni C, Sciumè G, Santoni A. How mucosal epithelia deal with stress: role of NKG2D/NKG2D ligands during inflammation. Front Immunol. 2017;8:1583.PubMedPubMedCentralCrossRef
142.
go back to reference Deng X, Gao F, Li N, Li Q, Zhou Y, Yang T, et al. Antitumor activity of NKG2D CAR-T cells against human colorectal cancer cells in vitro and in vivo. Am J Cancer Res. 2019;9:945.PubMedPubMedCentral Deng X, Gao F, Li N, Li Q, Zhou Y, Yang T, et al. Antitumor activity of NKG2D CAR-T cells against human colorectal cancer cells in vitro and in vivo. Am J Cancer Res. 2019;9:945.PubMedPubMedCentral
143.
go back to reference Xiao L, Cen D, Gan H, Sun Y, Huang N, Xiong H, et al. Adoptive transfer of NKG2D CAR mRNA-engineered natural killer cells in colorectal cancer patients. Mol Ther. 2019;27:1114–25.PubMedPubMedCentralCrossRef Xiao L, Cen D, Gan H, Sun Y, Huang N, Xiong H, et al. Adoptive transfer of NKG2D CAR mRNA-engineered natural killer cells in colorectal cancer patients. Mol Ther. 2019;27:1114–25.PubMedPubMedCentralCrossRef
144.
go back to reference Zhang T, Sentman CL. Mouse tumor vasculature expresses NKG2D ligands and can be targeted by chimeric NKG2D-modified T cells. J Immunol. 2013;190:2455–63.PubMedCrossRef Zhang T, Sentman CL. Mouse tumor vasculature expresses NKG2D ligands and can be targeted by chimeric NKG2D-modified T cells. J Immunol. 2013;190:2455–63.PubMedCrossRef
145.
go back to reference Zhang B-L, Li D, Gong Y-L, Huang Y, Qin D-Y, Jiang L, et al. Preclinical evaluation of chimeric antigen receptor-modified T cells specific to epithelial cell adhesion molecule for treating colorectal cancer. Hum Gene Ther. 2019;30:402–12.PubMedCrossRef Zhang B-L, Li D, Gong Y-L, Huang Y, Qin D-Y, Jiang L, et al. Preclinical evaluation of chimeric antigen receptor-modified T cells specific to epithelial cell adhesion molecule for treating colorectal cancer. Hum Gene Ther. 2019;30:402–12.PubMedCrossRef
146.
go back to reference Thor A, Ohuchi N, Szpak CA, Johnston WW, Schlom J. Distribution of oncofetal antigen tumor-associated glycoprotein-72 defined by monoclonal antibody B72.3. Cancer Res. 1986;46:3118–24.PubMed Thor A, Ohuchi N, Szpak CA, Johnston WW, Schlom J. Distribution of oncofetal antigen tumor-associated glycoprotein-72 defined by monoclonal antibody B72.3. Cancer Res. 1986;46:3118–24.PubMed
147.
go back to reference Hege KM, Bergsland EK, Fisher GA, Nemunaitis JJ, Warren RS, McArthur JG, et al. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J Immunother Cancer. 2017;5:22.PubMedPubMedCentralCrossRef Hege KM, Bergsland EK, Fisher GA, Nemunaitis JJ, Warren RS, McArthur JG, et al. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J Immunother Cancer. 2017;5:22.PubMedPubMedCentralCrossRef
148.
go back to reference Wang L-CS, Lo A, Scholler J, Sun J, Majumdar RS, Kapoor V, et al. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res. 2014;2:154–66.PubMedCrossRef Wang L-CS, Lo A, Scholler J, Sun J, Majumdar RS, Kapoor V, et al. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res. 2014;2:154–66.PubMedCrossRef
149.
go back to reference Caruana I, Savoldo B, Hoyos V, Weber G, Liu H, Kim ES, et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat Med. 2015;21:524–9.PubMedPubMedCentralCrossRef Caruana I, Savoldo B, Hoyos V, Weber G, Liu H, Kim ES, et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat Med. 2015;21:524–9.PubMedPubMedCentralCrossRef
150.
go back to reference Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–51.PubMedPubMedCentralCrossRef Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–51.PubMedPubMedCentralCrossRef
151.
go back to reference Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DAN, Feldman SA, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011;19:620–6.CrossRefPubMed Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DAN, Feldman SA, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011;19:620–6.CrossRefPubMed
152.
go back to reference Xia A-L, Wang X-C, Lu Y-J, Lu X-J, Sun B. Chimeric-antigen receptor T (CAR-T) cell therapy for solid tumors: challenges and opportunities. Oncotarget. 2017;8:90521.PubMedPubMedCentralCrossRef Xia A-L, Wang X-C, Lu Y-J, Lu X-J, Sun B. Chimeric-antigen receptor T (CAR-T) cell therapy for solid tumors: challenges and opportunities. Oncotarget. 2017;8:90521.PubMedPubMedCentralCrossRef
153.
go back to reference John LB, Devaud C, Duong CPM, Yong CS, Beavis PA, Haynes NM, et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin cancer Res. 2013;19:5636–46.PubMedCrossRef John LB, Devaud C, Duong CPM, Yong CS, Beavis PA, Haynes NM, et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin cancer Res. 2013;19:5636–46.PubMedCrossRef
Metadata
Title
Monoclonal antibodies and chimeric antigen receptor (CAR) T cells in the treatment of colorectal cancer
Authors
Ke-Tao Jin
Bo Chen
Yu-Yao Liu
H uan-Rong Lan
Jie-Ping Yan
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2021
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-01763-9

Other articles of this Issue 1/2021

Cancer Cell International 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine