HER2 in Metastatic Colorectal Cancer: Comparison
Please note this is a comparison between Version 2 by Rita Xu and Version 1 by Mariia Ivanova.

HER2 is an emerging biomarker in colorectal cancer (CRC). This oncogene plays an essential role in regulating cell proliferation, differentiation, migration, and, more in general, tumorigenesis and tumor progression. The most frequent types of HER2 alterations in CRC include gene amplification and missense mutations in 7–8% of CRC, often being mirrored by HER2 protein overexpression, representing founder events in solid tumors, including CRC.

  • HER2
  • colorectal cancer
  • pathology
  • biomarkers

1. Introduction

Human epidermal growth factor receptor 2 (HER2) is a proto-oncogene encoding for a transmembrane glycoprotein with a tyrosine kinase activity, a member of the ErbB receptor tyrosine kinases family, and one of the epidermal growth factor receptors (EGFRs) [1,2][1][2]. HER2 plays an essential role in normal biological and oncogenic processes, regulating cell proliferation, differentiation, and migration via numerous signaling pathways, such as mitogen-activated protein kinase/extracellular signal-regulated kinases (MAPK/ERK) and phosphoinositide 3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) [1,2][1][2]. The schematic overview of HER2 pathways is represented in Supplementary Figure S1. Alterations of HER2 include gene amplification and missense mutations and often lead to protein overexpression [3]. These types of molecular aberrations are considered founder events in tumorigenesis and tumor progression because of uncontrolled cell proliferation, inhibition of apoptosis, and migration [1,4,5,6][1][4][5][6].
In the current clinical practice, HER2 status is tested in breast and gastroesophageal cancers to select patients eligible for anti-HER2 treatment [5]. However, alterations in HER2 have been documented in a plethora of other solid tumors, including colorectal cancer (CRC) [2,3,4,5,6,7][2][3][4][5][6][7]. This tumor type is the second leading cause of cancer-associated deaths worldwide in both sexes [8]. In CRC, the frequency of HER2 overexpression is 5–6% with somatic HER2 gene alterations, including amplifications, reported in ~7% of patients [4,6,9,10,11][4][6][9][10][11]. HER2 mutations in colonic epithelial cells have been shown to be indicative of HER2 signaling pathway activations, promote independent cell growth, and potentially acquire resistance to EGFR-targeted therapies, subjecting patients to a worse prognosis [2,4,6,9,10,11,12][2][4][6][9][10][11][12]. There are currently no approved HER2-targeted therapy guidelines for CRC; however, several studies have shown that HER2 can be effectively targeted in metastatic CRC settings [2,12,13,14][2][12][13][14].

2. HER2 Targeting in Metastatic Colorectal Cancer

The administration of anti-HER2 drugs (e.g., trastuzumab, pertuzumab) is presently a standard of care in HER2-positive (i.e., score 3+ by immunohistochemistry (IHC) or 2+/in situ hybridization (ISH)-positive) breast and gastric cancer [15,16][15][16]. For CRC, HER2 first emerged as a negative predictive biomarker. Amplification or overexpression of HER2 was associated with a lack of response to anti-EGFR treatment [17]. The novel antibody–drug conjugate trastuzumab deruxtecan (T-DXd) has been approved by the United States Food and Drug Administration (FDA) for HER2-low (i.e., score 1+ or 2+/ISH-negative) breast cancers [18], and it is currently under investigation in histology-agnostic settings in the DESTINY-PanTumor02 study (NCT04482309). The first clinical trials using trastuzumab in mCRC evaluated the combination of this monoclonal antibody with chemotherapy. Clark et al. assessed the association of FOLFOX + trastuzumab in the second- or third-line treatment of HER2-positive mCRC. A total of 5 patients out of 21 (24%) had a partial (PR) or complete response (CR), with a median duration of response of 4.5 months [19]. Another phase II study assessed the combination of trastuzumab and irinotecan in HER2-positive mCRC pretreated with one line of therapy. Objective responses were recorded in five patients (71%), and these were maintained for at least 6 months [20]. Subsequent studies evaluated different strategies of an HER2 dual blockade with significant results. HERACLES-A tested the combination of trastuzumab and lapatinib in patients with KRAS exon-2 wild-type (WT) mCRC refractory to standard treatment and with HER2 amplification and/or overexpression. A total of 914 patients were screened, of whom 48 (5%) had HER2-positive diseases [14,21][14][21]. The long-term clinical results at a follow-up of 6.7 years reported an overall response rate (ORR) of 28% with one CR and eight PRs, a disease control rate (DCR) of 69%, median progression-free survival (mPFS) of 4.7 months, and median overall survival (OS) of 10 months for 32 treated patients. Two of them (6%) reported a grade 3 decrease in left ventricular ejection fraction, while fatigue was registered in five cases (16%) [22]. Differently, HERACLES-B tested the combination of pertuzumab and the antibody–drug conjugate trastuzumab emtansine (TDM-1) in RAS and BRAF WT and HER2-positive mCRC refractory to standard therapies. The primary endpoint was not reached, with ORR below the expected rate ≥30% (9.7%). Stable disease (SD) was seen in 21 patients (67.7%), and the DCR rate was 77.4%. Treatment was well-tolerated, with two patients suffering from grade (G) 3 thrombocytopenia [23]. A median PFS of 4.2 months was similar to the HERACLES-A study, and patients with a HER2 3+ score at immunohistochemistry (IHC) had a significantly higher mPFS compared to HER2 IHC and fluorescent in situ hybridization (FISH)-amplified tumors (HR: 0.20; 95% confidence interval (CI): 0.07–0.56; p = 0.0008) [17]. The MyPathway phase II trial enrolled 57 patients with HER2-amplified mCRC receiving a combination of trastuzumab and pertuzumab. One patient had a CR while seventeen (30%) benefited from a PR. On the whole, 18 patients (32%) achieved an objective response, and, in 4 cases, it was longer than 12 months. Treatment was well-tolerated, with the most common adverse events being G 1 or 2 diarrhea, fatigue, and nausea. Patients harboring a KRAS mutation had a significantly shorter PFS and OS compared to the KRAS WT population (PFS: 1.4 months; 95% CI, 1.2–2.8 months versus 5.3 months; 95% CI: 2.7–6.1 months for mutated and WT, respectively; OS: 8.5 months; 95% CI: 3.9–not estimable versus 14.0 months; 95% CI: 8.0–not estimable for mutated and WT, respectively) [24]. Two other phase II studies (TRIUMPH and TAPUR) evaluated the combination of trastuzumab + pertuzumab. In the first study, 19 patients with RAS wt mCRC and HER2 amplification in tissue samples achieved an ORR of 35% with a complete response and five partial responses. Interestingly, the TRIUMPH trial evaluated HER2 status on circulating tumor DNA (ctDNA). Similarly, to the patients with HER2 amplification detected on tissue, 15 patients with ctDNA positivity for HER2 amplification had an ORR of 33% with one CR and four PRs. With a median follow-up of 5.4 months, mPFS was 4 months [25]. In the TAPUR basket trial, a cohort of 28 patients with HER2 amplified mCRC was treated; ORR was 14% and DCR for at least 16 weeks was 50% with an mPFS of 3.8 months [26]. A new anti-HER2 agent, trastuzumab deruxtecan, was tested in the DESTINY-CRC01 phase II trial. This antibody–drug conjugate of a humanized anti-HER2 antibody with a topoisomerase I inhibitor was tested in HER2-positive RAS-BRAF WT mCRC progressed on two or more lines of treatment, with the possibility to include patients pretreated with different anti-HER2 agents. A total of 78 patients were enrolled with 53 placed in cohort A (HER2 IHC 3+ or 2+ and positive in situ hybridization), a total of 7 in cohort B (IHC 2+ and negative in situ hybridization), and 18 in cohort C (IHC 1+). After a median follow-up of 27.1 weeks, the ORR in group A was 45.3% (95% CI, 31.6–59.6), and patients pretreated with anti-HER2 agents obtained a high ORR of 43%, as well. Differently, no responses were seen in groups B and C of treatment [12]. At a longer median follow-up of 62.4 weeks with 86 patients treated, the ORR of the group A was confirmed (45.3%). Moreover, DCR was 83%, mPFS 6.9 months, and mOS 15.5 months. Pulmonary toxicity in terms of interstitial lung disease and pneumonitis was recorded in eight patients (9.3%). Two patients died because of grade 5 lung toxicity. Interestingly, trastuzumab deruxtecan was also effective in the group of patients with RAS mutation-positive ctDNA [27]. In the ongoing MOUNTAINEER trial, trastuzumab is associated with tucatinib, a tyrosine kinase inhibitor (TKI) of the HER2 protein. Twenty-six patients with chemorefractory, RAS WT, and HER2-positive tumors have been treated so far with an ORR of 52.2%, twelve PRs, and six cases of SD. Patients experienced a prolonged median response of 10.4 months, an mPFS of 8.1 months (95% CI, 3.8–not estimable), and a median OS of 18.7 months (95% CI, 12.3–not estimable) [28]. The HER2 FUSCC-G trial is testing the combination of trastuzumab + pyrotinib, an irreversible dual pan-ErbB tyrosine kinase inhibitor (TKI) [29]. A cohort of 11 mCRC HER2-positive patients have received this combination so far, with a global ORR of 45.5% and 55.6% in RAS WT tumors. With a median follow-up of 17.7 months, mPFS was 7.8 months, while mOS 14.9 months. Patients harboring KRAS mutation had poorer outcomes compared to the KRAS WT group (PFS, 7.7 versus 9.9 months; p = 0.19; OS, 12.4 versus 20.6 months; p = 0.021) [30]. Differently, the combination of the anti-HER2 TKI neratinib and anti-EGFR agent cetuximab was not effective in terms of objective responses. A total of 16 patients with KRAS, NRAS, BRAF, and PI3KCA WT tumors were treated; 6 of them (44%) had SD with 5 harboring HER2 amplification at baseline. G 3adverse events, such as diarrhea, skin rash, and an increased level of transaminases, were registered in 67% of patients [31]. Table 1 lists the main trials with HER2-targeted therapies in mCRC that have been conducted so far.
Table 1. Main phase II studies with HER2-targeted therapies in refractory HER2-positive mCRC.
Legend: IHC: immunohistochemistry; mOS: median overall survival; mPFS: median progression-free survival; Nr.: number; ORR: overall response rate.
While anti-HER2 therapy in CRC is awaiting approval, many early trials continue, demonstrating promising results [32]. Supposing that HER2 may be immunogenic and leads to T-cell activation suggests it is targetable for immunotherapy [33]. Some studies demonstrate the induction of HER2 downregulation in HER2-positive cancer cells with the immune effector cells’ engagement, revealing a new function of immune cells in trastuzumab-mediated antitumor efficacy and probably representing a novel mechanism of action of trastuzumab, predicting active immune effector cells’ recruitment in the tumor microenvironment [34]. The acquisition of drug resistance to trastuzumab has been recently explained in HER2-positive gastric cancer by vessel destabilization and activation of the glycolytic pathway inducing 6-phosphofructo-2-kinase (PFKFB3). The inhibition of PFKFB3 in patient-derived xenograft models has significantly diminished tumor proliferation and promoted vessel normalization. It has also been found that PFKFB3 promotes the interleukin-8 coding gene CXCL8 by activating the PI3K/AKT/NFκB p65 pathway, which leads to the idea that PFKFB3 inhibition might be effective in overcoming trastuzumab resistance in HER2-positive cancers [35]. HER2-activating mutations are also known for their association with microsatellite instability-high tumors, which has been observed in CRC [36]. With the evolution of immunotherapy, after the US Food and Drug Administration (FDA) approval of pembrolizumab, it has become a practice-changing treatment option for unresectable or mCRC in patients with high microsatellite instability (MSI-H) or mismatched repair deficiency (dMMR) [37]. In the metastatic settings, however, only a small proportion of CRC patients responded to immune checkpoint therapy, despite positive results in some phase I trials, and all these tumors were MSI-H/dMMR and had a high tumor mutation burden [38]. While tumor mutational burden has been associated with the immune checkpoint response rate in other tumor types, such as melanoma and non-small-cell lung cancer, the underlying mechanism is still unknown, although it might be related to immune cells’ reactivity, increasing T-cell infiltration [38,39,40][38][39][40]. An ongoing recruiting four-part, phase 1/2 dose-escalation/expansion study is aiming to evaluate the effects of BDC-1001 (immune stimulating antibody conjugate (ISAC), consisting of an anti-HER2 monoclonal antibody conjugated to a TLR 7/8 dual agonist) in combination with/without PD1 inhibitor pembrolizumab in patients with progressive HER2-expressing solid tumors (NCT04278144). Current results demonstrate BDC-1001 to be well tolerated and clinically efficient also in patients previously treated with anti-HER2 therapy; however, the safety and efficacy of combining with a PD1 inhibitor is yet to be studied [6[6][41],41], although the data on PD-L1 expression in CRC with its regards to microsatellite instability remain controversial [38]. Some authors advocate that HER2-targeted therapies may favorably be combined with other emerging therapeutic strategies for advanced CRC, including immune checkpoint inhibitors, increasing the tailored therapeutic approach [42,43][42][43].

References

  1. Gaye, E.; Penel, N.; Lebellec, L. Novel treatment approaches for HER2 positive solid tumors (excluding breast cancer). Curr. Opin. Oncol. 2022, 34, 570–574.
  2. Roy-Chowdhuri, S.; Davies, K.D.; Ritterhouse, L.L.; Snow, A.N. ERBB2 (HER2) Alterations in Colorectal Cancer. J. Mol. Diagn. 2022.
  3. Wang, H.; Miao, J.; Wen, Y.; Xia, X.; Chen, Y.; Huang, M.; Chen, S.; Zhao, Z.; Zhang, Y.; Chen, C.; et al. Molecular Landscape of ERBB2 Alterations in 14,956 Solid Tumors. Pathol. Oncol. Res. POR 2022, 28, 1610360.
  4. Oh, D.-Y.; Bang, Y.-J. HER2-targeted therapies—A role beyond breast cancer. Nat. Rev. Clin. Oncol. 2020, 17, 33–48.
  5. Vranić, S.; Bešlija, S.; Gatalica, Z. Targeting HER2 expression in cancer: New drugs and new indications. Bosn. J. Basic Med. Sci. 2021, 21, 1–4.
  6. Seo, A.N.; Kwak, Y.; Kim, D.W.; Kang, S.B.; Choe, G.; Kim, W.H.; Lee, H.S. HER2 status in colorectal cancer: Its clinical significance and the relationship between HER2 gene amplification and expression. PLoS ONE 2014, 9, e98528.
  7. Fusco, N.; Bosari, S. HER2 aberrations and heterogeneity in cancers of the digestive system: Implications for pathologists and gastroenterologists. World J. Gastroenterol. 2016, 22, 7926–7937.
  8. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249.
  9. Kavuri, S.M.; Jain, N.; Galimi, F.; Cottino, F.; Leto, S.M.; Migliardi, G.; Searleman, A.C.; Shen, W.; Monsey, J.; Trusolino, L.; et al. HER2 activating mutations are targets for colorectal cancer treatment. Cancer Discov. 2015, 5, 832–841.
  10. El-Deiry, W.S.; Vijayvergia, N.; Xiu, J.; Scicchitano, A.; Lim, B.; Yee, N.S.; Harvey, H.A.; Gatalica, Z.; Reddy, S. Molecular profiling of 6892 colorectal cancer samples suggests different possible treatment options specific to metastatic sites. Cancer Biol. Ther. 2015, 16, 1726–1737.
  11. Afrăsânie, V.A.; Marinca, M.V.; Alexa-Stratulat, T.; Gafton, B.; Păduraru, M.; Adavidoaiei, A.M.; Miron, L.; Rusu, C. KRAS, NRAS, BRAF, HER2 and microsatellite instability in metastatic colorectal cancer—Practical implications for the clinician. Radiol. Oncol. 2019, 53, 265–274.
  12. Siena, S.; Di Bartolomeo, M.; Raghav, K.; Masuishi, T.; Loupakis, F.; Kawakami, H.; Yamaguchi, K.; Nishina, T.; Fakih, M.; Elez, E.; et al. Trastuzumab deruxtecan (DS-8201) in patients with HER2-expressing metastatic colorectal cancer (DESTINY-CRC01): A multicentre, open-label, phase 2 trial. Lancet Oncol. 2021, 22, 779–789.
  13. Dienstmann, R.; Ciner, A.; Hochster, H.S. Should next-generation sequencing testing be routinely used in metastatic colorectal cancer? Lancet Oncol. 2018, 19, 1434–1435.
  14. Sartore-Bianchi, A.; Trusolino, L.; Martino, C.; Bencardino, K.; Lonardi, S.; Bergamo, F.; Zagonel, V.; Leone, F.; Depetris, I.; Martinelli, E.; et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): A proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 738–746.
  15. Henry, N.L.; Somerfield, M.R.; Dayao, Z.; Elias, A.; Kalinsky, K.; McShane, L.M.; Moy, B.; Park, B.H.; Shanahan, K.M.; Sharma, P.; et al. Biomarkers for Systemic Therapy in Metastatic Breast Cancer: ASCO Guideline Update. J. Clin. Oncol. 2022, JCO2201063.
  16. Bartley, A.N.; Washington, M.K.; Colasacco, C.; Ventura, C.B.; Ismaila, N.; Benson, A.B.; Carrato, A.; Gulley, M.L.; Jain, D.; Kakar, S.; et al. HER2 Testing and Clinical Decision Making in Gastroesophageal Adenocarcinoma: Guideline from the College of American Pathologists, American Society for Clinical Pathology, and the American Society of Clinical Oncology. J. Clin. Oncol. 2017, 35, 446–464.
  17. Djaballah, S.A.; Daniel, F.; Milani, A.; Ricagno, G.; Lonardi, S. HER2 in colorectal cancer: The long and winding road from negative predictive factor to positive actionable target. Am. Soc. Clin. Oncol. Educ. Book 2022, 42, 219–232.
  18. Modi, S.; Jacot, W.; Yamashita, T.; Sohn, J.; Vidal, M.; Tokunaga, E.; Tsurutani, J.; Ueno, N.T.; Chae, Y.S.; Lee, K.S.; et al. Trastuzumab deruxtecan (T-DXd) versus treatment of physician’s choice (TPC) in patients (pts) with HER2-low unresectable and/or metastatic breast cancer (mBC): Results of DESTINY-Breast04, a randomized, phase 3 study. J. Clin. Oncol. 2022, 40, LBA3.
  19. Clark, J.W.; Niedzwiecki, D.; Hollis, D.M.R. Phase-II trial of 5-fluororuacil (5-FU), leucovorin (LV), oxaliplatin (Ox), and trastuzamab (T) for patients with metastatic colorectal cancer (CRC) refractory to initial therapy. Onkologie 2003, 26, 13–46.
  20. Ramanathan, R.K.; Hwang, J.J.; Zamboni, W.C.; Sinicrope, F.A.; Safran, H.; Wong, M.K.; Earle, M.; Brufsky, A.; Evans, T.; Troetschel, M.; et al. Low overexpression of HER-2/neu in advanced colorectal cancer limits the usefulness of trastuzumab (Herceptin) and irinotecan as therapy. A phase II trial. Cancer Investig. 2004, 22, 858–865.
  21. Siena, S.; Sartore-Bianchi, A.; Marsoni, S.; Hurwitz, H.I.; McCall, S.J.; Penault-Llorca, F.; Srock, S.; Bardelli, A.; Trusolino, L. Targeting the human epidermal growth factor receptor 2 (HER2) oncogene in colorectal cancer. Ann. Oncol. 2018, 29, 1108–1119.
  22. Tosi, F.; Sartore-Bianchi, A.; Lonardi, S.; Amatu, A.; Leone, F.; Ghezzi, S.; Martino, C.; Bencardino, K.; Bonazzina, E.; Bergamo, F.; et al. Long-term Clinical Outcome of Trastuzumab and Lapatinib for HER2-positive Metastatic Colorectal Cancer. Clin. Colorectal Cancer 2020, 19, 256–262.
  23. Sartore-Bianchi, A.; Lonardi, S.; Martino, C.; Fenocchio, E.; Tosi, F.; Ghezzi, S.; Leone, F.; Bergamo, F.; Zagonel, V.; Ciardiello, F.; et al. Pertuzumab and trastuzumab emtansine in patients with HER2-amplified metastatic colorectal cancer: The phase II HERACLES-B trial. ESMO Open 2020, 5, e000911.
  24. Meric-Bernstam, F.; Hurwitz, H.; Raghav, K.P.S.; McWilliams, R.R.; Fakih, M.; VanderWalde, A.; Swanton, C.; Kurzrock, R.; Burris, H.; Sweeney, C.; et al. Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): An updated report from a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 2019, 20, 518–530.
  25. Nakamura, Y.; Okamoto, W.; Kato, T.; Esaki, T.; Kato, K.; Komatsu, Y.; Yuki, S.; Masuishi, T.; Nishina, T.; Ebi, H.; et al. Circulating tumor DNA-guided treatment with pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer: A phase 2 trial. Nat. Med. 2021, 27, 1899–1903.
  26. Gupta, M.; Wang, B.; Carrothers, T.J.; LoRusso, P.M.; Chu, Y.W.; Shih, T.; Loecke, D.; Joshi, A.; Saad, O.; Yi, J.H.; et al. Effects of Trastuzumab Emtansine (T-DM1) on QT Interval and Safety of Pertuzumab Plus T-DM1 in Patients with Previously Treated Human Epidermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer. Clin. Pharmacol. Drug Dev. 2013, 2, 11–24.
  27. Yoshino, T.; Di Bartolomeo, M.; Raghav, K.; Masuishi, T.; Loupakis, F.; Kawakami, H.; Yamaguchi, K.; Nishina, T.; Wainberg, Z.; Elez, E.; et al. Trastuzumab deruxtecan (T-DXd; DS-8201) in patients (pts) with HER2-expressing metastatic colorectal cancer (mCRC): Final results from a phase 2, multicenter, open-label study (DESTINY-CRC01). In Proceedings of the 2021 ASCO Annual Meeting, Virtual, 4–8 June 2021.
  28. Strickler, J.H.; Zemla, T.; Ou, F.S.; Cercek, A.; Wu, C.; Sanchez, F.A.; Hubbard, J.; Jaszeeski, B.; Bandel, L.; Schweitzer, B.; et al. Trastuzumab and tucatinib for the treatment of HER2 amplified metastatic colorectal cancer (mCRC): Initial results from the MOUNTAINEER trial. In Proceedings of the ESMO Annual Meeting, Barcelona, Spain, 27 September–1 October 2019.
  29. Li, X.; Yang, C.; Wan, H.; Zhang, G.; Feng, J.; Zhang, L.; Chen, X.; Zhong, D.; Lou, L.; Tao, W.; et al. Discovery and development of pyrotinib: A novel irreversible EGFR/HER2 dual tyrosine kinase inhibitor with favorable safety profiles for the treatment of breast cancer. Eur. J. Pharm. Sci. 2017, 110, 51–61.
  30. Li, W.; Chang, J.; Xu, M.; Zhu, X. Anti-HER2 therapy with pyrotinib and trastuzumab in refractory HER2-positive metastatic colorectal cancer: A preliminary report from HER2-FUSCC-G study. In Proceedings of the 2022 ASCO Gastrointestinal Cancers Symposium, San Francisco, CA, USA, 20–22 January 2022.
  31. Jacobs, S.A.; Lee, J.J.; George, T.J.; Wade, J.L., III; Stella, P.J.; Wang, D.; Sama, A.R.; Piette, F.; Pogue-Geile, K.L.; Kim, R.S.; et al. Neratinib-Plus-Cetuximab in Quadruple-WT (KRAS, NRAS, BRAF, PIK3CA) Metastatic Colorectal Cancer Resistant to Cetuximab or Panitumumab: NSABP FC-7, A Phase Ib Study. Clin. Cancer Res. 2021, 27, 1612–1622.
  32. Bitar, L.; Zouein, J.; Haddad, F.G.; Eid, R.; Kourie, H.R. HER2 in metastatic colorectal cancer: A new to target to remember. Biomark Med. 2021, 15, 133–136.
  33. Nazemalhosseini Mojarad, E.; Kuppen, P.J. HER2 and immunotherapy using monoclonal antibodies in colorectal cancer. Immunotherapy 2013, 5, 1267–1269.
  34. Shi, Y.; Fan, X.; Meng, W.; Deng, H.; Zhang, N.; An, Z. Engagement of immune effector cells by trastuzumab induces HER2/ERBB2 downregulation in cancer cells through STAT1 activation. Breast Cancer Res. 2014, 16, R33.
  35. Yao, X.; He, Z.; Qin, C.; Zhang, P.; Sui, C.; Deng, X.; Fang, Y.; Li, G.; Shi, J. Inhibition of PFKFB3 in HER2-positive gastric cancer improves sensitivity to trastuzumab by inducing tumour vessel normalisation. Br. J. Cancer 2022, 127, 811–823.
  36. Qiu, M.Z.; He, C.Y.; Yang, X.H.; Yang, L.Q.; Lin, J.Z.; Zhou, D.L.; Long, Y.K.; Guan, W.L.; Jin, Y.; Li, Y.H.; et al. Relationship of HER2 Alteration and Microsatellite Instability Status in Colorectal Adenocarcinoma. Oncologist 2021, 26, e1161–e1170.
  37. Andre, T.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.; Smith, D.; Garcia-Carbonero, R.; Benavides, M.; Gibbs, P.; et al. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N. Engl. J. Med. 2020, 383, 2207–2218.
  38. Passardi, A.; Canale, M.; Valgiusti, M.; Ulivi, P. Immune Checkpoints as a Target for Colorectal Cancer Treatment. Int. J. Mol. Sci. 2017, 18, 1324.
  39. Xie, Y.H.; Chen, Y.X.; Fang, J.Y. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct. Target Ther. 2020, 5, 22.
  40. Sahin, I.H.; Akce, M.; Alese, O.; Shaib, W.; Lesinski, G.B.; El-Rayes, B.; Wu, C. Immune checkpoint inhibitors for the treatment of MSI-H/MMR-D colorectal cancer and a perspective on resistance mechanisms. Br. J. Cancer 2019, 121, 809–818.
  41. Sharma, M.; Carvajal, R.D.; Hanna, G.J.; Li, B.T.; Moore, K.N.; Pegram, M.D.; Rasco, D.W.; Spira, A.I.; Alonso, M.; Fang, L.; et al. Preliminary results from a phase 1/2 study of BDC-1001, a novel HER2 targeting TLR7/8 immune-stimulating antibody conjugate (ISAC), in patients (pts) with advanced HER2-expressing solid tumors. In Proceedings of the ASCO Annual Meeting, Virtual, 4–8 June 2021.
  42. Lefler, D.S.; Snook, A.E.; Bashir, B. Immune checkpoint inhibitors in luminal gastrointestinal malignancies: Going beyond MSI-H/dMMR, TMB and PD-L1. Immunotherapy 2022, 14, 885–902.
  43. Guarini, C.; Grassi, T.; Pezzicoli, G.; Porta, C. Beyond RAS and BRAF: HER2, a New Actionable Oncotarget in Advanced Colorectal Cancer. Int. J. Mol. Sci. 2021, 22, 6813.
More
Video Production Service