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]. 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.