Abstract
Antibody-drug conjugates (ADCs) combine the high specificity of monoclonal antibodies with the high anti-tumor activity of small molecular cytotoxic payloads. The anti-tumor activity of ADCs is mainly achieved by the direct blocking of the receptor by monoclonal antibodies, direct action and bystander effect of cytotoxic drugs, and antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity. ADCs have been used in adjuvant therapy and rescue treatment of human epidermal receptor 2 (HER2)-positive breast cancer, greatly improving the prognosis of breast cancer patients. Several ongoing clinical trials of ADC for breast cancer and other solid tumors proved the potential of ADCs will provide more promising treatment options for patients with malignant tumors. This review introduces the mechanism and latest clinical progress of ADC drugs approved for HER2-positive breast cancer to guide clinical practice and conduct research.
Keywords: Antibody-drug conjugate, HER2-positive, Breast cancer, T-DM1, DS-8201
An antibody-drug conjugate (ADC) is composed of a monoclonal antibody linked to a small molecular cytotoxic payload through a cleavable or non-cleavable linker. With the approval of trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (DS-8201a),[1,2] ADC has created a new era of targeted therapy for breast cancer. However, the development process of ADCs is relatively tortuous. It took over 100 years from the proposal of the ADC design concept to the approval of the first drug for marketing.[3] This article summarizes the mechanism of ADC in human epidermal receptor 2 (HER2)-positive breast cancer therapy, as well as the latest clinical trial progress and common adverse effects (AEs) of approved ADCs.
Composition and Mechanism of ADCs
ADC combines the targeting specificity of monoclonal antibodies with the high toxicity of cytotoxic drugs. However, the anti-tumor mechanism of ADCs is complex, and any component may affect their efficacy and AEs. Several reviews have been published on the components and mechanisms of action of ADC.[4,5] This part of the content focuses on the properties of different components and pharmacokinetic parameters of T-DM1 and DS-8201a.
Target antigen and monoclonal antibody
The targeted antigen should be highly expressed in tumor tissues, but not expressed or expressed at low levels in normal tissues to facilitate highly specific binding with monoclonal antibodies and deliver an effective active dose of the payload to the tumor tissues. Currently, the approved agents for breast cancer target HER2 and human trophoblast cell-surface antigen 2 (TROP2), respectively. Among them, HER2 is overexpressed and amplified in 15% to 20% of breast cancers,[6] and TROP2 is overexpressed in various epithelial neoplasms, especially in triple-negative breast cancer.[7] Most clinical trials on other target antigens, such as epidermal growth factor receptor and Notch3, are still in phases I and II.[8,9] All ADCs currently approved for breast cancer use humanized immunoglobulin G (IgG) 1 as a monoclonal antibody.[10,11]
Linker
The linker conjugates the payload with the monoclonal antibody, which is a key factor in delivering a payload to tumor cells. It must have good stability in the blood circulation. An unstable linker releases the payload earlier, which reduces the efficacy and increases the off-target toxicity. Depending on whether the linker is lysed in the cell, it can be classified as cleavable and non-cleavable. ADCs with different types of linkers have different anti-tumor mechanisms, which will be discussed below.
Payload
The most commonly used payloads in ADCs are tubulin inhibitors and topoisomerase inhibitors. The number of antigens on the tumor cell surface is small, and the number of small-molecule cytotoxic payloads that ADCs can carry is limited. To achieve an effective therapeutic concentration, it is necessary to choose cytotoxic drugs with a low half-maximal inhibitory concentration (IC50). The damage to normal tissues is correspondingly reduced owing to the precise targeting of monoclonal antibodies. The number of small-molecule cytotoxic drugs conjugated to each antibody, also known as the drug-to-antibody ratio (DAR), is an important parameter for evaluating the efficacy and toxicity of ADCs. The currently recommended DAR range is 2 to 4, with an emphasis on uniformity. With an increase in DAR, the plasma clearance rate of the ADC is increased, and the half-life is shortened.
Mechanism of ADCs
The main anti-tumor mechanism of ADCs targeting HER2 is shown in Figure 1. The monoclonal antibody in the ADCs serves as a carrier to transport payloads to target tumor cells. Trastuzumab in ADCs binds to the extracellular domain IV of HER2 and inhibits HER2 homodimerization, thereby blocking HER2-mediated signaling pathways.[12] ADCs with the cleavable linkers directly release the payloads in the tumor microenvironment or in the target cell, whereas ADCs with non-cleavable linkers enter the tumor cell and are degraded in the lysosome to release the payloads. The highly membrane-permeable payloads can penetrate the cell membrane to kill adjacent HER2-negative tumor cells, known as the bystander effect, which can further improve the efficacy in heterogeneous tumors. Non-cleavable linkers provide greater stability and therapeutic efficacy, whereas cleavable linkers usually have a bystander effect. In addition, ADCs can inhibit the shedding of the extracellular domain of HER2 and induce tumor cell apoptosis through cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) [Figure 1].[13]
Comparison of features and pharmacokinetic parameters between T-DM1 and DS-8201a
Table 1 shows the comparison between T-DM1 and DS-8201a in terms of their features and pharmacokinetic parameters, which will affect the drug dosage, efficacy, and toxicity. Both DS-8201a and T-DM1 use IgG1 as a monoclonal antibody. Gemtuzumab ozogamicin and inotuzumab ozogamicin, which have been approved for hematological malignancies, use recombinant humanized IgG4 antibodies, and some ADCs in clinical trials use humanized IgG2 antibodies, such as glembatumumab vedotin and AGS-16C3F. Compared with IgG2 and IgG4, IgG1 and IgG3 can induce stronger ADCC and CDC.[14] However, IgG3 is not recommended because of its long-hinged region and polymorphic properties, which increase its instability and immunogenicity.[15] Therefore, IgG1 antibodies, such as trastuzumab, are the best choice for ADCs targeting HER2.
Table 1.
Parameter | Trastuzumab emtansine | Trastuzumab deruxtecan |
Alternative names | T-DM1 | DS-8201a |
Trade name | Kadcyla | Enhertu |
Target (antibody) | HER2 (trastuzumab) | HER2 (trastuzumab) |
Antibody type | Humanized IgG1 | Humanized IgG1 |
Payload | DM1 (derivative of maytansine) | DXd (derivative of DX-8951) |
Mechanism of payload | Tubulin polymerization inhibitor | Topoisomerase I inhibitor |
Bystander effect | No | Yes |
Linker | Thioether SMCC linker | Maleimide tetrapeptide linker |
Linker type | Uncleavable | Cleavable (protease) |
DAR | 3.5 | 8 |
Year of initial approval | 2013 | 2019 |
Approved indication(s) | HER2-positive mBC and HER2-positive EBC with invasive residual disease | HER2-positive mBC |
Standard dosage | 3.6 mg/kg, every 3 weeks | 5.4 mg/kg, every 3 weeks |
Pharmacodynamics | ||
IC50 (ng/mL) | 11–112 | 6.7–26.8 |
Pharmacokinetics | ||
Cmax (μg/mL) | 76.2 ± 19.1 | 127 ± 17.2 |
tmax (h) | 0.5 | 1.9 |
AUCinf (μg · day−1 · mL−1) | 300 ± 66 | 590 ± 186 |
Vdss (mL/kg) | 58.4 ± 12.4 | 75.2 ± 24.2 |
t1/2 (days) | 3.1 ± 0.7 | 6.03 ± 0.60 |
Clearance (mL · day−1 · kg−1) | 2.7 ± 3.6 | 10.1 ± 3.90 |
AUCinf: Area under the concentration-time curve from 0 to infinity; Cmax: Maximum observed plasma concentration; DAR: Drug-to-antibody ratio; EBC: Early-stage breast cancer; HER2-positive: Human epidermal receptor 2 positive; IgG: Immunoglobulin G; IC50: Inhibitory concentration; mBC: Metastatic breast cancer; SMCC: Succinimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate; tmax: Time of occurrence of the maximum concentration; t1/2: Elimination half-life.
The average DAR of T-DM1 is 3.5, which is also within the recommended DAR range, whereas DS-8201A has a high DAR of 8. Under the premise of controlled safety, ADCs with a higher DAR can deliver more payloads, thus showing better efficacy. Strikingly, the IC50 of payloads in T-DM1 and DS-8201a was higher than that of conventional chemotherapeutic agents.[16,17] However, the anti-tumor mechanisms of payloads in DS-8201a and T-DM1 are different. The former is a topoisomerase I inhibitor, whereas the latter is a tubulin polymerization inhibitor. Moreover, DS-8201a has a highly membrane-permeable payload and kills adjacent HER2-negative tumor cells through the bystander effect.
New advances in clinical research
Recently, the 5-year survival rate of breast cancer has reached 80%.[18] With the improvement in the overall survival (OS) of breast cancer, drug resistance has become a great challenge. ADCs are among the most promising drugs for treating tumors. The approval of DS-8201a and T-DM1 improved the prognosis of HER2-positive breast cancer.
ADCs in advanced HER2-positive breast cancer
ADCs revised the treatment guidelines for HER2-positive breast cancer and provided patients with promising treatment options. Compared with capecitabine plus lapatinib, T-DM1 significantly improved the median progression-free survival (PFS) (6.4 months vs. 9.6 months), and median OS (25.1 months vs. 30.9 months) in the phase III EMILIA study.[19] Based on the results of the EMILIA study, T-DM1 was approved by the Food and Drug Administration in 2013 for the treatment of patients with HER2-positive metastatic breast cancer (mBC) who previously received trastuzumab and taxane, separately or in combination.[1] TH3RESA further confirmed the efficacy and safety of T-DM1 in HER2-positive advanced breast cancer.[20]
DESTINY-Breast01 aimed to evaluate the safety and efficacy of DS-8201a in HER2-positive advanced breast cancer patients who previously received T-DM1.[21] The median number of previous lines of therapy for metastatic disease was 6, suggesting that the enrolled patients were severely resistant to anti-HER2 therapy. The results showed that the objective response rate (ORR) reached 60.9%, of which 6.0% were complete responses and 54.9% were partial responses.[21] Therefore, DS-8201a is significantly superior to other anti-HER2 therapies. It was approved in 2019 for the treatment of adult patients with HER2-positive, unresectable, or mBC who had received two or more anti-HER2 therapies.
The European Society for Medical Oncology Congress 2021 and American Society of Clinical Oncology Annual Meeting 2021 also announced the progress of ADCs in HER2-positive breast cancer. The results of the phase III DESTINY-Breast03 study showed that DS-8201a had better PFS and ORR than T-DM1 in patients previously treated with trastuzumab and taxane whereas the rates of adverse events were similar.[22] In the phase I study of ARX788, the ORR of the 1.5 mg/kg dose group reached 74%, and the disease control rate reached 100%.[23] It has obtained the fast-track qualification granted by the Food and Drug Administration. A166 is an ADC composed of a novel cytotoxic drug (Duo-5, anti-microtubule agent) and trastuzumab.[24] In the phase I study of A166, the ORR of the 6 mg/kg dose group also reached 59.1%.[24] The application of ADCs in HER2-positive breast cancer will become more and more common.
ADCs in adjuvant and neoadjuvant therapy
As the first optimized treatment study for residual lesions after neoadjuvant treatment of HER2-positive breast cancer, the KATHERINE study achieved breakthrough results. Compared with trastuzumab, T-DM1 treatment significantly improved the invasive disease-free survival (88.3% vs. 77.0%), with an absolute value benefit of 11.3%.[25] However, it is still unclear whether patients who have not received neoadjuvant therapy can benefit from T-DM1 adjuvant therapy.[26,27] Dual HER2-targeted neoadjuvant therapy with pertuzumab plus trastuzumab significantly reduced the risk of HER2-positive breast cancer recurrence. The efficacy and safety of neoadjuvant T-DM1 plus pertuzumab were also assessed in the phase III KRISTINE study but did not reach the primary clinical endpoint.[28] There have been no clinical studies of DS-8201a exploring adjuvant and neoadjuvant therapy in breast cancer, but due to its high incidence rate of serious AEs, caution should be taken while conducting clinical trials.
ADCs in HER2 low-expressing breast cancer
Monoclonal antibodies are only effective for HER2-positive (immunohistochemistry [IHC] 3+ or 2+ with fluorescence in situ hybridization [FISH] amplification) breast cancers and do not exert significant anti-tumor activity against low-expression HER2 (IHC1+ or IHC2+/FISH negative) breast cancers.[29] In fact, only 15% to 20% of breast cancers are HER2-positive, and most breast cancers have low HER2 expression.[30] Furthermore, several studies have confirmed that HER2 amplification is lost or downregulated in metastatic tumors.[31,32] DS-8201a showed satisfactory efficacy in patients with HER2 low-expressing mBC.[21,33] The ORR of DS-8201a in HER2 low-expressing breast cancer reached 37%, and the median PFS was 11.1 months.[33] There have been no prospective clinical studies on T-DM1 in breast cancer with low HER2 expression, but exploratory analyses have shown limited efficacy.[34] Novel ADCs, such as trastuzumab duocarmazine and ARX788, have also shown potential anti-tumor activity against breast cancer with low HER2 expression in preclinical models or clinical trials.[35,36] This suggests that there may be a need to redefine HER2-positive breast cancer and identify optimal candidates who would benefit from anti-HER2 therapy.
ADCs in HER2-positive breast cancer with brain metastases (BMs)
Most chemotherapeutic and targeted agents have difficulty passing through the blood–brain barrier; therefore, patients with BMs tend to have a poor prognosis. Patients with HER2-positive breast cancer have the most frequent BMs.[37] As a novel anti-HER2 ADC, T-DM1 exhibits favorable efficacy in patients with BMs. In the BMs subgroup of the EMILIA study, the median OS of T-DM1 was more than twice that of lapatinib plus capecitabine (26.8 months vs. 12.9 months).[38] Montemurro et al[39] analyzed 126 patients with measurable BMs in the KAMILLA study. The results showed that 67% of patients had target lesion reduction, and the clinical benefit rate reached 43%. Moreover, the median PFS was 18.1 months among the 24 patients with asymptomatic BMs in the DESTINY-Breast01 study.[21] The efficacy of the secondary prevention effect of T-DM1 combined with temozolomide rhythm chemotherapy on locally treated HER2-positive breast cancer with BMs was evaluated in a phase I/II study.[40]
ADCs combined with other treatments
NSABP FB-10 initially explored the efficacy of T-DM1 plus lenvatinib in patients with disease progression after previous trastuzumab plus pertuzumab treatment, with an ORR of 63%.[41] The combination of ADC with other targeted therapies and the combination of ADC with bispecial-specific antibodies is worthy of further exploration.[42,43]
T-DM1 combined immunotherapy has shown promising efficacy against breast cancer. ADCs can mediate ADCC, induce lymphocyte infiltration in the tumor microenvironment, upregulate the expression of immune checkpoint receptors of CTLA4, PD-1, and PD-L1, and enhance the immune response.[44] Phase II of the KATE2 study compared the efficacy and safety of T-DM1 plus atezolizumab with T-DM1 monotherapy for trastuzumab- and taxane-treated HER2-positive advanced breast cancer.[45] There were no significant benefits from PFS in the intention to treat the population. However, in the PD-L1 positive subgroup, the median PFS (8.5 months vs. 4.1 months) and 1-year OS rate (94.3% vs. 87.9%) of T-DM1 plus atezolizumab were significantly improved. For HER2 and PD-L1 “double positive” patients, targeted therapy combined with immunotherapy was hopeful to further improve the prognosis.
The WSG ADAPT study explored the efficacy of T-DM1 combined with endocrine therapy in neoadjuvant therapy. The pathological complete response rate of the two regimens containing T-DM1 was >40%, while that of the trastuzumab regimen was 15% (P < 0.001).[26] T-DM1 combined with endocrine therapy is expected to replace targeted therapy combined with chemotherapy in the future, as well as become a new adjuvant treatment option for HER2-positive and hormone receptor (HR)-positive breast cancer. The study also suggested that early breast cancer patients who are both HER2- and HR-positive have unique biological characteristics, and future studies should evaluate the efficacy of patients with different HR statuses separately.
Adverse Events of ADCs Approved in HER2-Positive Breast Cancer
The toxicity of ADCs can be caused by multiple mechanisms, including monoclonal antibodies, payloads, off-target effects, ADCC, and CDC. Therefore, the AEs of ADCs are diverse [Figure 2], and clinicians should pay close attention to their toxicity. The most common AEs were nausea, fatigue, alopecia, and thrombocytopenia.[21,46] The most commonly reported grade 3–5 AEs with T-DM1 were thrombocytopenia, anemia, and elevated serum concentrations of aspartate aminotransferase (AST) and alanine aminotransferase (ALT)[46]. Compared with T-DM1, DS-8201a had a higher frequency of grade 3 or higher AEs (52.2% vs. 47.6%). Grade 3 or 4 increased AST or ALT with DS-8201a was relatively rare, but hematologic toxicity (except thrombocytopenia), such as neutropenia, anemia, and leukopenia, was more serious.[21] Thrombocytopenia is a special AE of T-DM1, and the incidence of any grade events was 8.7% to 30.6% (incidence of grade 3–5 events is 2.7%–14.0%).[46,47] Compared with DS-8201a, peripheral neuropathy caused by T-DM1 was more serious.[46] Most AEs were grade 1 or 2 and did not require dose reduction or discontinuation of therapy. Approximately 10% to 15% of all patients discontinued therapy due to AEs.[19,21,34] Notably, the AEs related to the discontinuation of DS-8201a were mainly due to pneumonitis and interstitial lung disease.[21,48]
Conclusions
Till August 30, 2021, there have been >1000 clinical trials on ADCs registered on the website (https://clinicaltrials.gov). The excellent efficacy and controllable safety of T-DM1 and DS-8201a in HER2-positive breast cancer have been confirmed, but ADCs, such as A166, RC48, and ARX788, are still in phase I or II clinical trials.[36,49,50] Relevant studies on predicting efficacy, drug resistance biomarkers, and other evaluation methods are also actively carried out,[51–54] which will further promote and optimize the clinical application of ADCs. Additionally, clinical trials of ADC in other malignant solid tumors, such as lung cancer and gastric cancer, are also ongoing.[55,56] The potential anti-tumor activity of ADC will provide more promising treatment options for patients with malignant tumors in the future.
Funding
The study was supported by the grants from the National Natural Science Foundation of China (No. 81874122) and the Chinese Academy of Medical Sciences (CAMS) Initiative for Innovative Medicine (No. 2017-I2M-3-004).
Conflicts of interest
None.
Footnotes
How to cite this article: Li L, Zhang D, Liu B, Lv D, Zhai J, Guan X, Yi Z, Ma F. Antibody-drug conjugates in HER2-positive breast cancer. Chin Med J 2022;135:261–267. doi: 10.1097/CM9.0000000000001932
References
- 1.Amiri-Kordestani L, Blumenthal GM, Xu QC, Zhang L, Tang SW, Ha L, et al. FDA approval: ado-trastuzumab emtansine for the treatment of patients with HER2-positive metastatic breast cancer. Clin Cancer Res 2014; 20:4436–4441. doi: 10.1158/1078-0432.ccr-14-0012. [DOI] [PubMed] [Google Scholar]
- 2.Narayan P, Osgood CL, Singh H, Chiu HJ, Ricks TK, Chow ECY, et al. FDA approval summary: fam-trastuzumab deruxtecan-nxki for the treatment of unresectable or metastatic HER2-positive breast cancer. Clin Cancer Res 2021; 27:4478–4485. doi: 10.1158/1078-0432.ccr-20-4557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sorokin P. Mylotarg approved for patients with CD33+ acute myeloid leukemia. Clin J Oncol Nurs 2000; 4:279–280. [PubMed] [Google Scholar]
- 4.Thomas A, Teicher BA, Hassan R. Antibody-drug conjugates for cancer therapy. Lancet Oncol 2016; 17:e254–e262. doi: 10.1016/s1470-2045(16)30030-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Chau CH, Steeg PS, Figg WD. Antibody-drug conjugates for cancer. Lancet 2019; 394:793–804. doi: 10.1016/s0140-6736(19)31774-x. [DOI] [PubMed] [Google Scholar]
- 6.Howlader N, Altekruse SF, Li CI, Chen VW, Clarke CA, Ries LAG, et al. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. J Natl Cancer Inst 2014; 106:dju055.doi: 10.1093/jnci/dju055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Shvartsur A, Bonavida B. Trop2 and its overexpression in cancers: regulation and clinical/therapeutic implications. Genes Cancer 2015; 6:84–105. doi: 10.18632/genesandcancer.40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Goss GD, Vokes EE, Gordon MS, Gandhi L, Papadopoulos KP, Rasco DW, et al. Efficacy and safety results of depatuxizumab mafodotin (ABT-414) in patients with advanced solid tumors likely to overexpress epidermal growth factor receptor. Cancer 2018; 124:2174–2183. doi: 10.1002/cncr.31304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Rosen LS, Wesolowski R, Baffa R, Liao KH, Hua SY, Gibson BL, et al. A phase I, dose-escalation study of PF-06650808, an anti-Notch3 antibody-drug conjugate, in patients with breast cancer and other advanced solid tumors. Invest New Drugs 2019; 38:120–130. doi: 10.1007/s10637-019-00754-y. [DOI] [PubMed] [Google Scholar]
- 10.Ocean AJ, Starodub AN, Bardia A, Vahdat LT, Isakoff SJ, Guarino M, et al. Sacituzumab govitecan (IMMU-132), an anti-Trop-2-SN-38 antibody-drug conjugate for the treatment of diverse epithelial cancers: Safety and pharmacokinetics. Cancer 2017; 123:3843–3854. doi: 10.1002/cncr.30789. [DOI] [PubMed] [Google Scholar]
- 11.Doi T, Shitara K, Naito Y, Shimomura A, Fujiwara Y, Yonemori K, et al. Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody-drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: a phase 1 dose-escalation study. Lancet Oncol 2017; 18:1512–1522. doi: 10.1016/s1470-2045(17)30604-6. [DOI] [PubMed] [Google Scholar]
- 12.Greenblatt K, Khaddour K. Trastuzumab. Treasure Island: StatPearls; 2021. [PubMed] [Google Scholar]
- 13.Junttila TT, Li G, Parsons K, Phillips GL, Sliwkowski MX. Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancer. Breast Cancer Res Treat 2010; 128:347–356. doi: 10.1007/s10549-010-1090-x. [DOI] [PubMed] [Google Scholar]
- 14.Yu J, Song Y, Tian W. How to select IgG subclasses in developing anti-tumor therapeutic antibodies. J Hematol Oncol 2020; 13:45.doi: 10.1186/s13045-020-00876-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Graziano RF, Engelhardt JJ. Role of Fc(Rs in antibody-based cancer therapy. Curr Top Microbiol Immunol 2019; 423:13–34. doi: 10.1007/82_2019_150. [DOI] [PubMed] [Google Scholar]
- 16.Phillips GDL, Li G, Dugger DL, Crocker LM, Parsons KL, Mai E, et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res 2008; 68:9280–9290. doi: 10.1158/0008-5472.can-08-1776. [DOI] [PubMed] [Google Scholar]
- 17.Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, et al. DS-8201a, a novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising antitumor efficacy with differentiation from T-DM1. Clin Cancer Res 2016; 22:5097–5108. doi: 10.1158/1078-0432.ccr-15-2822. [DOI] [PubMed] [Google Scholar]
- 18.Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Nikšić M, et al. Global surveillance of trends in cancer survival 2000-14 (CONCORD-3): analysis of individual records for 37,513,025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet 2018; 391:1023–1075. doi: 10.1016/s0140-6736(17)33326-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med 2012; 367:1783–1791. doi: 10.1056/NEJMoa1209124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Krop IE, Kim SB, Martin AG, LoRusso PM, Ferrero JM, Badovinac-Crnjevic T, et al. Trastuzumab emtansine versus treatment of physician's choice in patients with previously treated HER2-positive metastatic breast cancer (TH3RESA): final overall survival results from a randomised open-label phase 3 trial. Lancet Oncol 2017; 18:743–754. doi: 10.1016/s1470-2045(17)30313-3. [DOI] [PubMed] [Google Scholar]
- 21.Modi S, Saura C, Yamashita T, Park YH, Kim SB, Tamura K, et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N Engl J Med 2019; 382:610–621. doi: 10.1056/NEJMoa1914510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Cortés J, Kim S, Chung W, Im S, Park YH, Hegg R, et al. Trastuzumab deruxtecan (T-DXd) vs trastuzumab emtansine (T-DM1) in patients (Pts) with HER2+ metastatic breast cancer (mBC): results of the randomized phase III DESTINY-Breast03 study. Ann Oncol 2021; 32:S1283–S1346. doi: 10.1016/annonc/annonc741. [Google Scholar]
- 23.Zhang Y, Qiu M, Wang J, Zhang Y, Yuan X, Zhang T, et al. A phase 1 multicenter, dose expansion study of ARX788 as monotherapy in patients with HER2-positive advanced gastric and gastroesophageal junction adenocarcinoma (ACE-Gastric-01). J Clin Oncol 2021; 39:e16059.doi: 10.1200/JCO.2021.39.15_suppl.e16059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hu X, Zhang J, Liu R, Gao S, Qing Y, Yi S, et al. Phase I study of A166 in patients with HER2-expressing locally advanced or metastatic solid tumors. J Clin Oncol 2021; 39:1024.doi: 10.1200/JCO.2021.39.15_suppl.1024. [Google Scholar]
- 25.von Minckwitz G, Huang CS, Mano MS, Loibl S, Mamounas EP, Untch M, et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med 2018; 380:617–628. doi: 10.1056/NEJMoa1814017. [DOI] [PubMed] [Google Scholar]
- 26.Harbeck N, Gluz O, Christgen M, Kates RE, Braun M, Küemmel S, et al. De-escalation strategies in human epidermal growth factor receptor 2 (HER2)-positive early breast cancer (BC): final analysis of the west german study group adjuvant dynamic marker-adjusted personalized therapy trial optimizing risk assessment and therapy response prediction in early BC HER2- and hormone receptor-positive phase II randomized trial-efficacy, safety, and predictive markers for 12 weeks of neoadjuvant trastuzumab emtansine with or without endocrine therapy (ET) versus trastuzumab plus ET. J Clin Oncol 2017; 35:3046–3054. doi: 10.1200/jco.2016.71.9815. [DOI] [PubMed] [Google Scholar]
- 27.Tolaney SM, Trippa L, Barry W, Hu J, Dang C, Yardley D, et al. Abstract GS1-05: TBCRC 033: a randomized phase II study of adjuvant trastuzumab emtansine (T-DM1) vs paclitaxel (T) in combination with trastuzumab (H) for stage I HER2-positive breast cancer (BC) (ATEMPT). Cancer Res 2020; 80: doi: 10.1158/1538-7445.sabcs19-gs1-05. [Google Scholar]
- 28.Hurvitz SA, Martin M, Jung KH, Huang CS, Harbeck N, Valero V, et al. Neoadjuvant trastuzumab emtansine and pertuzumab in human epidermal growth factor receptor 2-positive breast cancer: three-year outcomes from the phase III KRISTINE study. J Clin Oncol 2019; 37:2206–2216. doi: 10.1200/jco.19.00882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Fehrenbacher L, Cecchini RS, Geyer CE, Jr, Rastogi P, Costantino JP, Atkins JN, et al. NSAB B-47/NRG oncology phase III randomized trial comparing adjuvant chemotherapy with or without trastuzumab in high-risk invasive breast cancer negative for HER2 by FISH and with IHC 1+ or 2. J Clin Oncol 2020; 38:444–453. doi: 10.1200/jco.19.01455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tarantino P, Hamilton E, Tolaney SM, Cortes J, Morganti S, Ferraro E, et al. HER2-low breast cancer: pathological and clinical landscape. J Clin Oncol 2020; 38:1951–1962. doi: 10.1200/jco.19.02488. [DOI] [PubMed] [Google Scholar]
- 31.Grinda T, Joyon N, Lusque A, Lefèvre S, Arnould L, Penault-Llorca F, et al. Phenotypic discordance between primary and metastatic breast cancer in the large-scale real-life multicenter French ESME cohort. NPJ Breast Cancer 2021; 7:41.doi: 10.1038/s41523-021-00252-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Niikura N, Liu J, Hayashi N, Mittendorf EA, Gong Y, Palla SL, et al. Loss of human epidermal growth factor receptor 2 (HER2) expression in metastatic sites of HER2-overexpressing primary breast tumors. J Clin Oncol 2012; 30:593–599. doi: 10.1200/jco.2010.33.8889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Modi S, Park H, Murthy RK, Iwata H, Tamura K, Tsurutani J, et al. Antitumor activity and safety of trastuzumab deruxtecan in patients with HER2-low-expressing advanced breast cancer: results from a Phase Ib Study. J Clin Oncol 2020; 38:1887–1896. doi: 10.1200/jco.19.02318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Krop IE, LoRusso P, Miller KD, Modi S, Yardley D, Rodriguez G, et al. A phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. J Clin Oncol 2011; 30:3234–3241. doi: 10.1200/jco.2011.40.5902. [DOI] [PubMed] [Google Scholar]
- 35.Banerji U, van Herpen CML, Saura C, Thistlethwaite F, Lord S, Moreno V, et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol 2019; 20:1124–1135. doi: 10.1016/s1470-2045(19)30328-6. [DOI] [PubMed] [Google Scholar]
- 36.Skidmore L, Sakamuri S, Knudsen NA, Hewet AG, Milutinovic S, Barkho W, et al. ARX788, a site-specific anti-HER2 antibody-drug conjugate, demonstrates potent and selective activity in HER2-low and T-DM1-resistant breast and gastric cancers. Mol Cancer Ther 2020; 19:1833–1843. doi: 10.1158/1535-7163.mct-19-1004. [DOI] [PubMed] [Google Scholar]
- 37.Guo Y, Arciero CA, Jiang R, Behera M, Peng L, Li X. Different breast cancer subtypes show different metastatic patterns: a Study from a Large Public Database. Asian Pac J Cancer Prev 2020; 21:3587–3593. doi: 10.31557/apjcp.2020.21.12.3587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Krop IE, Lin NU, Blackwell K, Guardino E, Huober J, Lu M, et al. Trastuzumab emtansine (T-DM1) versus lapatinib plus capecitabine in patients with HER2-positive metastatic breast cancer and central nervous system metastases: a retrospective, exploratory analysis in EMILIA. Ann Oncol 2015; 26:113–119. doi: 10.1093/annonc/mdu486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Montemurro F, Ellis P, Delaloge S, Wuerstlein R, Anton A, Button P, et al. Abstract P1-12-10: Safety and efficacy of trastuzumab emtansine (T-DM1) in 399 patients with central nervous system metastases: exploratory subgroup analysis from the KAMILLA study. Cancer Res 2017; 77: doi: 10.1158/1538-7445.sabcs16-p1-12-10. [Google Scholar]
- 40.Zimmer AS, Steinberg SM, Smart DD, Gilbert MR, Armstrong TS, Burton E, et al. Temozolomide in secondary prevention of HER2-positive breast cancer brain metastases. Future Oncol 2020; 16:899–909. doi: 10.2217/fon-2020-0094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Abraham J, Montero AJ, Jankowitz RC, Salkeni MA, Beumer JH, Kiesel BF, et al. Safety and efficacy of T-DM1 plus neratinib in patients with metastatic HER2-positive breast cancer: NSABP foundation Trial FB-10. J Clin Oncol 2019; 37:2601–2609. doi: 10.1200/jco.19.00858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Andreev J, Thambi N, Bay AEP, Delfino F, Martin J, Kelly MP, et al. Bispecific antibodies and antibody-drug conjugates (ADCs) bridging HER2 and prolactin receptor improve efficacy of HER2 ADCs. Mol Cancer Ther 2017; 16:681–693. doi: 10.1158/1535-7163.mct-16-0658. [DOI] [PubMed] [Google Scholar]
- 43.Meric-Bernstam F, Beeram M, Mayordomo JI, Hanna DL, Ajani JA, Murphy MAB, et al. Single agent activity of ZW25, a HER2-targeted bispecific antibody, in heavily pretreated HER2-expressing cancers. J Clin Oncol 2018; 36:2500.doi: 10.1200/JCO.2018.36.15_suppl.2500. [Google Scholar]
- 44.Müller P, Kreuzaler M, Khan T, Thommen DS, Martin K, Glatz K, et al. Trastuzumab emtansine (T-DM1) renders HER2+ breast cancer highly susceptible to CTLA-4/PD-1 blockade. Sci Transl Med 2015; 7:315ra188.doi: 10.1126/scitranslmed.aac4925. [DOI] [PubMed] [Google Scholar]
- 45.Emens LA, Esteva F, Beresford M, Saura C, De Laurentiis M, Kim SB, et al. Abstract PD3-01: results from KATE2, a randomized phase 2 study of atezolizumab (atezo)+trastuzumab emtansine (T-DM1) vs placebo (pbo)+T-DM1 in previously treated HER2+ advanced breast cancer (BC). Cancer Res 2019; 79:PD3-01.doi: 10.1158/1538-7445.SABCS18-PD3-01. [Google Scholar]
- 46.Diéras V, Miles D, Verma S, Pegram M, Welslau M, Baselga J, et al. Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol 2017; 18:732–742. doi: 10.1016/s1470-2045(17)30312-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Montemurro F, Ellis P, Anton A, Wuerstlein R, Delaloge S, Bonneterre J, et al. Safety of trastuzumab emtansine (T-DM1) in patients with HER2-positive advanced breast cancer: primary results from the KAMILLA study cohort 1. Eur J Cancer 2019; 109:92–102. doi: 10.1016/j.ejca.2018.12.022. [DOI] [PubMed] [Google Scholar]
- 48.Tamura K, Tsurutani J, Takahashi S, Iwata H, Krop IE, Redfern C, et al. Trastuzumab deruxtecan (DS-8201a) in patients with advanced HER2-positive breast cancer previously treated with trastuzumab emtansine: a dose-expansion, phase 1 study. Lancet Oncol 2019; 20:816–826. doi: 10.1016/s1470-2045(19)30097-x. [DOI] [PubMed] [Google Scholar]
- 49.Gong J, Shen L, Wang W, Fang J. Safety, pharmacokinetics and efficacy of RC48-ADC in a phase I study in patients with HER2-overexpression advanced solid cancer. J Clin Oncol 2018; 36:e16059.doi: 10.1200/JCO.2018.36.15_suppl.e16059. [Google Scholar]
- 50.Liu Y, Lian W, Zhao X, Qi W, Xu J, Xiao L, et al. A first in-human study of A166 in patients with locally advanced/metastatic solid tumors which are HER2-positive or HER2-amplified who did not respond or stopped responding to approved therapies. J Clin Oncol 2020; 38:1049.doi: 10.1200/JCO.2020.38.15_suppl.1049. [Google Scholar]
- 51.Baselga J, Phillips GDL, Verma S, Ro J, Huober J, Guardino AE, et al. Relationship between tumor biomarkers and efficacy in EMILIA, a phase III study of trastuzumab emtansine in HER2-positive metastatic breast cancer. Clin Cancer Res 2016; 22:3755–3763. doi: 10.1158/1078-0432.ccr-15-2499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kim SB, Wildiers H, Krop IE, Smitt M, Yu R, de Haas SL, et al. Relationship between tumor biomarkers and efficacy in TH3RESA, a phase III study of trastuzumab emtansine (T-DM1) vs. treatment of physician's choice in previously treated HER2-positive advanced breast cancer. Int J Cancer 2016; 139:2336–2342. doi: 10.1002/ijc.30276. [DOI] [PubMed] [Google Scholar]
- 53.Perez EA, de Haas SL, Eiermann W, Barrios CH, Toi M, Im YH, et al. Relationship between tumor biomarkers and efficacy in MARIANNE, a phase III study of trastuzumab emtansine ± pertuzumab versus trastuzumab plus taxane in HER2-positive advanced breast cancer. BMC Cancer 2019; 19:517.doi: 10.1186/s12885-019-5687-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Denkert C, Lambertini C, Fasching PA, Pogue-Geile KL, Mano MS, Untch M, et al. Biomarker data from KATHERINE: a phase III study of adjuvant trastuzumab emtansine (T-DM1) versus trastuzumab (H) in patients with residual invasive disease after neoadjuvant therapy for HER2-positive breast cancer. J Clin Oncol 2020; 38:502.doi: 10.1200/JCO.2020.38.15_suppl.502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Peters S, Stahel R, Bubendorf L, Bonomi P, Villegas A, Kowalski DM, et al. Trastuzumab emtansine (T-DM1) in patients with previously treated HER2-overexpressing metastatic non-small cell lung cancer: efficacy, Safety, and Biomarkers. Clin Cancer Res 2018; 25:64–72. doi: 10.1158/1078-0432.ccr-18-1590. [DOI] [PubMed] [Google Scholar]
- 56.Thuss-Patience PC, Shah MA, Ohtsu A, Van Cutsem E, Ajani JA, Castro H, et al. Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GATSBY): An international randomised, open-label, adaptive, phase 2/3 study. Lancet Oncol 2017; 18:640–653. doi: 10.1016/s1470-2045(17)30111-0. [DOI] [PubMed] [Google Scholar]