echemi logo
Product
  • Product
  • Supplier
  • Inquiry
    Home > Active Ingredient News > Antitumor Therapy > Based on the unique design and mechanism, T-DXd opens up a sea of stars for HER2-positive advanced gastric cancer

    Based on the unique design and mechanism, T-DXd opens up a sea of stars for HER2-positive advanced gastric cancer

    • Last Update: 2022-11-04
    • Source: Internet
    • Author: User
    Search more information of high quality chemicals, good prices and reliable suppliers, visit www.echemi.com
    *For medical professionals only to read and refer from structural design, mechanism of action to research progress, revealing the mystery
    of T-DXd in the treatment of HER2-positive advanced gastric cancer.


    Human epidermal growth factor receptor 2 (HER2) is a proto-oncogene located on chromosome 17, and its encoded HER2 protein has tyrosine kinase activity, which is involved in a variety of intracellular signaling, mainly leading to tumorigenesis by promoting cell proliferation and inhibiting apoptosis[1].

    HER2 overexpression or gene amplification has been found in about 12-20% of patients with gastric cancer/gastroesophageal junction adenocarcinoma (hereinafter referred to as gastric cancer) [2], suggesting the possibility of anti-HER2 therapy in this part of
    gastric cancer patients.
    The phase III ToGA study confirmed for the first time that trastuzumab can prolong the survival of HER2-positive advanced gastric cancer [3], and since then gastric cancer has entered a new era of targeted therapy, which has also opened up further exploration
    of anti-HER2 drugs in the field of HER2-positive gastric cancer.


    However, in the exploration of first-line therapy, the research of tripapa bitarget and lapatinib has ended in failure [4,5]; In second-line therapy, neither monoclonal antibodies (margetuximab), TKI (lapatinib), nor conventional antibody conjugates (ADC, T-DM1) significantly improved patient survival outcomes compared with conventional chemotherapy [6-8].

    Until the emergence of a new generation of ADC drug T-DXd, the treatment of HER2-positive advanced gastric cancer once again ushered in a breakthrough, with an objective response rate (ORR) of 51.
    3% and a median overall survival (OS) of 1 year after T-DXd third-line therapy [9]; In contrast, the current CSCO guideline recommends a third-line therapy with an apatinib ORR of 2.
    84 percent and a median OS of 6.
    5 months and nivolumab of 5.
    26 months [10,11].

    T-DXd performed well in the third-line treatment of HER2-positive advanced gastric cancer, becoming the first HER2 ADC drug
    to be approved for HER2-positive advanced gastric cancer indications.


    The unique properties build the basis of the efficacy of T-DXd in the treatment of HER2-positive advanced gastric cancer


    The ADC consists of antibodies, cytotoxic drug loaders, and linkers; The main mechanism is to use the specific targeting effect of antibodies to deliver the drug to the target cells, so that the drug can be concentrated on the tumor cells to exert anti-tumor killing effect, and can reduce the drug concentration in other normal tissues and organs, so as to obtain a wider treatment window
    .
    Specifically, after the ADC enters the body, it selectively binds to the antigen of the target cell through the targeting effect of the antibody; The formed ADC-antigen complex enters tumor cells
    by endocytosis.
    Subsequently, under the action of lysosomes, cytotoxic drugs are released through cleavage of linkers, which play the role of killing tumor cells [12].

    In the above process, the affinity of antigens and antibodies, the stability of linkers, the potency of cytotoxic drug loading, membrane permeability, and release timing will all affect
    the efficacy of ADC drugs.


    Figure 1.
    ADC structure and mechanism of action [12].

    Figure A.
    General ADC structure
    .
    These include antibodies, linkers, and
    payloads.
    Figure B The mechanism
    of action of the general ADC.
    After distribution through the hematological system to tumor tissue (I), the ADC binds to its cell surface antigen, and the ADC-antigen complex is internalized by antigen-mediated endocytosis (II).

    ADC processing (linker cleavage and/or antibody degradation) occurs during endosome-lysosome (IIIa, IIIb) transport, resulting in the release of the drug load into the cytoplasm (IV) in a biologically active form, ultimately leading to cell death (V).

    If the drug load is permeable to the cell membrane, it can enter adjacent cells to achieve the bystander effect (VI).


    As an emerging anti-tumor drug, the research and development exploration of ADC has never stopped, and the continuous optimization of various components has effectively promoted the development process of ADC, which has undergone three generations of technological changes
    so far.
    T-DXd targeting HER2 has improved off-target toxicity, hydrophobicity and homogeneity, and has become a "bright star"
    in the third-generation ADC drugs.
    T-DXd is formed by humanized HER2 antibody conjugated to topoisomerase I inhibitor (DXd) via cleavable linkers, which has many advantages
    in structure and mechanism of action.


    Figure 2.
    T-DXd structure diagram [13].

    Structure of T-DXd: the antibody moiety is conjugated to the linker-drug-loading moiety by cysteine residues, and the DAR reaches a maximum of 8
    .


    Antibody conjugation of DXd did not affect its HER2 recognition ability and ADCC effect


    The antibody component of T-DXd is humanized trastuzumab
    .
    Preclinical studies have found [14] that T-DXd and naked antibodies bind to HER2 at different concentrations with similar affinities and have similar dissociation constants (Kd, 7.
    3 ng/mL and 7.
    8 ng/mL, respectively).

    Therefore, antibody conjugation of DXd did not affect its HER2 recognition ability
    .
    In addition to the direct killing of drugs, antibody-dependent cell-mediated cytotoxicity (ADCC) effects also play an important role
    in the process of ADC drugs killing tumors.
    Preclinical studies have found that T-DXd has similar ADCC effects
    to naked antibodies.


    Figure 3.
    T-DXd contrasts HER2 binding capacity and ADCC activity of naked antibodies[14].

    Figure A.
    The affinity of T-DXd (DS-8201a) and naked antibody (anti-HER2 Ab, trastuzumab) for HER2 protein is determined by ELISA
    .
    Figure B The ADCC activity
    of T-DXd and naked antibody was compared by measuring the degree of lysis of SK-BR-3 cells by human peripheral blood mononuclear cells.


    Optimized cleassurable linkers provide a bystander effect, and unique coupling techniques maximize the DAR of T-DXd to 8


    The linker is the key structure of the ADC, which determines whether the ADC drug has a bystander effect and the size of the drug-load/antibody ratio (DAR), which will seriously affect the efficacy
    of the drug.
    First, the linker must have some stability to ensure the integrity of the ADC drug during blood circulation before reaching the target cell to avoid damage to normal tissues or cells
    by early drug release.
    T-DXd uses tetrapeptide-based digestion to cleave linkers, which are more
    stable than traditional aminobenzyl linkers.
    Preclinical studies have shown [14] that the presence of free DXd
    in plasma is detected only at the beginning after a single injection into mice, rats, monkeys, and humans.
    After 21 days, the release rate of DXd in plasma is only 1.
    2%-3.
    9%, of which the release rate in human plasma is about 2%, while the release rate of T-DM1 is as high as 18.
    4%
    on day 4.
    It shows that the tetrapeptide linker of T-DXd is highly stable in the blood, and the DXd shedding rate is extremely low, which ensures the safety
    of the system.


    Figure 4.
    The T-DXd linker is stable in plasma and has a very low drop-off rate [14].

    Left: Pharmacokinetics
    of T-DXd in cynomolgus monkeys.
    T-DXd is administered at a dose of 3 mg/kg
    intravenously.
    Right: DXd release rate
    in plasma.


    When the ADC enters the target cell, the linker needs to be effectively lysed to quickly release the drug load to function
    .
    After T-DXd enters the cell, it only needs to be recognized by lysosomal protease to recognize the linker cleavage site to release the drug load [14,15], which is simple and more
    efficient.
    This characteristic makes T-DXd a bystander effect, which is of great significance
    for the treatment of gastric cancer, a highly heterogeneous tumor.
    Preclinical studies have demonstrated that T-DXd significantly reduces the volume of mixed tumors (HER2-positive/HER2-negative) [16].


    Figure 5.
    Only T-DXd can effectively kill rodent mixed graft tumors, and significantly reduce the volume of mixed tumors [16].


    The conjugation method used for the linker is also important and will determine the homogeneity and DAR
    of the drug.
    Natural conjugation methods include lysine coupling and cysteine coupling, and cysteine conjugation results in better
    homogeneity of ADC drugs.
    The homogeneity will directly affect the distribution and metabolism of ADC drugs in vivo, and high homogeneity will improve drug purity and benefit quality control [13,17].

    On the basis of cysteine coupling, the coupling technology of T-DXd was further optimized to make it more homogeneous and reach the highest theoretical value of DAR 8[13].


    Figure 6.
    Lysine is coupled to cysteine[13,17].


    Drug loading has the characteristics of unique mechanism, high antitumor activity and strong cell membrane permeability


    The drug carrier DXd is a topoisomerase I inhibitor, which is different from the mechanism of chemotherapy drugs commonly used in gastric cancer such as microtubule inhibitors (taxane), DNA damage factors (platinum), and antimetabolites (fluorouracil) to avoid cross-resistance [18].

    Secondly, DXd has 10 times the antitumor activity of its metabolite SN38 compared to irinotecan [13], while SN38 is 100-1000 times
    more active than irinotecan.


    The system half-life of free DXd is short, and T-DXd is conducive to control system toxicity after drug release [19].

    Moreover, DXd's self-deamination design gives it strong cell membrane permeability [13], which is another condition for
    T-DXd to produce a bystander effect.


    Overall, thanks to the optimized linker technology, the homogeneity of T-DXd is greatly improved and the DAR reaches 8
    .
    Moreover, the efficient release of drug loading can be achieved by lysosomal protease cleavage linkers, and because the drug load has high cell membrane permeability, T-DXd has a strong bystander effect, showing great potential
    in the treatment of gastric cancer with high heterogeneity.
    The unique mechanism and excellent pharmacokinetic characteristics of drug-loaded DXd are also indispensable for
    T-DXd to exert strong anti-tumor effects.
    All in all, the systematic optimization of each key element of the ADC has played a huge role in improving the clinical efficacy of the ADC, and T-DXd has "emerged"
    with technological advancements.


    All-round layout, T-DXd in HER2-positive advanced gastric cancer research exploration


    Figure 7.
    Summary of studies of T-DXd in HER2-positive advanced gastric cancer


    A-J101 study, the first human trial to explore the anti-tumor effect of T-DXd in the treatment of HER2-positive gastric cancer


    Phase I clinical study A-J101 evaluates the safety and tolerability of
    T-DXd in patients with advanced solid tumors.
    Part 1 is a dose escalation study, and Part 2 is a dose expansion study
    .
    The study identified a dose of 6.
    4 mg/kg for T-DXd in patients with gastric cancer [20,21].


    Figure 8.
    A-J101 study design[20].


    A total of 44 patients were enrolled in this study, and 27% of them had received at least 5-line system therapy
    in the past.
    After a median follow-up of 5.
    5 months, the ORR was as high as 43.
    2% in the total population and the disease control rate (DCR) was 79.
    5%; The median progression-free survival (PFS) was 5.
    6 months and the median OS was 12.
    8 months
    .
    Post-hoc subgroup analysis also found that gastric cancer patients who had previously received irinotecan could continue to benefit from T-DXd treatment, with an ORR of 41.
    7% and a DCR of 79.
    2% in 24 patients treated with irinotecan, which was comparable
    to the general population.
    ≥ grade 3 TEAE was 64%, and there were
    no AE-induced drug-related deaths.


    A-J101 initially demonstrated the good anti-tumor activity of T-DXd in the field of advanced gastric cancer, on the basis of which T-DXd opened the DESTINY-Gastric (DG) series of studies in the field of gastric cancer to verify its efficacy and safety
    .


    DG01 study, the first randomized controlled study to confirm the efficacy of T-DXd in advanced gastric cancer


    The Phase II DG01 study evaluated the efficacy and safety of T-DXd versus physician-selected chemotherapy (paclitaxel or irinotecan) in patients with advanced gastric cancer who had progressed to at least two treatment regimens, including fluorouracil, platinum-based drugs, and trastuzumab, in Japan and South Korea [9].

    。 HER2-positive patients (IHC3+ or IHC2+/ISH+) treated with trastuzumab were included in the primary cohort, and patients with low HER2 expression who had not been treated with trastuzumab were included in the exploratory cohort (cohort 1: IHC2+/ISH-, cohort 2: IHC1+).

    The main cohort included a total of 187 treated patients, of whom 125 were enrolled in the T-DXd group and 62 were enrolled in the chemotherapy group (55 received irinotecan and 7 received paclitaxel).


    Figure 9.
    DG01 study design[9].


    A preliminary analysis of the main cohort, published in the New England Journal of Medicine (NEJM) in 2020 [9], showed that the ORR in the T-DXd group was significantly higher than that in the chemotherapy group (51.
    3% vs.
    14.
    3%), and 10 patients achieved complete remission (CR), while no patients in the chemotherapy group selected by the physician achieved CR.

    The confirmatory DCRs were 86% and 62%, respectively, with a median confirmed duration of response (DoR) of 11.
    3 and 3.
    9 months, and a median OS of 12.
    5 months and 8.
    4 months
    , respectively.
    Safety results were similar to those previously reported, with no new safety signals
    emerging.


    Based on this research, in May 2020, the US Food and Drug Administration (FDA) granted T-DXd breakthrough therapy designation (BTD).

    In September 2020, Japan's Ministry of Health, Labor and Welfare (MHLW) approved T-DXd for the treatment
    of patients with HER2-positive unresectable advanced or recurrent gastric cancer who have previously received chemotherapy.
    In January 2021, the FDA approved T-DXd for the treatment of
    adults with locally advanced or metastatic HER2-positive advanced gastric cancer who had previously received trastuzumab.


    The 2020 ESMO Annual Meeting reported results from an exploratory cohort of low HER2 expression in DG01 [22], with an ORR of 36.
    8% in cohort 1, a median confirmed DoR of 7.
    6 months, a median OS of 7.
    8 months, and a median PFS of 4.
    4 months
    .
    Cohort 2 had an ORR of 19%, a median confirmed DoR of 12.
    5 months, a median OS of 8.
    5 months, and a median PFS of 2.
    8 months
    .
    This exploratory cohort showed excellent efficacy in patients with low HER2 expression, confirming the structural advantages of
    T-DXd.


    Figure 10.
    T-DXd has been used for low HER2 expression and has achieved long-term benefits [22].


    DG06 is a Chinese bridging trial of the DG01 study, which includes Chinese patients with advanced gastric cancer who are HER2-positive (IHC 3+/2+) and have received at least two previous systemic therapies [23], and it is expected that this study will add new evidence to T-DXd in the treatment of HER2-positive advanced gastric cancer patients in China and bring good news
    to more Chinese patients.


    Figure 11.
    DESTINY-Gastric 06 Study Design [23].


    DG02 study to verify the benefit of second-line treatment of T-DXd in European and American populations


    The DG02 study is an open-label, single-arm phase II study that includes patients with HER2-positive advanced gastric cancer who have progressed after trastuzumab treatment to explore the efficacy and safety
    of T-DXd in the second-line treatment of European and American populations.
    The latest data from the 2022 ESMO Congress [24] shows that as of August 2021, the median follow-up time was 10.
    2 months, and the independent center review (ICR) confirmed ORR was 41.
    8%, of which 5.
    1% of patients achieved CR, confirmed DCR was 81.
    0%, median DoR was 8.
    1 months, and median response time (TTR) was 1.
    4 months
    .
    The median OS was 12.
    1 months and the median PFS was 5.
    6 months
    .


    Safety results were consistent with initial analyses and reported in previous studies, suggesting that adverse effects of T-DXd were tolerated
    with longer follow-up.
    It is worth noting that the ESMO conference also released patient-reported outcome (PRO) data in the DG02 study, and the evaluation of EQ-5D VAS and FACT-Ga scale showed that there was no significant decrease
    in the quality of life of patients from T-DXd treatment to cycle 7.


    Figure 12.
    PRO benefited from the DG02 study [24].


    In addition, an ongoing global, multicenter, randomized, open-label, phase III DG04 study will compare head-to-head the differences between T-DXd monotherapy and ramoximab plus paclitaxel regimen in trastuzumab treated HER2-positive advanced gastric cancer [25] to further strengthen the therapeutic position
    of T-DXd in HER2-positive advanced gastric cancer.


    Multi-dimensional exploration to broaden the boundaries of beneficiary groups


    While the success of T-DXd monotherapy has also opened the exploration
    of combination therapy.
    The DG03 study included patients with advanced gastric cancer with HER2-positive (IHC3+ or 2+/ISH+) in North America, Europe, and Asia, with multiple therapeutic arms to evaluate the safety and antitumor activity of T-DXd monotherapy and combination regimens [26].

    Part 1 is the ongoing dose escalation phase that includes at least second-line gastric cancer patients
    who have progressed after treatment with trastuzumab-containing regimens.
    Part 2 is the dose expansion phase, enrolling patients with untreated gastric cancer
    .


    Figure 13.
    DG03 study design[26].


    The 2022 ASCOGI reported preliminary results from the DG03 study-Part 1 T-DXd in combination with 5-fluorouracil (5-FU) or capecitabine (Cap) [26].

    As of 1 November 2021, the median follow-up was 5.
    2 months
    .
    PR2D is T-DXd 6.
    4 mg/kg + 5-FU 600mg/m 2, T-DXd 6.
    4 mg/kg + Cap 1000mg/m2
    .
    The ORR of T-DXd combined with 5-FU group reached 66.
    7%, and the ORR of T-DXd combined with Cap group reached 71.
    4%.


    In addition, the EPOC2003 study initiated by investigators evaluated the antitumor activity of T-DXd neoadjuvant therapy in patients with HER2-positive gastric cancer [27].

    The primary cohort included HER2-positive (IHC3+ or IHC2+/ISH+) patients, and the exploratory cohort included patients
    with low HER2 expression (IHC1+ or IHC2+/ISH-) and HER2-ECD>11.
    6 ng/mL.
    It is hoped that the publication of the research data will also bring new treatment options
    for the perioperative administration of HER2-positive gastric cancer.


    Figure 14.
    EPOC2003 study design[27].


    summary


    In summary, optimized linkon technology, highly active drug delivery with unique mechanisms, high DAR and potent bystander effect constitute the key characteristics
    of T-DXd to achieve breakthrough clinical benefits in the highly heterogeneous field of gastric cancer treatment.
    With the DG01 study T-DXd has been approved for HER2-positive advanced gastric cancer in the United States and Japan, it is the first ADC drug approved for indications in the field of gastric cancer, and it is currently under approval for indications in other countries and regions such as the European Union
    .
    Moreover, T-DXd is also actively conducting a series of explorations in the field of HER2-positive advanced gastric cancer, and it is believed that with the accumulation of research evidence, the benefit population of T-DXd will be expanded
    .
    During the same period, other HER2-targeted ADC drugs have also emerged at home and abroad, and it is expected that a hundred flowers will bloom under the successful leadership of T-DXd and bring new hope
    for the treatment of more gastric cancer patients.


    Expert commentary


    Professor Li Enxiao pointed out that HER2 as a target of targeted therapy was first approved by the US FDA for HER2-positive advanced breast cancer
    in September 1998.
    Secondly, based on the results of the ToGA study reported by Lancet in 2010, which confirmed that trastuzumab combined with chemotherapy can prolong the survival of HER2-positive advanced gastric cancer to 13.
    8 months, trastuzumab was again approved by the US FDA as the standard first-line treatment for HER2-positive advanced gastric cancer
    .
    The exploration of a new stage of targeted therapy for advanced gastric cancer has begun, especially the application
    of anti-HER2 molecular targeted drugs.
    Unfortunately, in the first- and second-line exploration, many clinical studies of multiple drugs such as tyrosine kinase inhibitor lapatinib and traditional ADC drug T-DM1 have failed
    one after another.


    Until 2020, New England Journal reported that the new ADC drug T-DXd third-line treatment of advanced gastric cancer ORR was as high as 51.
    3%, with a median OS of 12.
    5 months, and T-DXd also became the first ADC drug
    approved for HER2-positive advanced gastric cancer indications.
    The new generation of ADC drug T-DXd has improved off-target toxicity, hydrophobicity and homogeneity, and has optimized linker technology, highly active drug loading with unique mechanism, high DAR and strong bystander effect, which has greatly improved its clinical efficacy
    .
    The A-J101 study preliminarily confirmed the safety and tolerability of T-DXd in patients with advanced gastric cancer, the DG01 study and the DG02 study verified the benefits of second-line treatment of T-DXd in Asian and European and American populations, respectively, and clinical trials of T-DXd combination therapy and perioperative treatment are also being explored
    .
    We have reason to expect that in the future, T-DXd combined with anti-angiogenic macromolecular monoclonal antibodies or multi-target anti-angiogenic TKIs, immune checkpoint inhibitors and other drugs will further improve its efficacy and bring multi-faceted treatment benefits
    to more gastric cancer patients.


    Expert Profile
    Dr.
    Yangwei Fan

    Subspecialty of digestive tract oncology, Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University

    MD Research Assistant Resident

    Member and Secretary of the Neuroendocrine Oncology Professional Committee of Shaanxi Anti-Cancer Association

    Member of the Anti-Cancer Drug Professional Committee of Shaanxi Anti-Cancer Association

    Member of the Tumor Biotherapy Professional Committee of Shaanxi Anti-Cancer Association

    Member of the Youth Committee of Translational Medicine of Shaanxi Anti-Cancer Association

    Member of the MDT Professional Committee of Liver Tumor of Shaanxi International Medical Exchange Promotion Association

    Member of the Precision Treatment Professional Committee of Xi'an Cancer Rehabilitation Association

    • His main research interests focus on basic and clinical research on HER2-positive gastrointestinal tumors

    • He has published 26 academic papers, including 12 SCI papers as the first author or corresponding author, and 5 oral reports or poster presentations
      at high-end academic conferences at home and abroad.

    • He has presided over or participated in more than 10 national, provincial and ministerial and university-level natural science funds, presided over 1 CSCO fund, and participated in more than 10 national clinical research projects
      of new anti-tumor drugs.



    Expert profile
    Professor Li Eunxiao


    Leader of the Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University

    Chief Physician, Professor, Doctor of Medicine, Doctoral Supervisor

    Chairman of the Tumor Biotherapy Professional Committee of Shaanxi Anti-Cancer Association

    CSCO Governor

    Vice Chairman of CSCO Biliary Tract Cancer Committee, member of CSCO Pancreatic Cancer Committee

    Member of the Standing Committee of GP-NET Special Committee and Standing Committee of Integrated Traditional Chinese and Western Medicine

    Member of the National Health Commission's Capacity Building and Continuing Education Oncology Committee

    Member of the Standing Committee of the Cancer Rehabilitation Committee of the Chinese Rehabilitation Medical Association

    Member of the Standing Committee of the Molecular Targeted Therapy Committee of the Chinese Society of Biomedical Engineering

    Member of the Standing Committee of the Molecular Targeted Therapy Committee of the Chinese Anti-Cancer Association

    Member of the Standing Committee of the MDT Professional Committee of Precision Medicine and Oncology of China Research Hospital Association

    Editorial Board Member of Modern Oncology



    References:

    [1] Boku N.
    HER2-positive gastric cancer.
    Gastric Cancer.
    2014 Jan; 17(1):1-12.

    [2] Bartley AN, Washington MK, Colasacco C, 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 Feb; 35(4):446-464.

    [3] Bang YJ, Van Cutsem E, Feyereislova A, et al.
    Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial.
    Lancet.
    2010 Aug 28; 376(9742):687-697.

    [4] Tabernero J, Hoff PM, Shen L, et al.
    Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): final analysis of a double-blind, randomised, placebo-controlled phase 3 study.
    Lancet Oncol.
    2018 Oct; 19(10):1372-1384.

    [5] Hecht JR, Bang YJ, Qin SK, et al.
    Lapatinib in Combination With Capecitabine Plus Oxaliplatin in Human Epidermal Growth Factor Receptor 2-Positive Advanced or Metastatic Gastric, Esophageal, or Gastroesophageal Adenocarcinoma: TRIO-013/LOGiC--A Randomized Phase III Trial.
    J Clin Oncol.
    2016 Feb 10; 34(5):443-451.

    [6] Catenacci DVT, Kang YK, Park H, et al.
    Margetuximab plus pembrolizumab in patients with previously treated, HER2-positive gastro-oesophageal adenocarcinoma (CP-MGAH22-05): a single-arm, phase 1b-2 trial.
    Lancet Oncol.
    2020 Aug; 21(8):1066-1076.

    [7] Satoh T, Xu RH, Chung HC, et al.
    Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN--a randomized, phase III study.
    J Clin Oncol.
    2014 Jul 1; 32(19):2039-2049.

    [8] Thuss-Patience PC, Shah MA, Ohtsu A, 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 May; 18(5):640-653.

    [9] Shitara K, Bang YJ, Iwasa S, et al; Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer.
    N Engl J Med.
    2020 Jun 18; 382(25):2419-2430.

    [10] Li J, Qin S, Xu J, et al.
    Randomized, Double-Blind, Placebo-Controlled Phase III Trial of Apatinib in Patients With Chemotherapy-Refractory Advanced or Metastatic Adenocarcinoma of the Stomach or Gastroesophageal Junction.
    J Clin Oncol.
    2016 May 1; 34(13):1448-1454.

    [11] Kang YK, Boku N, Satoh T, et al.
    Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial.
    Lancet.
    2017 Dec 2; 390(10111):2461-2471.

    [12] Birrer MJ, Moore KN, Betella I, et al.
    Antibody-Drug Conjugate-Based Therapeutics: State of the Science.
    J Natl Cancer Inst.
    2019 Jun 1; 111(6):538-549.

    [13] Nakada T, Sugihara K, Jikoh T, et al.
    The Latest Research and Development into the Antibody-Drug Conjugate, [fam-] Trastuzumab Deruxtecan (DS-8201a), for HER2 Cancer Therapy.
    Chem Pharm Bull (Tokyo).
    2019; 67(3):173-185.

    [14] Ogitani Y, Aida T, Hagihara K, 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 Oct 15; 22(20):5097-5108.

    [15] LoRusso PM, Weiss D, Guardino E, et al.
    Trastuzumab emtansine: a unique antibody-drug conjugate in development for human epidermal growth factor receptor 2-positive cancer.
    Clin Cancer Res.
    2011 Oct 15; 17(20):6437-6447.

    [16] Ogitani Y, Hagihara K, Oitate M, et al.
    Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity.
    Cancer Sci.
    2016 Jul; 107(7):1039-1046.

    [17] Conilh L, Fournet G, Fourmaux E, et al.
    Exatecan Antibody Drug Conjugates Based on a Hydrophilic Polysarcosine Drug-Linker Platform.
    Pharmaceuticals (Basel).
    2021 Mar 9; 14(3):247.

    [18] Guidelines Working Committee of Chinese Society of Clinical Oncology.
    Chinese Society of Clinical Oncology (CSCO) guidelines for the diagnosis and treatment of gastric cancer 2022[M].
    Beijing:People's Medical Publishing House, 2022.

    [19] Nagai Y, Oitate M, Shiozawa H, et al.
    Comprehensive preclinical pharmacokinetic evaluations of trastuzumab deruxtecan (DS-8201a), a HER2-targeting antibody-drug conjugate, in cynomolgus monkeys.
    Xenobiotica.
    2019 Sep; 49(9):1086-1096.

    [20] Doi T, Shitara K, Naito Y, 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-oesophagealtumours: a phase 1 dose-escalation study.
    Lancet Oncol.
    2017 Nov; 18(11):1512-1522.

    [21] Shitara K, Iwata H, Takahashi S, et al.
    Trastuzumab deruxtecan (DS-8201a) in patients with advanced HER2-positive gastric cancer: a dose-expansion, phase 1 study.
    Lancet Oncol.
    2019 Jun; 20(6):827-836.

    [22] Trastuzumab deruxtecan (T-DXd; DS-8201) in patients with HER2-low, advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma: Results of the exploratory cohorts in the phase II, multicenter, open-label DESTINY-Gastric01 study.
    2020 ESMO Mini Oral 1422.

    [23] https://clinicaltrials.
    gov/ct2/show/NCT04989816

    [24] Updated analysis of DESTINY-Gastric02: A phase II single-arm trial of trastuzumab deruxtecan (T-DXd) in western patients (Pts) with HER2-positive (HER2+) unresectable/metastatic gastric/gastroesophageal junction (GEJ) cancer who progressed on or after trastuzumab-containing regimen.
    2022 ESMO 1205MO.

    [25] https://clinicaltrials.
    gov/ct2/show/NCT04704934

    [26] Dose-escalation and dose-expansion study of trastuzumab deruxtecan (T-DXd) monotherapy and combinations in patients (pts) with advanced/metastatic HER2+ gastric cancer (GC)/ gastroesophageal junction adenocarcinoma (GEJA): DESTINY-Gastric03.
    2022 ASCO GI.
    Abstract 295.

    [27] Phase 2 study of trastuzumab deruxtecan in the neoadjuvant treatment for patients with HER2-positive gastric and gastroesophageal junction adenocarcinoma (EPOC2003).
    2022 ASCO.
    Abstract TPS4161.

    *This article is only for providing scientific information to medical professionals and does not represent the views of this platform


    This article is an English version of an article which is originally in the Chinese language on echemi.com and is provided for information purposes only. This website makes no representation or warranty of any kind, either expressed or implied, as to the accuracy, completeness ownership or reliability of the article or any translations thereof. If you have any concerns or complaints relating to the article, please send an email, providing a detailed description of the concern or complaint, to service@echemi.com. A staff member will contact you within 5 working days. Once verified, infringing content will be removed immediately.

    Contact Us

    The source of this page with content of products and services is from Internet, which doesn't represent ECHEMI's opinion. If you have any queries, please write to service@echemi.com. It will be replied within 5 days.

    Moreover, if you find any instances of plagiarism from the page, please send email to service@echemi.com with relevant evidence.