echemi logo
Product
  • Product
  • Supplier
  • Inquiry
    Home > Active Ingredient News > Antitumor Therapy > Review the research and development process of "Miracle Drug T-DXd (DS-8201)"

    Review the research and development process of "Miracle Drug T-DXd (DS-8201)"

    • Last Update: 2022-09-14
    • Source: Internet
    • Author: User
    Search more information of high quality chemicals, good prices and reliable suppliers, visit www.echemi.com

    The T-DXd is arguably the most successful ADC


    Discovery and optimization of innovative connector-payloads

    Camptothecin (CPT) analogues were selected as cytotoxic payloads


    Table 1.


    Compared to the other ADCs in Table 1, the simplest structure of the ADC Entry 4 shows low cytotoxicity due to the steric hindrance of the drug fraction and high levels of aggregation


    The payload metabolite released by ADC Entry 3 contains primary amino groups derived from a partial linker in the structure, so its membrane permeability is impaired


    Link sub-payload system key properties

    T-DXd is a new ADC coupled to a TOP1 inhibitor with seven key characteristics that distinguish it from traditional ADCs by linking subloads (Figure 1


    Figure 1.


    The novel exatecan derivative DXd inhibits TOP1 1 10 times more


    Despite the high DAR, the pharmacokinetic curve of T-DXd in cynomolgus monkeys is near-perfect


    By conducting plasma stability studies of T-DXd in several species in vitro, it was found that T-DXd exhibited high stability in plasma of all species, including cynomolgus monkeys


    The linker between the antibody and the drug fraction contains an enzyme cleavable peptide (GGFG) and an aminomethyl fraction, which reduces hydrophobicity and provides stability


    Non-clinical studies

    • Antitumor efficacy in non-clinical studies

    In mouse transplant models with different HER2 expressions, the efficacy of T-DXd was compared with T-DM1 and lower DAR HER2 ADC (DAR 3.


    Figure 6.


    • Contribution of bystanders to antitumor effects

    Due to the membrane permeability of the released cytotox payload, the released DXd remains killer not only in target cells, but also in neighboring tumor cells (Figure 7


    Figure 7.


    Xeno-tumor tissues were mixed with NCI-N87 (HER2-positive cell line) and MDA-MB-468-Luc (HER2-negative cell lines) to establish a xenograft mouse model to confirm the bystander antitumor effects


    In the T-DM1 treatment group, immunohistochemical analysis showed that HER2-positive cells were cleared, while HER2-negative cancer cells accounted for a large portion of
    the tumor tissue.

    In the T-DXd treatment group, HER2-positive and HER2-negative cells almost all disappeared, with little or no cancer cell residue in the tissues (Figure 9
    ).

    These results clearly show that under co-inoculation conditions, T-DXd has anti-tumor activity not only for HER2-positive tumor cells, but also for HER2-negative tumor cells, while T-DM1 has no effect
    on HER2-negative tumor cells.

    Therefore, T-DXd has a bystander anti-tumor effect and is expected to show a higher therapeutic effect
    in heterogeneous tumors.

    Figure 9.
    Immunohistochemical study of HER2 in co-inoculated xenomeric mouse models

    • Non-clinical safety assessment

    T-DXd was well
    tolerated in both rats of non-cross-reactive species and monkeys of cross-reactive species.

    When the dose of T-DXd was 20 to 197 mk in rats and 10 to 78.
    8 mpk in monkeys, once every 3 weeks, for
    a total of 3 times.

    In rats, the severe toxic dose for 10% of animals > 197 mpk, and the highest non-severe toxic dose (HNSTD) for monkeys was 30 mpk (Table 2
    ).

    Table 2.
    Toxicity studies of repeated administration in rats and monkeys

    The results of these non-clinical studies provide strong evidence
    for the powerful anti-tumor activity of T-DXd as well as good pharmacokinetics and safety.

    • Non-clinical pharmacokinetic evaluation

    Non-clinical pharmacokinetics studied the pharmacokinetics of T-DXd and DXd in cynomolgus monkeys
    .

    After intravenous injection, the blood concentration of T-DXd is similar
    to the downward trend of blood concentration of total antibodies.

    This indicates that the connecton is stable in plasma and that systemic DXd exposure is low
    .

    Biological distribution analysis showed that intact T-DXd was mainly present in the bloodstream without tissue-specific retention
    .

    After intravenous administration of radiolabeled T-DXd, the main route of excretion of DXd is feces
    .

    The only undimbolized DXd detectable in urine and feces (Figure 10
    ).

    Figure 10.
    Biological distribution of T-DXd in cynomolgus monkeys

    In summary, the connector exhibits a high degree of stability in the loop, and the high clearance of the DXd when released results in low system exposure
    .

    In addition, minimal tissue-specific retention and rapid excretion of DXd into the feces in its non-metabolic form have limited potential effects on drug-drug interactions and are also key factors
    in the pharmacokinetics of T-DXd.

    Clinical studies

    Between 10% and 20% of breast cancer patients are HER2 positive
    .

    Tumors in these patients tend to grow and spread more aggressively
    .

    Therefore, a number of drugs
    have been developed against HER2.

    Trastuzumab, a humanized anti-HER2 IgG1 monoclonal antibody, is the first epoch-making antiher2 drug
    to significantly improve patient survival.

    However, although patients with metastatic disease have a good antitumor response when combined with trastuzumab with common chemotherapy drugs, most patients eventually show signs of
    disease progression.

    T-DM1 significantly prolonged PFS and OS in patients with HER2-positive advanced breast cancer previously treated with trastuzumab and taxane, and was less toxic than lapatinib plus capecitabine
    .

    However, her2-targeted therapy for third-line or more treatment routes for HER2-positive metastatic breast cancer has not been established, including T-DM1-resistant tumors
    .

    In non-clinical studies, T-DXd has been shown to be effective in inhibiting the growth of her2-high- and low-expression tumors, including those that are resistant to T-DM1
    .

    In addition, non-clinical safety is good
    .

    Therefore, a phase I clinical dose escalation study was conducted to investigate the safety and tolerability of T-DXd, as well as key studies
    of advanced HER2-positive metastatic breast and gastric cancers.

    • Phase I dose escalation studies

    In the dose-escalation study (NCT02564900) of the Phase I trial, 41 patients received an initial intravenous dose of 0.
    8 to 8.
    0 mpk (N=3,0.
    8, 1.
    6, 3.
    2, and 8.
    0 mpk; N =6, 5.
    4 and 6.
    4 mpk), dose limiting toxicity (DLT) was assessed
    on a 21-day cycle.

    There is no DLT and no single dose of MTD is reached
    .

    The most common adverse effects are mild or moderate gastrointestinal and hematototoxicity
    .

    Pharmacokinetic profile analysis showed that the pharmacokinetic curve at all doses was nonlinear, with higher doses having a longer half-life, which is consistent
    with known HER2-targeted antibodies.

    Plasma concentrations of complete T-DXd, released DXd, and total antibodies after treatment with 6.
    4 mpk T-DXd are shown in
    Figure 11.

    At all time points of the assessment, the total antibody concentration was similar to the T-DXd concentration, and only a fairly low concentration of DXd was observed, indicating that the connecton payload had high stability in the patient's in vivo circulation, similar
    to non-clinical outcomes.

    Figure 11.
    In the phase I trial, the dose was 6.
    4 mpk, and the average blood concentration-time curve of T-DXd, DXd, and total antibodies in cycles 1 to 3

    On the basis of weighing efficacy and safety, in the dose expansion part of the Phase I study, doses of T-DXd monotherapy 5.
    4 and 6.
    4 mpk were selected for further study
    .

    T-DXd is used for HER2-positive metastatic breast cancer treated with T-DM1, HER2-positive stomach cancer (GC) treated with trastuzumab, breast cancer with low HER2 expression, her2 expression, or other solid tumors
    with HER2 mutations.

    Based on the interim analysis of Phase I (Table 3), the latest subgroup analysis of 99 patients with HER2-positive metastatic breast cancer who received trastuzumab and T-DM1 pre-treatment showed that 54.
    5% (54/99) confirmed ORR and 93.
    9% (93/99) of DCR
    .

    In addition, a recent subpopulation analysis of 34 patients with HER2-low expression HER2-positive metastatic breast cancer showed that 50.
    0% (17/34) of patients with ORR and 85.
    3% (29/34) of DCR
    were confirmed after T-DXd treatment.

    In 44 patients with HER2-positive gastric cancer or gastric-esophageal junction adenocarcinoma who received trastuzumab combined chemotherapy, the ORR of T-DXd was 43.
    2% (19/44) and the DCR was 79.
    5% (35/44).


    Table 3.
    T-DXd studies efficacy results in phase I dose-escalation portions by cancer type

    In addition, confirmed responses have also been observed in a range of other solid tumors with HER2 expression or HER2 mutations, including non-small cell lung cancer (NSCLC), colorectal cancer (CRC), and salivary gland cancer
    .

    In all 241 patients, the most common adverse reactions were in the gastrointestinal tract or blood system (Table 4
    ).

    Overall, T-DXd has acceptable security
    .

    Table 4.
    The most common treatment in phase I studies – emergency adverse events

    • A key clinical study for HER2-positive breast cancer

    The Phase II Destiny-Breast01 Pivot Study (NCT03248492) evaluated adult patients
    with HER2-positive metastatic breast cancer who had been treated with T-DM1.

    The first part of the study evaluated three different doses of T-DXd to determine the recommended dose (5.
    4, 6.
    4 and 7.
    4 mpk); The second part evaluates the efficacy and safety
    of the recommended dose (5.
    4 mpk).

    184 patients who had previously undergone median 6 treatments received 5.
    4 mpk of T-DXd (once every 3 weeks) with an ORR of 60.
    9%.


    The median effective time was 14.
    8 months and the median PFS was 16.
    4 months
    (Table 5).

    Table 5.
    Clinical primary endpoint assessment of the Phase II Destination-Breast01 key study

    During the study period, the most common grade 3 or higher adverse reactions were a decrease in neutrophil count (20.
    7%), anemia (8.
    7%), and nausea (7.
    6%)
    .

    Trial drugs accounted for 13.
    6% of patients associated with interstitial lung disease (ILD) (grade 1 or 2, 10.
    9%); Level 3 or Level 4, 0.
    5%; Level 5, 2.
    2%) (Table 6
    ).

    Table 6.
    Adverse events in the Phase II Destination-Breast01 study

    These results show that T-DXd has long-lasting antitumor activity
    in the population of pre-treated patients with HER2-positive metastatic breast cancer.

    In addition to nausea and bone marrow suppression, ILD was observed, so close monitoring of patients' lung symptoms
    was required.

    • A key clinical study of HER2-positive stomach cancer

    The Phase II DISTY-GASTEC01 key study (NCT03329690) evaluated T-DXd compared with chemotherapy in patients with HER2-positive advanced gastric cancer
    .

    Patients who developed progressing HER2-positive gastric or gastroesophageal junction adenocarcinoma at at least two previous treatments, including trastuzumab, were randomly assigned to receive T-DXd or chemotherapy
    in a 2:1 ratio.

    Of the 187 patients treated, 125 received T-DXd and 62 received chemotherapy (55 received irinotecan and 7 received paclitaxel
    ).

    The ORR in the T-DXd treatment group was 51%, compared to 14% in the doctor-selected chemotherapy group (Table 7
    ).

    Overall survival (OS) The T-DXd group was four months (12.
    5 vs.
    8.
    4 months)
    than the chemotherapy group.

    Table 7.
    Evaluation of the primary clinical endpoint of the Phase II DISTY-GASTEC01 key study

    The most common adverse events of grade 3 or higher were a decrease in neutrophil count (51% and 24% in the T-DXd group and the doctor-selected group, respectively), anemia (38% and 23%, respectively), and a decrease in white blood cell count (21% and 11%, respectively) (Table 8
    ).

    A total of 12 patients had T-DXd-associated ILD or pneumonia (grade 1 or 2 in 9 patients and grade 3 or 4 in 3 patients
    ).

    One treatment-related death (pneumonia)
    was found in the T-DXd treatment group.

    Table 8.
    Adverse events in the Phase II DISTY-GASTEC01 study

    These results show that T-DXd therapy significantly improves response and overall survival in
    patients with HER2-positive gastric cancer compared with standard therapy.

    The toxic side effects of bone marrow suppression and ILD should be taken seriously
    .

    Summary

    T-DXd as a proof-of-concept study established a new ADC connector payload system that demonstrated superior efficacy and acceptable safety
    by using the unique TOP1 inhibitor DXd and stable connectors.

    Despite the high DAR8 with hydrophobic payload, the optimized connector produces an ADC with excellent physico-chemical and pharmacokinetic properties, and there is no inclusion of hydrophilic masking parts in the linker structure, such as polyethylene glycol – an unprecedented achievement
    .

    T-DXd became the first FDA-approved ADC
    with an average DAR of about 8.

    The success of T-DXd proves the great potential of the new generation of ADC drugs, brings unlimited imagination to everyone, and opens up another wave of ADC research and development boom
    .

    Congratulations to the T-DXd research team, but also to all those who
    have worked quietly on the road to the development of new tumor drugs.

    T-DXd is not an end, but a new beginning
    .

    With more breakthroughs in the future, we believe that cancer has become a chronic disease, and it is
    getting closer and closer.

    References

    1.
    Chemical Linkers in Antibody–Drug Conjugates (ADCs).

    2.
    Trastuzumab Deruxtecan in Previously TreatedHER2-Low Advanced Breast Cancer.

    3.
    Trastuzumab Deruxtecan: Changing the Destiny of HER2 Expressing Solid Tumors

    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.