Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.761
1.
BMC Cancer ; 24(1): 631, 2024 May 23.
Article En | MEDLINE | ID: mdl-38783218

BACKGROUND: Cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) combined with endocrine therapy (ET) are currently recommended by the National Comprehensive Cancer Network (NCCN) guidelines and the European Society for Medical Oncology (ESMO) guidelines as the first-line (1 L) treatment for patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative, locally advanced/metastatic breast cancer (HR+/HER2- LABC/mBC). Although there are many treatment options, there is no clear standard of care for patients following 1 L CDK4/6i. Understanding the real-world effectiveness of subsequent therapies may help to identify an unmet need in this patient population. This systematic literature review qualitatively synthesized effectiveness and safety outcomes for treatments received in the real-world setting after 1 L CDK4/6i therapy in patients with HR+/ HER2- LABC/mBC. METHODS: MEDLINE®, Embase, and Cochrane were searched using the Ovid® platform for real-world evidence studies published between 2015 and 2022. Grey literature was searched to identify relevant conference abstracts published from 2019 to 2022. The review was conducted in accordance with PRISMA guidelines (PROSPERO registration: CRD42023383914). Data were qualitatively synthesized and weighted average median real-world progression-free survival (rwPFS) was calculated for NCCN/ESMO-recommended post-1 L CDK4/6i treatment regimens. RESULTS: Twenty records (9 full-text articles and 11 conference abstracts) encompassing 18 unique studies met the eligibility criteria and reported outcomes for second-line (2 L) treatments after 1 L CDK4/6i; no studies reported disaggregated outcomes in the third-line setting or beyond. Sixteen studies included NCCN/ESMO guideline-recommended treatments with the majority evaluating endocrine-based therapy; five studies on single-agent ET, six studies on mammalian target of rapamycin inhibitors (mTORi) ± ET, and three studies with a mix of ET and/or mTORi. Chemotherapy outcomes were reported in 11 studies. The most assessed outcome was median rwPFS; the weighted average median rwPFS was calculated as 3.9 months (3.3-6.0 months) for single-agent ET, 3.6 months (2.5-4.9 months) for mTORi ± ET, 3.7 months for a mix of ET and/or mTORi (3.0-4.0 months), and 6.1 months (3.7-9.7 months) for chemotherapy. Very few studies reported other effectiveness outcomes and only two studies reported safety outcomes. Most studies had heterogeneity in patient- and disease-related characteristics. CONCLUSIONS: The real-world effectiveness of current 2 L treatments post-1 L CDK4/6i are suboptimal, highlighting an unmet need for this patient population.


Breast Neoplasms , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Protein Kinase Inhibitors , Receptor, ErbB-2 , Humans , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Female , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Progression-Free Survival
2.
J Pharm Biomed Anal ; 245: 116185, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38723556

Human epidermal growth factor receptor 2 (HER2) is a key player in the pathogenesis and progression of breast cancer and is currently a primary target for breast cancer immunotherapy. Bioactivity determination is necessary to guarantee the safety and efficacy of therapeutic antibodies targeting HER2. Nevertheless, currently available bioassays for measuring the bioactivity of anti-HER2 mAbs are either not representative or have high variability. Here, we established a reliable reporter gene assay (RGA) based on T47D-SRE-Luc cell line that expresses endogenous HER2 and luciferase controlled by serum response element (SRE) to measure the bioactivity of anti-HER2 antibodies. Neuregulin-1 (NRG-1) can lead to the heterodimerization of HER2 on the cell membrane and induce the expression of downstream SRE-controlled luciferase, while pertuzumab can dose-dependently reverse the reaction, resulting in a good dose-response curve reflecting the activity of the antibody. After optimizing the relevant assay parameters, the established RGA was fully validated based on ICH-Q2 (R1), which demonstrated that the method had excellent specificity, accuracy, precision, linearity, and stability. In summary, this robust and innovative bioactivity determination assay can be applied in the development and screening, release control, biosimilar assessment and stability studies of anti-HER2 mAbs.


Antibodies, Monoclonal, Humanized , Biological Assay , Genes, Reporter , Luciferases , Neuregulin-1 , Receptor, ErbB-2 , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptor, ErbB-2/antagonists & inhibitors , Humans , Cell Line, Tumor , Antibodies, Monoclonal, Humanized/pharmacology , Biological Assay/methods , Luciferases/genetics , Neuregulin-1/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/genetics , Female , Antineoplastic Agents, Immunological/pharmacology , Reproducibility of Results , Response Elements
3.
Bioorg Chem ; 147: 107371, 2024 Jun.
Article En | MEDLINE | ID: mdl-38643564

Due to the strong selectivity and permeability of tumor tissue, anti-cancer peptide-drug conjugates (PDCs) can accumulate high concentration of toxic payloads at the target, effectively killing tumor cells. This approach holds great promise for tumor-targeted treatment. In our previous study, we identified the optimal peptide P1 (NPNWGRSWYNQRFK) targeting HER2 from pertuzumab, a monoclonal antibody that blocks the HER2 signaling pathway. Here, a series of PDCs were constructed through connecting P1 and CPT with different linkers. Among these, Z8 emerged as the optimal compound, demonstrating good antitumor activity and targeting ability in biological activity tests. Z8 exhibited IC50 values of 1.04 ± 0.24 µM and 1.91 ± 0.71 µM against HER2-positive SK-BR-3 and NCI-N87 cells, respectively. Moreover, superior antitumor activity and higher biosafety of Z8 were observed compared to the positive control CPT in vivo, suggesting a novel idea for the construction of PDCs.


Antineoplastic Agents , Camptothecin , Cell Proliferation , Drug Screening Assays, Antitumor , Peptides , Receptor, ErbB-2 , Humans , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Camptothecin/pharmacology , Camptothecin/chemistry , Structure-Activity Relationship , Animals , Cell Proliferation/drug effects , Peptides/chemistry , Peptides/pharmacology , Peptides/chemical synthesis , Molecular Structure , Dose-Response Relationship, Drug , Mice , Drug Discovery , Cell Line, Tumor , Female , Mice, Inbred BALB C , Mice, Nude
4.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38673831

Designed ankyrin repeat protein (DARPin) G3 is an engineered scaffold protein. This small (14.5 kDa) targeting protein binds with high affinity to human epidermal growth factor receptor 2 (HER2). HER2 is overexpressed in several cancers. The use of the DARPin G3 for radionuclide therapy is complicated by its high renal reabsorption after clearance via the glomeruli. We tested the hypothesis that a fusion of the DARPin G3 with an albumin-binding domain (ABD) would prevent rapid renal excretion and high renal reabsorption resulting in better tumour targeting. Two fusion proteins were produced, one with the ABD at the C-terminus (G3-ABD) and another at the N-terminus (ABD-G3). Both variants were labelled with 177Lu. The binding properties of the novel constructs were evaluated in vitro and their biodistribution was compared in mice with implanted human HER2-expressing tumours. Fusion with the ABD increased the retention time of both constructs in blood compared with the non-ABD-fused control. The effect of fusion with the ABD depended strongly on the order of the domains in the constructs, resulting in appreciably better targeting properties of [177Lu]Lu-G3-ABD. Our data suggest that the order of domains is critical for the design of targeting constructs based on scaffold proteins.


Receptor, ErbB-2 , Animals , Female , Humans , Mice , Albumins/metabolism , Ankyrin Repeat , Cell Line, Tumor , Lutetium , Protein Binding , Protein Domains , Radioisotopes , Radiopharmaceuticals/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/chemistry , Tissue Distribution , Molecular Targeted Therapy
5.
Theranostics ; 14(6): 2442-2463, 2024.
Article En | MEDLINE | ID: mdl-38646654

Rationale: Resistance to targeted therapies like trastuzumab remains a critical challenge for HER2-positive breast cancer patients. Despite the progress of several N-terminal HSP90 inhibitors in clinical trials, none have achieved approval for clinical use, primarily due to issues such as induction of the heat shock response (HSR), off-target effects, and unfavorable toxicity profiles. We sought to examine the effects of HVH-2930, a novel C-terminal HSP90 inhibitor, in overcoming trastuzumab resistance. Methods: The effect of HVH-2930 on trastuzumab-sensitive and -resistant cell lines in vitro was evaluated in terms of cell viability, expression of HSP90 client proteins, and impact on cancer stem cells. An in vivo model with trastuzumab-resistant JIMT-1 cells was used to examine the efficacy and toxicity of HVH-2930. Results: HVH-2930 was rationally designed to fit into the ATP-binding pocket interface cavity of the hHSP90 homodimer in the C-terminal domain of HSP90, stabilizing its open conformation and hindering ATP binding. HVH-2930 induces apoptosis without inducing the HSR but by specifically suppressing the HER2 signaling pathway. This occurs with the downregulation of HER2/p95HER2 and disruption of HER2 family member heterodimerization. Attenuation of cancer stem cell (CSC)-like properties was associated with the downregulation of stemness factors such as ALDH1, CD44, Nanog and Oct4. Furthermore, HVH-2930 administration inhibited angiogenesis and tumor growth in trastuzumab-resistant xenograft mice. A synergistic effect was observed when combining HVH-2930 and paclitaxel in JIMT-1 xenografts. Conclusion: Our findings highlight the potent efficacy of HVH-2930 in overcoming trastuzumab resistance in HER2-positive breast cancer. Further investigation is warranted to fully establish its therapeutic potential.


Breast Neoplasms , Drug Resistance, Neoplasm , HSP90 Heat-Shock Proteins , Receptor, ErbB-2 , Trastuzumab , Xenograft Model Antitumor Assays , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Drug Resistance, Neoplasm/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Animals , Female , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Cell Line, Tumor , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Mice, Nude , Apoptosis/drug effects , Cell Survival/drug effects , Antineoplastic Agents/pharmacology
6.
Biomed Pharmacother ; 174: 116522, 2024 May.
Article En | MEDLINE | ID: mdl-38565055

In recent decades, there has been a surge in the approval of monoclonal antibodies for treating a wide range of hematological and solid malignancies. These antibodies exhibit exceptional precision in targeting the surface antigens of tumors, heralding a groundbreaking approach to cancer therapy. Nevertheless, monoclonal antibodies alone do not show sufficient lethality against cancerous cells compared to chemotherapy. Consequently, a new class of anti-tumor medications, known as antibody-drug conjugates (ADCs), has been developed to bridge the divide between monoclonal antibodies and cytotoxic drugs, enhancing their therapeutic potential. ADCs are chemically synthesized by binding tumor-targeting monoclonal antibodies with cytotoxic payloads through linkers that are susceptible to cleavage by intracellular proteases. They combined the accurate targeting of monoclonal antibodies with the potent efficacy of cytotoxic chemotherapy drugs while circumventing systemic toxicity and boasting superior lethality over standalone targeted drugs. The human epidermal growth factor receptor (HER) family, which encompasses HER1 (also known as EGFR), HER2, HER3, and HER4, plays a key role in regulating cellular proliferation, survival, differentiation, and migration. HER2 overexpression in various tumors is one of the most frequently targeted antigens for ADC therapy in HER2-positive cancers. HER2-directed ADCs have emerged as highly promising treatment modalities for patients with HER2-positive cancers. This review focuses on three approved anti-HER2 ADCs (T-DM1, DS-8201a, and RC48) and reviews ongoing clinical trials and failed trials based on anti-HER2 ADCs. Finally, we address the notable challenges linked to ADC development and underscore potential future avenues for tackling these hurdles.


Immunoconjugates , Neoplasms , Receptor, ErbB-2 , Humans , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
7.
Sci Rep ; 14(1): 9869, 2024 04 30.
Article En | MEDLINE | ID: mdl-38684839

Cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) are the standard agents for treating patients with estrogen receptor-positive and human epidermal growth factor receptor 2-negative advanced breast cancer (ER + HER2 - ABC). However, markers predicting the outcomes of CDK4/6i treatment have yet to be identified. This study was a single-center retrospective cohort study. We retrospectively evaluated 101 patients with ER + HER2 - ABC receiving CDK4/6i in combination with endocrine therapy at Fukuyama City Hospital between November 2017 and July 2021. We investigated the clinical outcomes and the safety of CDK4/6i treatment, and the absolute lymphocyte count (ALC) and neutrophil-to-lymphocyte ratio (NLR) as predictive markers for CDK4/6i. We defined the cut-off values as 1000/µL for ALC and 3 for NLR, and divided into "low" and "high" groups, respectively. We evaluated 43 and 58 patients who received abemaciclib and palbociclib, respectively. Patients with high ALC and low NLR had significantly longer overall survival than those with low ALC and high NLR (high vs. low; ALC: HR 0.29; 95% CI 0.12-0.70; NLR: HR 2.94; 95% CI 1.21-7.13). There was no significant difference in efficacy between abemaciclib and palbociclib and both had good safety profiles. We demonstrated that ALC and NLR might predict the outcomes of CDK4/6i treatment in patients with ER + HER2 - ABC.


Breast Neoplasms , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Lymphocytes , Neutrophils , Protein Kinase Inhibitors , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/mortality , Breast Neoplasms/blood , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Middle Aged , Lymphocytes/metabolism , Lymphocyte Count , Retrospective Studies , Protein Kinase Inhibitors/therapeutic use , Aged , Adult , Pyridines/therapeutic use , Piperazines/therapeutic use , Aminopyridines/therapeutic use , Benzimidazoles/therapeutic use , Aged, 80 and over , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Treatment Outcome
8.
Anticancer Res ; 44(5): 1837-1844, 2024 May.
Article En | MEDLINE | ID: mdl-38677753

BACKGROUND/AIM: Although there are curative treatment options for non-muscle-invasive bladder cancer, the recurrence of this tumor is high. Therefore, novel targeted therapies are needed for the complete removal of bladder cancer cells in stages of localized disease, in order to avoid local recurrence, to spare bladder cancer patients from stressful and expensive treatment procedures and to increase their quality of life and life expectancy. This study tested a new approach for the photoimmunotherapy (PIT) of bladder cancer. MATERIALS AND METHODS: We generated a cysteine modified recombinant version of the antibody cetuximab targeting the epidermal growth factor receptor (EGFR) on the surface of bladder cancer cells. Then, we coupled the novel photoactivatable phthalocyanine dye WB692-CB1 via a maleimide linker to the free cysteines of the antibody. PIT was performed by incubating bladder cancer cells with the antibody dye conjugate followed by irradiation with visible red light. RESULTS: The conjugate was able to induce specific cytotoxicity in EGFR-positive bladder cancer cells in a light dose-dependent manner. Enhanced cytotoxicity in RT112 bladder cancer cells was evoked by addition of a second antibody dye conjugate targeting HER2 or by repeated cycles of PIT. CONCLUSION: Our new antibody dye conjugate targeting EGFR-expressing bladder cancer cells is a promising candidate for the future PIT of bladder cancer patients.


ErbB Receptors , Immunoconjugates , Immunotherapy , Receptor, ErbB-2 , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , ErbB Receptors/immunology , ErbB Receptors/antagonists & inhibitors , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Immunotherapy/methods , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Cell Line, Tumor , Cetuximab/pharmacology , Cetuximab/therapeutic use , Cetuximab/chemistry , Phototherapy/methods
9.
Bull Cancer ; 111(5): 441-451, 2024 May.
Article Fr | MEDLINE | ID: mdl-38480056

INTRODUCTION: Quality of life (QoL) and patient satisfaction are major concerns in oncology. METHODS: The aim of this prospective observational study was to evaluate these parameters according to the mode of administration of anti-HER2 (subcutaneous [SC] versus intravenous [IV]), the place of administration (Home Hospitalization or HOD versus hospital) for patients supervised by an advanced practice nurse (APN). RESULTS: Between January 2022 and June 2023, 32 patients were included. They were statistically more satisfied with subcutaneous management (P=0.0004), a result explained by the speed of administration (43.5%), comfort during administration (26%) even though some expressed pain on injection and felt less anxiety (26%). Management by the APN seems more appropriate when anti-HER2 drugs were administered in HOD. In HOD, patients perceived an overall improvement in their quality of life, appetite and cognitive abilities, with a reduction in fatigue, pain and depression (P<0.05). However, the rate of outsourcing to HOD remained too low (30.4%), as 56.3% of patients would have liked to be cared for in HOD if they had had the opportunity. CONCLUSION: SC administration of anti-HER2 under the supervision of an APN has advantages for the patient, resulting in greatest satisfaction and improved patient QOL, preferably in HOD.


Breast Neoplasms , Patient Satisfaction , Quality of Life , Receptor, ErbB-2 , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/psychology , Prospective Studies , Middle Aged , Receptor, ErbB-2/antagonists & inhibitors , Injections, Subcutaneous , Aged , Anxiety , Hospitalization , Adult , Trastuzumab/therapeutic use , Trastuzumab/administration & dosage , Depression , Fatigue , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , Appetite/drug effects
11.
Curr Treat Options Oncol ; 25(5): 585-604, 2024 May.
Article En | MEDLINE | ID: mdl-38539034

OPINION STATEMENT: Targeted treatment strategies are available for human epidermal growth factor receptor 2 (HER2)-positive (amplified and/or overexpressed) metastatic colorectal cancer (mCRC), and HER2 testing is indicated in patients with mCRC. At present, standard of care first-line treatment for those with HER2-positive mCRC remains chemotherapy in combination with epidermal growth factor receptor (EGFR) inhibitors or bevacizumab, depending on RAS/BRAF mutational status and tumor sidedness. HER2-targeted agents should be considered for those with RAS/BRAF wild-type disease in subsequent-line treatment and in first-line treatment for patients not appropriate for intensive therapy. While the choice of anti-HER2 therapy is empiric given lack of head-to-head comparisons, the combination of trastuzumab plus tucatinib has received FDA accelerated approval for use in this setting and is generally the authors' preference. Trastuzumab plus lapatinib, trastuzumab plus pertuzumab, and trastuzumab deruxtecan (T-DXd) also have evidence of efficacy in this setting. As T-DXd has demonstrated activity following treatment with other HER2-targeted regimens and carries an increased risk of high-grade toxicities, the authors favor reserving it for use after progression on prior anti-HER2 therapy. HER2-targeted therapies that inhibit signal transduction appear to have limited activity in those with RAS mutations, including trastuzumab-containing regimens. However, the antibody drug conjugate T-DXd has some data showing efficacy in this setting, and the authors would consider T-DXd in subsequent-line therapy for HER2-positive, RAS-mutated mCRC. Several areas of uncertainty remain regarding how to best utilize HER2-targeted therapies in mCRC. These include the optimal sequence of anti-HER2 therapies with chemotherapy and anti-EGFR therapies, the optimal combination partners for anti-HER2 therapies, and the incorporation of predictive biomarkers to guide use of anti-HER2 therapies. Results of ongoing studies may thus alter the treatment paradigm above in the coming years.


Antineoplastic Combined Chemotherapy Protocols , Colorectal Neoplasms , Molecular Targeted Therapy , Receptor, ErbB-2 , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Neoplasm Metastasis , Biomarkers, Tumor , Disease Management , Treatment Outcome , Clinical Trials as Topic
12.
Hematol Oncol Clin North Am ; 38(3): 659-675, 2024 Jun.
Article En | MEDLINE | ID: mdl-38485551

Gastroesophageal cancers are highly diverse tumors in terms of their anatomic and molecular characteristics, making drug development challenging. Recent advancements in understanding the molecular profiles of these cancers have led to the identification of several new biomarkers. Ongoing clinical trials are investigating new targeted agents with promising results. CLDN18.2 has emerged as a biomarker with established activity of associated targeted therapies. Other targeted agents, such as bemarituzumab and DKN-01, are under active investigation. As new agents are incorporated into the treatment continuum, the questions of biomarker overlap, tumor heterogeneity, and toxicity management will need to be addressed.


Biomarkers, Tumor , Esophageal Neoplasms , Molecular Targeted Therapy , Receptor, ErbB-2 , Stomach Neoplasms , Humans , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Molecular Targeted Therapy/methods , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Biomarkers, Tumor/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
13.
Hematol Oncol Clin North Am ; 38(3): 585-598, 2024 Jun.
Article En | MEDLINE | ID: mdl-38521686

The Trastuzumab for Gastric Cancer (ToGA) trial marked a pivotal moment in the adoption of trastuzumab for treating advanced human epidermal growth factor receptor 2 (HER2)-positive esophagogastric (EG) cancer. The KEYNOTE-811 trial brought to light the synergistic effect of immune modulation and HER2 targeting. Additionally, the emergence of trastuzumab deruxtecan (T-DXd) highlighted the potential of new pharmaceutical technologies to extend response, particularly for patients who have advanced beyond initial HER2-targeted therapies. This review aims to navigate through both the successes and challenges encountered historically, as well as promising current trials on innovative and transformative therapeutic strategies, including promising first-in-class and novel first-in-human agents.


Camptothecin/analogs & derivatives , Immunoconjugates , Molecular Targeted Therapy , Receptor, ErbB-2 , Humans , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Molecular Targeted Therapy/methods , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/therapy , Trastuzumab/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Antineoplastic Agents, Immunological/therapeutic use , Clinical Trials as Topic
14.
Drug Resist Updat ; 74: 101078, 2024 May.
Article En | MEDLINE | ID: mdl-38503142

AIMS: Human epidermal growth factor receptor 2 (HER2) is an oncogenic receptor tyrosine kinase amplified in approximately 20% of breast cancer (BC). HER2-targeted therapies are the linchpin of treating HER2-positive BC. However, drug resistance is common, and the main resistance mechanism is unknown. We tested the hypothesis that drug resistance results mainly from inadequate or lack of inhibition of HER2 and its family member epidermal growth factor receptor (EGFR). METHODS: We used clinically relevant cell and tumor models to assess the impact of targeted degradation of HER2 and EGFR on trastuzumab resistance. Trastuzumab is the most common clinically used HER2 inhibitor. Targeted degradation of HER2 and EGFR was achieved using recombinant human protein PEPDG278D, which binds to the extracellular domains of the receptors. siRNA knockdown was used to assess the relative importance of EGFR and HER2 in trastuzumab resistance. RESULTS: Both HER2 and EGFR are overexpressed in all trastuzumab-resistant HER2-positive BC cell and tumor models and that all trastuzumab-resistant models are highly vulnerable to targeted degradation of HER2 and EGFR. Degradation of HER2 and EGFR induced by PEPDG278D causes extensive inhibition of oncogenic signaling in trastuzumab-resistant HER2-positive BC cells. This is accompanied by strong growth inhibition of cultured cells, orthotopic patient-derived xenografts, and metastatic lesions in the brain and lung of trastuzumab-resistant HER2-positive BC. siRNA knockdown indicates that eliminating both HER2 and EGFR is necessary to maximize therapeutic outcome. CONCLUSIONS: This study unravels the therapeutic vulnerability of trastuzumab-resistant HER2-positive BC and shows that an agent that targets the degradation of both HER2 and EGFR is highly effective in overcoming drug resistance in this disease. The findings provide new insights and innovations for advancing treatment of drug-resistant HER2-positive breast cancer that remains an unmet problem.


Breast Neoplasms , Drug Resistance, Neoplasm , ErbB Receptors , Receptor, ErbB-2 , Signal Transduction , Trastuzumab , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Animals , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Mice , Signal Transduction/drug effects , Cell Line, Tumor , Xenograft Model Antitumor Assays , Proteolysis/drug effects , Cell Proliferation/drug effects
15.
Cancer Discov ; 14(5): 828-845, 2024 May 01.
Article En | MEDLINE | ID: mdl-38358339

Zanidatamab is a bispecific human epidermal growth factor receptor 2 (HER2)-targeted antibody that has demonstrated antitumor activity in a broad range of HER2-amplified/expressing solid tumors. We determined the antitumor activity of zanidatamab in patient-derived xenograft (PDX) models developed from pretreatment or postprogression biopsies on the first-in-human zanidatamab phase I study (NCT02892123). Of 36 tumors implanted, 19 PDX models were established (52.7% take rate) from 17 patients. Established PDXs represented a broad range of HER2-expressing cancers, and in vivo testing demonstrated an association between antitumor activity in PDXs and matched patients in 7 of 8 co-clinical models tested. We also identified amplification of MET as a potential mechanism of acquired resistance to zanidatamab and demonstrated that MET inhibitors have single-agent activity and can enhance zanidatamab activity in vitro and in vivo. These findings provide evidence that PDXs can be developed from pretreatment biopsies in clinical trials and may provide insight into mechanisms of resistance. SIGNIFICANCE: We demonstrate that PDXs can be developed from pretreatment and postprogression biopsies in clinical trials and may represent a powerful preclinical tool. We identified amplification of MET as a potential mechanism of acquired resistance to the HER2 inhibitor zanidatamab and MET inhibitors alone and in combination as a therapeutic strategy. This article is featured in Selected Articles from This Issue, p. 695.


Antibodies, Bispecific , Receptor, ErbB-2 , Xenograft Model Antitumor Assays , Humans , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Mice , Female , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology
16.
Cell Chem Biol ; 31(5): 989-999.e7, 2024 May 16.
Article En | MEDLINE | ID: mdl-38307028

Human epidermal growth factor receptor 2 (HER2)-targeted agents have proven to be effective, however, the development of resistance to these agents has become an obstacle in treating HER2+ breast cancer. Evidence implicates HUNK as an anti-cancer target for primary and resistant HER2+ breast cancers. In this study, a selective inhibitor of HUNK is characterized alongside a phosphorylation event in a downstream substrate of HUNK as a marker for HUNK activity in HER2+ breast cancer. Rubicon has been established as a substrate of HUNK that is phosphorylated at serine (S) 92. Findings indicate that HUNK-mediated phosphorylation of Rubicon at S92 promotes both autophagy and tumorigenesis in HER2/neu+ breast cancer. HUNK inhibition prevents Rubicon S92 phosphorylation in HER2/neu+ breast cancer models and inhibits tumorigenesis. This study characterizes a downstream phosphorylation event as a measure of HUNK activity and identifies a selective HUNK inhibitor that has meaningful efficacy toward HER2+ breast cancer.


Antineoplastic Agents , Breast Neoplasms , Receptor, ErbB-2 , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Female , Phosphorylation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice , Animals , Cell Proliferation/drug effects , Cell Line, Tumor , Mice, Nude , Structure-Activity Relationship
17.
Acta Pharmacol Sin ; 45(6): 1287-1304, 2024 Jun.
Article En | MEDLINE | ID: mdl-38360930

HER2-positive (HER2+) metastatic breast cancer (mBC) is highly aggressive and a major threat to human health. Despite the significant improvement in patients' prognosis given the drug development efforts during the past several decades, many clinical questions still remain to be addressed such as efficacy when combining different therapeutic modalities, best treatment sequences, interindividual variability as well as resistance and potential coping strategies. To better answer these questions, we developed a mechanistic quantitative systems pharmacology model of the pathophysiology of HER2+ mBC that was extensively calibrated and validated against multiscale data to quantitatively predict and characterize the signal transduction and preclinical tumor growth kinetics under different therapeutic interventions. Focusing on the second-line treatment for HER2+ mBC, e.g., antibody-drug conjugates (ADC), small molecule inhibitors/TKI and chemotherapy, the model accurately predicted the efficacy of various drug combinations and dosing regimens at the in vitro and in vivo levels. Sensitivity analyses and subsequent heterogeneous phenotype simulations revealed important insights into the design of new drug combinations to effectively overcome various resistance scenarios in HER2+ mBC treatments. In addition, the model predicted a better efficacy of the new TKI plus ADC combination which can potentially reduce drug dosage and toxicity, while it also shed light on the optimal treatment ordering of ADC versus TKI plus capecitabine regimens, and these findings were validated by new in vivo experiments. Our model is the first that mechanistically integrates multiple key drug modalities in HER2+ mBC research and it can serve as a high-throughput computational platform to guide future model-informed drug development and clinical translation.


Breast Neoplasms , Receptor, ErbB-2 , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Humans , Female , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology , Network Pharmacology , Models, Biological , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Mice , Cell Line, Tumor , Neoplasm Metastasis
18.
Drug Resist Updat ; 74: 101068, 2024 May.
Article En | MEDLINE | ID: mdl-38402670

The treatment for trastuzumab-resistant breast cancer (BC) remains a challenge in clinical settings. It was known that CD47 is preferentially upregulated in HER2+ BC cells, which is correlated with drug resistance to trastuzumab. Here, we developed a novel anti-CD47/HER2 bispecific antibody (BsAb) against trastuzumab-resistant BC, named IMM2902. IMM2902 demonstrated high binding affinity, blocking activity, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and internalization degradation effects against both trastuzumab-sensitive and trastuzumab-resistant BC cells in vitro. The in vivo experimental data indicated that IMM2902 was more effective than their respective controls in inhibiting tumor growth in a trastuzumab-sensitive BT474 mouse model, a trastuzumab-resistant HCC1954 mouse model, two trastuzumab-resistant patient-derived xenograft (PDX) mouse models and a cord blood (CB)-humanized HCC1954 mouse model. Through spatial transcriptome assays, multiplex immunofluorescence (mIFC) and in vitro assays, our findings provided evidence that IMM2902 effectively stimulates macrophages to generate C-X-C motif chemokine ligand (CXCL) 9 and CXCL10, thereby facilitating the recruitment of T cells and NK cells to the tumor site. Moreover, IMM2902 demonstrated a high safety profile regarding anemia and non-specific cytokines release. Collectively, our results highlighted a novel therapeutic approach for the treatment of HER2+ BCs and this approach exhibits significant anti-tumor efficacy without causing off-target toxicity in trastuzumab-resistant BC cells.


Antibodies, Bispecific , Breast Neoplasms , CD47 Antigen , Drug Resistance, Neoplasm , Immunotherapy , Receptor, ErbB-2 , Trastuzumab , Xenograft Model Antitumor Assays , Humans , Animals , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Female , Drug Resistance, Neoplasm/drug effects , Mice , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/immunology , Immunotherapy/methods , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Cell Line, Tumor , Antibody-Dependent Cell Cytotoxicity/drug effects , Phagocytosis/drug effects
19.
Breast ; 72: 103597, 2023 Dec.
Article En | MEDLINE | ID: mdl-37944341

BACKGROUND: Inetetamab is a novel recombinant humanized anti-HER2 monoclonal antibody. This study aimed to evaluate the efficacy and safety of inetetamab and predictive factors for response in HER2-positive metastatic breast cancer (MBC) patients. METHODS: A cohort of HER2-positive MBC patients who received inetetamab-based therapy between June 2020 and August 2021 was evaluated. The primary endpoint was progression-free survival (PFS), and the secondary endpoints included objective response rate (ORR) and disease control rate (DCR). Adverse events (AEs) were graded according to the National Cancer Institute Common Toxicity Criteria. RESULTS: A total of 141 patients were included in the final analysis. The median PFS of the entire cohort was 7.1 months. The median number of treatment lines administered was three. The ORR was 36.9 %, and the DCR was 80.9 %. The most frequently employed treatment strategy was inetetamab + chemotherapy (49/141, 34.8 %), followed by inetetamab + HER2-tyrosine kinase inhibitors (HER2-TKIs) + chemotherapy, inetetamab + pertuzumab + chemotherapy, inetetamab + endocrine treatment and inetetamab + HER2-TKIs. Cox multivariate analysis revealed that PFS was associated with liver metastasis (hazard ratio [HR] 2.112, 95 % confidence interval [CI] 1.334-3.343, p = 0.001), previous HER2-TKI treatment (HR 2.019, 95 % CI 1.133-3.597, p = 0.017) and estrogen receptor positivity (HR 0.587, 95 % CI 0.370-0.934, p = 0.024). The toxicity was tolerable, with neutropenia being the most common treatment-related grade 3/4 AE (14.9 %). CONCLUSION: Inetetamab demonstrates effectiveness with a manageable safety profile, offering a promising therapeutic option for HER2-positive breast cancer patients who have shown resistance to prior anti-HER2 treatments.


Antibodies, Monoclonal , Antineoplastic Agents , Breast Neoplasms , Receptor, ErbB-2 , Female , Humans , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/secondary , East Asian People , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/therapeutic use , Treatment Outcome , Antibodies, Monoclonal/therapeutic use
20.
Int J Mol Sci ; 24(14)2023 Jul 13.
Article En | MEDLINE | ID: mdl-37511163

Gastric cancer is an aggressive disease with increasing global incidence in recent years. Human epidermal growth receptor 2 (HER2) is overexpressed in approximately 10-20% of gastric cancers. The implementation of targeted therapy against HER2 as part of the standard of care treatment in metastatic disease has improved the prognosis of this subset of patients. However, gastric cancer still has high mortality rates and urgently requires new treatment strategies. The combination of immunotherapy with HER2-targeted therapies has shown synergistic effects in preclinical models, this being the rationale behind exploring this combination in clinical trials in locally advanced and metastatic settings. Additionally, the irruption of antibody-drug conjugates and other novel HER2-targeted agents has led to the development of numerous clinical trials showing promising results. This review presents the molecular mechanisms supporting the use of HER2-targeted drugs in combination with immunotherapy and provides an overview of the therapeutic scenario of HER2-positive disease. We focus on the role of immunotherapy but also summarize emerging therapies and combinations under clinical research that may change the standard treatment in HER-2 positive disease in the future.


Antineoplastic Agents , Immunoconjugates , Receptor, ErbB-2 , Stomach Neoplasms , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Immunotherapy , Receptor, ErbB-2/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Trastuzumab/pharmacology , Trastuzumab/therapeutic use
...