Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 482
Filtrer
1.
J Exp Clin Cancer Res ; 43(1): 236, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39164784

RÉSUMÉ

BACKGROUND: Anti-HER2 therapies, including the HER2 antibody-drug conjugates (ADCs) trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd), have led to improved survival outcomes in patients with HER2-overexpressing (HER2+) metastatic breast cancer. However, intrinsic or acquired resistance to anti-HER2-based therapies remains a clinical challenge in these patients, as there is no standard of care following disease progression. The purpose of this study was to elucidate the mechanisms of resistance to T-DM1 and T-DXd in HER2+ BC patients and preclinical models and identify targets whose inhibition enhances the antitumor activity of T-DXd in HER2-directed ADC-resistant HER2+ breast cancer in vitro and in vivo. METHODS: Targeted DNA and whole transcriptome sequencing were performed in breast cancer patient tissue samples to investigate genetic aberrations that arose after anti-HER2 therapy. We generated T-DM1 and T-DXd-resistant HER2+ breast cancer cell lines. To elucidate their resistance mechanisms and to identify potential synergistic kinase targets for enhancing the efficacy of T-DXd, we used fluorescence in situ hybridization, droplet digital PCR, Western blotting, whole-genome sequencing, cDNA microarray, and synthetic lethal kinome RNA interference screening. In addition, cell viability, colony formation, and xenograft assays were used to determine the synergistic antitumor effect of T-DXd combinations. RESULTS: We found reduced HER2 expression in patients and amplified DNA repair-related genes in patients after anti-HER2 therapy. Reduced ERBB2 gene amplification in HER2-directed ADC-resistant HER2+ breast cancer cell lines was through DNA damage and epigenetic mechanisms. In HER2-directed ADC-resistant HER2+ breast cancer cell lines, our non-biased RNA interference screening identified the DNA repair pathway as a potential target within the canonical pathways to enhance the efficacy of T-DXd. We validated that the combination of T-DXd with ataxia telangiectasia and Rad3-related inhibitor, elimusertib, led to significant breast cancer cell death in vitro (P < 0.01) and in vivo (P < 0.01) compared to single agents. CONCLUSIONS: The DNA repair pathways contribute to HER2-directed ADC resistance. Our data justify exploring the combination treatment of T-DXd with DNA repair-targeting drugs to treat HER2-directed ADC-resistant HER2+ breast cancer in clinical trials.


Sujet(s)
Tumeurs du sein , Réparation de l'ADN , Résistance aux médicaments antinéoplasiques , Immunoconjugués , Récepteur ErbB-2 , Trastuzumab , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Trastuzumab/pharmacologie , Trastuzumab/usage thérapeutique , Animaux , Immunoconjugués/pharmacologie , Immunoconjugués/usage thérapeutique , Souris , Récepteur ErbB-2/métabolisme , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Camptothécine/analogues et dérivés , Camptothécine/pharmacologie , Camptothécine/usage thérapeutique , Synergie des médicaments
2.
Cancer Med ; 13(16): e70096, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39157928

RÉSUMÉ

BACKGROUND: Sacituzumab govitecan (sacituzumab) emerged as an important agent in metastatic and locally recurrent HER2-negative breast cancer treatment. UGT1A1 polymorphisms have also been shown to predict sacituzumab toxicity. METHODS: In this retrospective study, we sought to evaluate the associations between UGT1A1 status, toxicity, and therapeutic outcomes in sacituzumab recipients with advanced breast cancer who underwent genotype testing for UGT1A1 alleles (N = 68). RESULTS: We found 17 (25%) of our patients to be homozygous for UGT1A1*28 and 24 (35.3%) were heterozygous. Of seven African American patients with triple-negative breast cancer, five were homozygous for UGT1A1*28 and two were heterozygous. Patients with a homozygous UGT1A1*28 genotype were significantly more likely to have treatment terminated because of adverse effects. However, the polymorphism was not associated with treatment discontinuation because of disease progression. CONCLUSION: This retrospective, real-world analysis suggests potential clinical utility in UGT1A1 testing for patients receiving sacituzumab, but future trials are needed to confirm the association between genotypes and treatment outcomes.


Sujet(s)
Anticorps monoclonaux humanisés , Tumeurs du sein , Camptothécine , Évolution de la maladie , Glucuronosyltransferase , Humains , Femelle , Glucuronosyltransferase/génétique , Adulte d'âge moyen , Études rétrospectives , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Camptothécine/analogues et dérivés , Camptothécine/usage thérapeutique , Camptothécine/effets indésirables , Adulte , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Sujet âgé , Polymorphisme génétique , Génotype , Immunoconjugués
3.
Cancer ; 130(S17): 3054-3066, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39092590

RÉSUMÉ

Antibody-drug conjugates (ADCs) have demonstrated effectiveness in treating various cancers, particularly exhibiting specificity in targeting human epidermal growth factor receptor 2 (HER2)-positive breast cancer. Recent advancements in phase 3 clinical trials have broadened current understanding of ADCs, especially trastuzumab deruxtecan, in treating other HER2-expressing malignancies. This expansion of knowledge has led to the US Food and Drug Administration's approval of trastuzumab deruxtecan for HER2-positive and HER2-low breast cancer, HER2-positive gastric cancer, and HER2-mutant nonsmall cell lung cancer. Concurrent with the increasing use of ADCs in oncology, there is growing concern among health care professionals regarding the rise in the incidence of interstitial lung disease or pneumonitis (ILD/p), which is associated with anti-HER2 ADC therapy. Studies on anti-HER2 ADCs have reported varying ILD/p mortality rates. Consequently, it is crucial to establish guidelines for the diagnosis and management of ILD/p in patients receiving anti-HER2 ADC therapy. To this end, a panel of Chinese experts was convened to formulate a strategic approach for the identification and management of ILD/p in patients treated with anti-HER2 ADC therapy. This report presents the expert panel's opinions and recommendations, which are intended to guide the management of ILD/p induced by anti-HER2 ADC therapy in clinical practice.


Sujet(s)
Immunoconjugués , Pneumopathies interstitielles , Récepteur ErbB-2 , Humains , Pneumopathies interstitielles/traitement médicamenteux , Pneumopathies interstitielles/induit chimiquement , Chine , Immunoconjugués/usage thérapeutique , Immunoconjugués/effets indésirables , Pneumopathie infectieuse/traitement médicamenteux , Femelle , Consensus , Trastuzumab/usage thérapeutique , Trastuzumab/effets indésirables , Tumeurs du sein/traitement médicamenteux , Camptothécine/analogues et dérivés
4.
J Transl Med ; 22(1): 766, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39143619

RÉSUMÉ

BACKGROUND: Delta-like ligand 3 (DLL3) is highly expressed on the cell surface of small cell lung cancer (SCLC), one of the most lethal malignancies, but minimally or not in normal tissues, making it an attractive target for SCLC. However, none of the DLL3-targeting antibody-drug conjugates (ADCs) have been approved for SCLC therapy yet. We developed DB-1314, the new anti-DLL3 ADC composed of a novel humanized anti-DLL3 monoclonal antibody (DB131401) conjugated with eight molecules of P1021 (topoisomerase I inhibitor), and described its preclinical profiles. METHODS: The binding epitope for DB131401 and Rovalpituzumab was tested by biolayer interferometry. The binding affinity and specificity of DB-1314 to DLL3 and other homologous proteins were respectively measured by surface plasmon resonance and enzyme-linked immunosorbent assay. Internalization, bystander effects, and antibody-dependent cell-mediated cytotoxicity (ADCC) were assessed by respective assay. DLL3 was quantified by antibodies bound per cell assay and immunohistochemistry. In vitro and in vivo growth inhibition studies were evaluated in SCLC cell lines, and cell line/patient-derived xenograft models. The safety profile was measured in cynomolgus monkeys. RESULTS: DB-1314 induces potent, durable, and dose-dependent antitumor effects in cells in vitro and in cell/patient-derived xenograft models in vivo. The killing activity of DB-1314 mechanically arises from P1021-induced DNA damage, whereby P1021 is delivered and released within tumor cells through DLL3-specific binding and efficient internalization. Bystander effects and ADCC also contribute to the antitumor activity of DB-1314. DB-1314 displays favorable pharmacokinetic and toxicokinetic profiles in rats and cynomolgus monkeys; besides, DB-1314 is well-tolerated at a dose of up to 60 mg/kg in monkeys. CONCLUSIONS: These results suggest that DB-1314 may be a candidate ADC targeting DLL3 for the treatment of DLL3-positive SCLC, supporting further evaluation in the clinical setting.


Sujet(s)
Immunoconjugués , Tumeurs du poumon , Protéines membranaires , Carcinome pulmonaire à petites cellules , Inhibiteurs de la topoisomérase-I , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Carcinome pulmonaire à petites cellules/anatomopathologie , Animaux , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Humains , Inhibiteurs de la topoisomérase-I/pharmacologie , Inhibiteurs de la topoisomérase-I/usage thérapeutique , Lignée cellulaire tumorale , Immunoconjugués/pharmacologie , Immunoconjugués/usage thérapeutique , Protéines membranaires/métabolisme , Protéines membranaires/antagonistes et inhibiteurs , Macaca fascicularis , Tests d'activité antitumorale sur modèle de xénogreffe , Rats , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/antagonistes et inhibiteurs , Femelle , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/pharmacologie , Souris , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux/pharmacologie , Benzodiazépinones
5.
Cancer Treat Rev ; 129: 102806, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39094332

RÉSUMÉ

Hepato-Pancreato-Biliary (HPB) malignancies constitute a highly aggressive group of cancers that have a dismal prognosis. Patients not amenable to curative intent surgical resection are managed with systemic chemotherapy which, however, confers little survival benefit. Antibody-Drug Conjugates (ADCs) are tripartite compounds that merge the intricate selectivity and specificity of monoclonal antibodies with the cytodestructive potency of attached supertoxic payloads. In view of the unmet need for drugs that will enhance the survival rates of HPB cancer patients, the assessment of ADCs for treating HPB malignancies has become the focus of extensive clinical and preclinical investigation, showing encouraging preliminary results. In the current review, we offer a comprehensive overview of the growing body of evidence on ADC approaches tested for HPB malignancies. Starting from a concise discussion of the functional principles of ADCs, we summarize here all available data from preclinical and clinical studies evaluating ADCs in HPB cancers.


Sujet(s)
Tumeurs des voies biliaires , Immunoconjugués , Tumeurs du foie , Tumeurs du pancréas , Humains , Immunoconjugués/usage thérapeutique , Tumeurs des voies biliaires/traitement médicamenteux , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du foie/traitement médicamenteux , Antinéoplasiques immunologiques/usage thérapeutique
6.
JCO Precis Oncol ; 8: e2400179, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39151109

RÉSUMÉ

Antibody-drug conjugates (ADCs) are fusions of therapeutic drugs and antibodies conjugated by a linker, designed to deliver a therapeutic payload to cells expressing the target antigen. By delivering the highly cytotoxic agent directly to cancer cells, ADCs are designed to enhance safety and broaden the therapeutic window. Recently, ADCs have demonstrated promising efficacy in various solid tumors and are rapidly expanding their indications. The prognosis of patients with advanced head and neck squamous cell carcinoma (HNSCC) remains poor, with no new therapeutics since the advent of anti-PD-1 antibodies in 2016, highlighting a critical need for innovative therapies. Recent preliminary results suggest that ADCs could be promising treatment options for HNSCC as they explore a variety of target antigens, payloads, and linkers. However, for successful adaptation of ADCs in the treatment of HNSCC, addressing key challenges such as payload toxicities, antigen heterogeneity, and adaptive resistance will be essential. Current research focused on new ADC structures, including multispecific antibodies and noncytotoxic payloads, and diverse combination approaches, show promise for future advancements.


Sujet(s)
Tumeurs de la tête et du cou , Immunoconjugués , Humains , Immunoconjugués/usage thérapeutique , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/immunologie , Développement de médicament
7.
Nat Commun ; 15(1): 6707, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39112464

RÉSUMÉ

Sacituzumab Govitecan (SG) is an antibody-drug conjugate that has demonstrated efficacy in patients with TROP-2 expressing epithelial cancers. In a xenograft model of intracranial breast cancer, SG inhibited tumor growth and increased mouse survival. We conducted a prospective window-of-opportunity trial (NCT03995706) at the University of Texas Health Science Center at San Antonio to examine the intra-tumoral concentrations and intracranial activity of SG in patients undergoing craniotomy for breast cancer with brain metastases (BCBM) or recurrent glioblastoma (rGBM). We enrolled 25 patients aged ≥18 years diagnosed with BCBM and rGBM to receive a single intravenous dose of SG at 10 mg/kg given one day before resection and continued on days 1 and 8 of 21-day cycles following recovery. The PFS was 8 months and 2 months for BCBM and rGBM cohorts, respectively. The OS was 35.2 months and 9.5 months, respectively. Grade≥3 AE included neutropenia (28%), hypokalemia (8%), seizure (8%), thromboembolic event (8%), urinary tract infection (8%) and muscle weakness of the lower limb (8%). In post-surgical tissue, the median total SN-38 was 249.8 ng/g for BCBM and 104.5 ng/g for rGBM, thus fulfilling the primary endpoint. Biomarker analysis suggests delivery of payload by direct release at target site and that hypoxic changes do not drive indirect release. Secondary endpoint of OS was 35.2 months for the BCBM cohort and 9.5 months for rGBM. Non-planned exploratory endpoint of ORR was 38% for BCBM and 29%, respectively. Exploratory endpoint of Trop-2 expression was observed in 100% of BCBM and 78% of rGBM tumors. In conclusion, SG was found to be well tolerated with adequate penetration into intracranial tumors and promising preliminary activity within the CNS. Trial Registration: Trial (NCT03995706) enrolled at Clinical Trials.gov as Neuro/Sacituzumab Govitecan/Breast Brain Metastasis/Glioblastoma/Ph 0: https://clinicaltrials.gov/study/NCT03995706?cond=NCT03995706 .


Sujet(s)
Anticorps monoclonaux humanisés , Tumeurs du cerveau , Tumeurs du sein , Glioblastome , Immunoconjugués , Récidive tumorale locale , Humains , Femelle , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Adulte d'âge moyen , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie , Adulte , Sujet âgé , Immunoconjugués/usage thérapeutique , Camptothécine/analogues et dérivés , Camptothécine/usage thérapeutique , Études prospectives , Composés hétérocycliques bicycliques/usage thérapeutique , Antigènes néoplasiques/métabolisme , Molécules d'adhérence cellulaire/métabolisme
8.
J Am Chem Soc ; 146(33): 23240-23251, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39113488

RÉSUMÉ

Selective cleavage of amide bonds holds prominent significance by facilitating precise manipulation of biomolecules, with implications spanning from basic research to therapeutic interventions. However, achieving selective cleavage of amide bonds via mild synthetic chemistry routes poses a critical challenge. Here, we report a novel amide bond-cleavage reaction triggered by Na[AuCl4] in mild aqueous conditions, where a crucial cyclization step leads to the formation of a 5-membered ring intermediate that rapidly hydrolyses to release the free amine in high yields. Notably, the reaction exhibits remarkable site-specificity to cleave peptide bonds at the C-terminus of allyl-glycine. The strategic introduction of a leaving group at the allyl position facilitated a dual-release approach through π-acid catalyzed substitution. This reaction was employed for the targeted release of the cytotoxic drug monomethyl auristatin E in combination with an antibody-drug conjugate in cancer cells. Finally, Au-mediated prodrug activation was shown in a colorectal zebrafish xenograft model, leading to a significant increase in apoptosis and tumor shrinkage. Our findings reveal a novel metal-based cleavable reaction expanding the utility of Au complexes beyond catalysis to encompass bond-cleavage reactions for cancer therapy.


Sujet(s)
Amides , Antinéoplasiques , Promédicaments , Danio zébré , Animaux , Amides/composition chimique , Humains , Promédicaments/composition chimique , Promédicaments/synthèse chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Antinéoplasiques/pharmacologie , Oligopeptides/composition chimique , Lignée cellulaire tumorale , Or/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Structure moléculaire , Immunoconjugués/composition chimique
9.
J Labelled Comp Radiopharm ; 67(10): 341-348, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39107085

RÉSUMÉ

Radioimmunoconjugates (RICs) composed of tumor-targeting monoclonal antibodies and radionuclides have been developed for diagnostic and therapeutic application. A new radiolabeling method using microfluidic devices is expected to facilitate simpler and more rapid synthesis of RICs. In the microfluidic method, microfluidic chips can promote the reaction between reactants by mixing them efficiently, and pumping systems enable automated synthesis. In this study, we synthesized RICs by the pre-labeling method, in which the radiometal is coordinated to the chelator and then the radiolabeled chelator is incorporated into the antibodies, using microfluidic devices for the first time. As a result of examining the reaction parameters including the material of mixing units, reaction temperature, and flow rate, RICs with radiochemical purity (RCP) exceeding 90% were obtained. These high-purity RICs were successfully synthesized without any purification simply by pumping three solutions of a chelating agent, radiometal, and antibody into microfluidic devices. Under the same conditions, the RCP of RICs labeled by conventional methods was below 50%. These findings indicate the utility of microfluidic devices for automatic and rapid synthesis of high-quality RICs.


Sujet(s)
Immunoconjugués , Marquage isotopique , Immunoconjugués/composition chimique , Techniques d'analyse microfluidique/méthodes , Techniques d'analyse microfluidique/instrumentation , Anticorps monoclonaux/composition chimique , Chélateurs/composition chimique , Laboratoires sur puces , Automatisation , Radiopharmaceutiques/composition chimique , Radiopharmaceutiques/synthèse chimique
10.
Bioconjug Chem ; 35(8): 1142-1147, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39129506

RÉSUMÉ

Ferritin-drug conjugates (FDCs) and antibody-drug conjugates (ADCs) respectively represent the innovative and traditional mainstream approaches in drug delivery systems, each offering unique advantages and challenges. This viewpoint delves into the evolving landscape of drug delivery technologies, specifically focusing on FDCs and ADCs. Each method exhibits unique advantages and inherent challenges, shaping their roles in therapeutic applications. The article provides a comparative analysis of two delivery systems, FDCs and ADCs, in terms of targeting accuracy, drug loading capacity, and the nature of the payload itself. This comparison offers valuable insights into the distinct advantages and disadvantages associated with each system, enabling a clearer understanding of their potential applications and limitations in therapeutic contexts. This analysis is crucial for optimizing the use of these delivery systems across varying medical contexts, offering a comprehensive overview of their impact on the field of drug delivery.


Sujet(s)
Systèmes de délivrance de médicaments , Ferritines , Immunoconjugués , Immunoconjugués/composition chimique , Humains , Systèmes de délivrance de médicaments/méthodes , Ferritines/composition chimique , Animaux , Préparations pharmaceutiques/composition chimique
11.
J Ovarian Res ; 17(1): 161, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39118097

RÉSUMÉ

Ovarian cancer stands as the deadliest gynecologic malignancy, responsible for nearly 65% of all gynecologic cancer-related deaths. The challenges in early detection and diagnosis, coupled with the widespread intraperitoneal spread of cancer cells and resistance to chemotherapy, contribute significantly to the high mortality rate of this disease. Due to the absence of specific symptoms and the lack of effective screening methods, most ovarian cancer cases are diagnosed at advanced stages. While chemotherapy is a common treatment, it often leads to tumor recurrence, necessitating further interventions. In recent years, antibody-drug conjugates (ADCs) have emerged as a valuable tool in targeted cancer therapy. These complex biotherapeutics combine an antibody that specifically targets tumor specific/associated antigen(s) with a high potency anti-cancer drug through a linker, offering a promising approach for ovarian cancer treatment. The identification of molecular targets in various human tumors has paved the way for the development of targeted therapies, with ADCs being at the forefront of this innovation. By delivering cytotoxic agents directly to tumors and metastatic lesions, ADCs show potential in managing chemo-resistant ovarian cancers. Mucins such as MUC16, MUC13, and MUC1 have shown significantly higher expression in ovarian tumors as compared to normal and/or benign samples, thus have become promising targets for ADC generation. While traditional markers are limited by their elevated levels in non-cancerous conditions, mucins offer a new possibility for targeted treatment in ovarian cancer. This review comprehensively described the potential of mucins for the generation of ADC therapy, highlighting their importance in the quest to improve the outcome of ovarian cancer patients.


Sujet(s)
Immunoconjugués , Mucines , Tumeurs de l'ovaire , Humains , Femelle , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/métabolisme , Immunoconjugués/usage thérapeutique , Immunoconjugués/pharmacologie , Mucines/métabolisme , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Animaux
12.
ACS Appl Mater Interfaces ; 16(32): 41939-41948, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39093637

RÉSUMÉ

Antibody-drug conjugates (ADCs), which combine the precise targeting capabilities of antibodies with the powerful cytotoxicity of small-molecule drugs, have evolved into a promising approach for tumor treatment. However, the traditional covalent coupling method requires the design of a specific linker tailored to the properties of the small-molecule drugs, which greatly limits the development of ADCs and the range of drugs that can be used. Herein, a novel type of antibody-calixarene drug conjugates (ACDCs) that function similarly to ADCs by delivering drugs to their targets using antibodies but without the requirement of covalent conjugation of the drugs with antibodies is presented. By replacement of conventional linkers with supramolecular linkers, the ACDCs can load various chemotherapeutic drugs through host-guest interactions. Furthermore, ACDCs are readily reduced upon reaching the hypoxic microenvironment, resulting in rapid release of the drugs. With this precise drug encapsulation and controlled release mechanism, ACDCs deliver drugs to tumor tissues effectively and achieve a significantly enhanced antitumor effect. Considering that the ACDCs can be easily prepared by combining antibody-calixarene conjugates derived from tumor-targeting antibodies with various small-molecule drugs, ACDCs may provide a promising platform technology to accelerate ADC development and thus improve the therapeutic efficacy of chemotherapy.


Sujet(s)
Antinéoplasiques , Calixarènes , Immunoconjugués , Calixarènes/composition chimique , Immunoconjugués/composition chimique , Immunoconjugués/pharmacologie , Immunoconjugués/usage thérapeutique , Humains , Animaux , Souris , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Tumeurs/traitement médicamenteux , Systèmes de délivrance de médicaments , Souris de lignée BALB C , Vecteurs de médicaments/composition chimique , Femelle , Libération de médicament
13.
Anticancer Drugs ; 35(8): 769-773, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39115060

RÉSUMÉ

Trastuzumab deruxtecan (T-DXd) is a novel anti-HER2 antibody-drug conjugate formed by the combination of trastuzumab and deruxtecan. It is used in human epidermal growth factor 2 receptor (HER2) mutant breast, stomach and colorectal cancers as well as non-small cell lung cancer (NSCLC). The 58-year-old denovo metastatic NSCLC patient we will discuss here progressed with newly developing brain metastasis under first-line carboplatin/paclitaxel treatment. After next generation sequencing revealed a mutation in the ERBB2 gene located in exon 20, we administered T-DXd to our patient. While a significant improvement was observed in the clinical condition of the patient after one course of treatment, brain metastases were found to be in complete response in control screening after four courses of treatment. Systemic screening with PET/computed tomography showed nearly complete regression of the primary lesion, metastatic lymphadenopathies, and surrenal metastases. T-DXd may be successfully used in HER2 mutant metastatic NSCLC patients. In addition, it can also be successfully used in patients with central nervous system metastases with or without cranial radiotherapy.


Sujet(s)
Tumeurs du cerveau , Camptothécine , Carcinome pulmonaire non à petites cellules , Exons , Tumeurs du poumon , Récepteur ErbB-2 , Trastuzumab , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Trastuzumab/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Adulte d'âge moyen , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Récepteur ErbB-2/génétique , Camptothécine/analogues et dérivés , Camptothécine/usage thérapeutique , Immunoconjugués/usage thérapeutique , Mutation , Femelle , Mâle , Antinéoplasiques immunologiques/usage thérapeutique
14.
MAbs ; 16(1): 2387240, 2024.
Article de Anglais | MEDLINE | ID: mdl-39113562

RÉSUMÉ

Prostate stem cell antigen (PSCA) is expressed in all stages of prostate cancer, including in advanced androgen-independent tumors and bone metastasis. PSCA may associate with prostate carcinogenesis and lineage plasticity in prostate cancer. PSCA is also a promising theranostic marker for a variety of other solid tumors, including pancreatic adenocarcinoma and renal cell carcinoma. Here, we identified a novel fully human PSCA antibody using phage display methodology. The structure-based affinity maturation yielded a high-affinity binder, F12, which is highly specific and does not bind to 6,000 human membrane proteins based on a membrane proteome array assay. F12 targets PSCA amino acids 63-69 as tested by the peptide scanning microarray, and it cross-reacts with the murine PSCA. IgG1 F12 efficiently internalizes into PSCA-expressing tumor cells. The antimitotic reagent monomethyl auristatin E (MMAE)-conjugated IgG1 F12 (ADC, F12-MMAE) exhibits dose-dependent efficacy and specificity in a human prostate cancer PC-3-PSCA xenograft NSG mouse model. This is a first reported ADC based on a fully human PSCA antibody and MMAE that is characterized in a xenograft murine model, which warrants further optimizations and investigations in additional preclinical tumor models, including prostate and other solid tumors.


Sujet(s)
Antigènes néoplasiques , Protéines liées au GPI , Immunoconjugués , Protéines tumorales , Tumeurs de la prostate , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Mâle , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/immunologie , Immunoconjugués/pharmacologie , Animaux , Antigènes néoplasiques/immunologie , Souris , Protéines liées au GPI/immunologie , Protéines tumorales/immunologie , Protéines tumorales/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Oligopeptides/immunologie , Oligopeptides/pharmacologie , Immunoglobuline G/immunologie , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/pharmacologie
15.
PLoS One ; 19(8): e0308247, 2024.
Article de Anglais | MEDLINE | ID: mdl-39088571

RÉSUMÉ

BACKGROUND: Diffuse large B-cell lymphoma (DLBCL) is an aggressive non-Hodgkin lymphoma subtype with a significant relapse rate and poor prognosis in relapsed/refractory (R/R) patients. Polatuzumab vedotin in combination with bendamustine and rituximab (Pola-BR) has demonstrated promising efficacy and safety as salvage therapy for R/R DLBCL. This systematic review protocol aims to comprehensively evaluate the efficacy of Pola-BR for the treatment of R/R DLBCL by synthesizing data from relevant randomized controlled trials. METHODS: This protocol details the eligibility criteria, search strategy, study selection, data extraction, and analysis methods for the systematic review. Randomized controlled trials comparing Pola-BR with other interventions for R/R DLBCL will be included. The primary endpoint is overall survival, with secondary endpoints being progression-free survival and incidence of adverse events. A comprehensive search will be conducted across databases such as Medline/PubMed, Cochrane Library, Web of Science, Scopus, EMBASE, ProQuest, EU Clinical Trials Register, WHO International Clinical Trials Registry Platform (ICTRP), and ClinicalTrials.gov from the January 2000 to April 2024. To assess the potential risk of bias, the Cochrane Risk of Bias 1 tool will be used. Data synthesis will utilize fixed-effect or random-effects models, and subgroup and meta-regression analyses will examine heterogeneity. Additionally, publication bias and sensitivity analyses will be performed, and the GRADE approach will be applied to assess the certainty of the evidence. CONCLUSION: This systematic review and meta-analysis protocol provides a rigorous framework for evaluating the efficacy of Pola-BR in the treatment of R/R DLBCL. The results will inform clinical decision-making and guideline development, addressing the unmet need for effective and tolerable treatments for this challenging patient population. Potential limitations and biases will be acknowledged, and future research directions will be discussed.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Chlorhydrate de bendamustine , Lymphome B diffus à grandes cellules , Rituximab , Revues systématiques comme sujet , Humains , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/anatomopathologie , Rituximab/administration et posologie , Rituximab/usage thérapeutique , Chlorhydrate de bendamustine/administration et posologie , Chlorhydrate de bendamustine/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Récidive tumorale locale/traitement médicamenteux , Immunoconjugués/usage thérapeutique , Immunoconjugués/administration et posologie , Immunoconjugués/effets indésirables , Essais contrôlés randomisés comme sujet , Anticorps monoclonaux
16.
Clin Cancer Res ; 30(15): 3179-3188, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39086310

RÉSUMÉ

PURPOSE: Human trophoblast cell surface antigen 2 (Trop-2) is a protein highly expressed in urothelial cancer (UC). Sacituzumab govitecan (SG) is a Trop-2-directed antibody drug conjugate with a hydrolysable linker and a potent SN-38 payload. This study explored Trop-2 expression in tumors treated with SG in cohorts 1 to 3 (C1-3) from the TROPHY-U-01 study and evaluated whether efficacy was associated with Trop-2 expression. PATIENTS AND METHODS: TROPHY-U-01 (NCT03547973) is an open-label phase II study that assessed the efficacy and safety of SG (alone or in combinations) in patients with unresectable locally advanced or metastatic UC (mUC). Archival tumor samples collected at enrollment for C1-3 were analyzed for Trop-2 membrane expression by considering histological scores (H-scores; scale 0-300) and the percentage of membrane positive tumor cells at low magnification (4×). The association of Trop-2 with clinical endpoints [objective response rate (ORR), progression-free survival (PFS), and overall survival (OS)] was evaluated. RESULTS: In C1-3, tissue was collected from 158 (82%) of 192 treated patients, and 146 (76%) had evaluable Trop-2 data. Trop-2 was highly expressed in tumor samples. The median [interquartile range (IQR)] Trop-2 H-score was 215 (180-246), and the median (IQR) percentage of membrane positive tumor cells was 91% (80-98). Trop-2 expression at any level was observed in 98% of patients. Furthermore, ORR, PFS, and OS benefits were observed across all Trop-2 expression levels. CONCLUSIONS: Trop-2 protein is highly expressed in UC, as confirmed by examining tumors from patients enrolled in the TROPHY-U-01 trial. The results indicate that SG demonstrates efficacy in mUC across Trop-2 expression levels.


Sujet(s)
Anticorps monoclonaux humanisés , Antigènes néoplasiques , Camptothécine , Molécules d'adhérence cellulaire , Immunoconjugués , Humains , Molécules d'adhérence cellulaire/métabolisme , Anticorps monoclonaux humanisés/usage thérapeutique , Femelle , Mâle , Sujet âgé , Adulte d'âge moyen , Camptothécine/analogues et dérivés , Camptothécine/usage thérapeutique , Immunoconjugués/usage thérapeutique , Sujet âgé de 80 ans ou plus , Adulte , Marqueurs biologiques tumoraux/métabolisme , Tumeurs urologiques/traitement médicamenteux , Tumeurs urologiques/anatomopathologie , Tumeurs urologiques/mortalité , Tumeurs urologiques/métabolisme , Résultat thérapeutique , Stadification tumorale
17.
Theranostics ; 14(9): 3674-3692, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948057

RÉSUMÉ

Trophoblast cell surface antigen 2 (Trop2) is overexpressed in a range of solid tumors and participants in multiple oncogenic signaling pathways, making it an attractive therapeutic target. In the past decade, the rapid development of various Trop2-targeted therapies, notably marked by the advent of the antibody-drug conjugate (ADC), revolutionized the outcome for patients facing Trop2-positive tumors with limited treatment opinions, such as triple-negative breast cancer (TNBC). This review provides a comprehensive summary of advances in Trop2-targeted therapies, including ADCs, antibodies, multispecific agents, immunotherapy, cancer vaccines, and small molecular inhibitors, along with in-depth discussions on their designs, mechanisms of action (MOAs), and limitations. Additionally, we emphasize the clinical research progress of these emerging Trop2-targeted agents, focusing on their clinical application and therapeutic efficacy against tumors. Furthermore, we propose directions for future research, such as enhancing our understanding of Trop2's structure and biology, exploring the best combination strategies, and tailoring precision treatment based on Trop2 testing methodologies.


Sujet(s)
Antigènes néoplasiques , Molécules d'adhérence cellulaire , Immunoconjugués , Thérapie moléculaire ciblée , Tumeurs , Humains , Antigènes néoplasiques/immunologie , Molécules d'adhérence cellulaire/antagonistes et inhibiteurs , Molécules d'adhérence cellulaire/métabolisme , Immunoconjugués/usage thérapeutique , Immunoconjugués/pharmacologie , Thérapie moléculaire ciblée/méthodes , Tumeurs/traitement médicamenteux , Tumeurs/thérapie , Immunothérapie/méthodes , Animaux , Vaccins anticancéreux/usage thérapeutique
18.
Signal Transduct Target Ther ; 9(1): 175, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39013849

RÉSUMÉ

Traditional therapeutic approaches such as chemotherapy and radiation therapy have burdened cancer patients with onerous physical and psychological challenges. Encouragingly, the landscape of tumor treatment has undergone a comprehensive and remarkable transformation. Emerging as fervently pursued modalities are small molecule targeted agents, antibody-drug conjugates (ADCs), cell-based therapies, and gene therapy. These cutting-edge treatment modalities not only afford personalized and precise tumor targeting, but also provide patients with enhanced therapeutic comfort and the potential to impede disease progression. Nonetheless, it is acknowledged that these therapeutic strategies still harbour untapped potential for further advancement. Gaining a comprehensive understanding of the merits and limitations of these treatment modalities holds the promise of offering novel perspectives for clinical practice and foundational research endeavours. In this review, we discussed the different treatment modalities, including small molecule targeted drugs, peptide drugs, antibody drugs, cell therapy, and gene therapy. It will provide a detailed explanation of each method, addressing their status of development, clinical challenges, and potential solutions. The aim is to assist clinicians and researchers in gaining a deeper understanding of these diverse treatment options, enabling them to carry out effective treatment and advance their research more efficiently.


Sujet(s)
Thérapie génétique , Tumeurs , Humains , Tumeurs/thérapie , Tumeurs/génétique , Tumeurs/traitement médicamenteux , Immunoconjugués/usage thérapeutique , Thérapie moléculaire ciblée , Thérapie cellulaire et tissulaire , Antinéoplasiques/usage thérapeutique
19.
Zhongguo Fei Ai Za Zhi ; 27(6): 431-440, 2024 Jun 20.
Article de Chinois | MEDLINE | ID: mdl-39026494

RÉSUMÉ

Lung cancer is the most common malignant tumor and the second most common malignant tumor in terms of mortality in the world. Non-small cell lung cancer (NSCLC) is the most common pathological type of lung cancer. Currently, the first-line standard treatment for advanced NSCLC is immunotherapy and targeted therapy. Although these treatments prolong the survival of patients, acquired drug resistance is still inevitable. Antibody-drug conjugates (ADCs) are a new type of anti-tumor drug made by coupling cytotoxic payloads to specific monoclonal antibodies via linkers. Compared with chemotherapy drugs, ADCs have the advantages of accurate recognition, local release, and high patient tolerance. In recent years, they have shown good clinical benefits in the treatment of NSCLC. This article provides an overview of the mechanism of action of ADCs, clinical studies progress in advanced NSCLC, and existing problems and challenges.
.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Immunoconjugués , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/immunologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Immunoconjugués/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Animaux , Anticorps monoclonaux/usage thérapeutique
20.
Taiwan J Obstet Gynecol ; 63(4): 471-478, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39004472

RÉSUMÉ

Platinum-resistant ovarian cancer (PROC) refers to disease progression within 6 months after the completion of platinum-based chemotherapy. Historically, treatment options for PROC were limited with a poor prognosis and non-platinum single agent plus bevacizumab has been the mainstay of treatment. Fortunately, there have been notable advancements in recent years, leading to an advance in treatment paradigms for this challenging disease. Various combinations of chemotherapy, targeted agents such as poly (ADP-ribose) polymerase (PARP) inhibitors, and immunotherapy are being explored for an improved treatment outcome. Antibody-drug conjugates targeting folate receptor alpha, which deliver a cytotoxic payload directly to cancer cells, have emerged as a promising therapeutic approach for PROC. WEE1 inhibitors, such as adavosertib, function by inhibiting the WEE1 kinase activity, leading to premature entry of a cell into mitosis phase and thus increased DNA damage. It has been observed that cancer cells with TP53 mutations may be more sensitive to WEE1 inhibitors. Biomarker testing such as analysis of the expression level of folate receptor alpha or mutation in TP53 may be applicable for identifying patients who are more likely to respond to the specific therapy, enabling a more personalized treatment approach. This overview summarizes key clinical findings on the efficacy and safety of theses novel biomarker-driven therapeutic approaches.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Tumeurs de l'ovaire , Humains , Femelle , Tumeurs de l'ovaire/traitement médicamenteux , Protein-tyrosine kinases/antagonistes et inhibiteurs , Antinéoplasiques/usage thérapeutique , Récepteur-1 des folates/antagonistes et inhibiteurs , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Immunothérapie/méthodes , Immunoconjugués/usage thérapeutique , Pyrazoles/usage thérapeutique , Protéine p53 suppresseur de tumeur , Pyrimidinones/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE