Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.164
Filter
1.
J Immunother Cancer ; 12(10)2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39357981

ABSTRACT

BACKGROUND: Antibody-drug conjugates (ADCs) offer a promising approach, combining monoclonal antibodies with chemotherapeutic drugs to target cancer cells effectively while minimizing toxicity. METHODS: This study examined the therapeutic efficacy and potential mechanisms of a bispecific ADC (BsADC) in laryngeal squamous cell carcinoma. This BsADC selectively targets the immune checkpoints programmed cell death ligand-1 (PD-L1) and B7-H3, and the precise delivery of the small-molecule toxin monomethyl auristatin E. RESULTS: Our findings demonstrated that the BsADC outperformed its bispecific antibody and PD-L1 or B7-H3 ADC counterparts, particularly in terms of in vitro/in vivo tumor cytotoxicity, demonstrating remarkable immune cytotoxicity. Additionally, we observed potent activation of tumor-specific immunity and significant induction of markers of immunogenic cell death (ICD) and potential endoplasmic reticulum stress. CONCLUSION: In conclusion, this novel BsADC, through immune checkpoint inhibition and promotion of ICD, amplified durable tumor immune cytotoxicity, providing novel insights and potential avenues for future cancer treatments and overcoming resistance.


Subject(s)
Antibodies, Bispecific , B7 Antigens , B7-H1 Antigen , Immunoconjugates , Humans , Animals , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Mice , B7-H1 Antigen/antagonists & inhibitors , B7 Antigens/antagonists & inhibitors , Cell Line, Tumor , Female
2.
J Ovarian Res ; 17(1): 196, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39367438

ABSTRACT

Platinum is a key component of ovarian cancer systemic therapy. However, most patients will eventually face a recurrence, leading to chemotherapy resistance, especially against platinum. For individuals with platinum-resistant ovarian cancer (PROC), treatment options are limited, and their survival prospects are grim. The emergence of antibody-drug conjugates (ADCs) shows promises as a future treatment for PROC. This review synthesizes current research on the effectiveness of ADCs in treating PROC. It encapsulates the advancements and clinical trials of novel ADCs that target specific antigens such as Folate Receptor alpha (FRα), MUC16, NaPi2b, Mesothelin, Dipeptidase 3(DPEP3), and human epidermal growth factor receptor 2 (HER2), as well as tissue factor, highlighting their potential anti-tumor efficacy and used in combination with other therapies. The ADCs landscape in ovarian cancer therapeutics is swiftly evolving, promising more potent and efficacious treatment avenues.


Subject(s)
Drug Resistance, Neoplasm , Immunoconjugates , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/drug therapy , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology , Platinum/therapeutic use , Platinum/pharmacology , Antineoplastic Agents/therapeutic use
3.
Cancer Discov ; 14(9): 1577-1580, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39228298

ABSTRACT

Polatuzumab vedotin, an antibody-drug conjugate targeting CD79B, is the first new drug approved for first-line therapy of diffuse large B-cell lymphoma in more than two decades, although factors determining treatment responses to polatuzumab vedotin remain unknown. Two new studies identified central mechanisms of lower sensitivity, namely reduced accessibility of the CD79B epitope through N-linked glycosylation of CD79B and lower CD79B surface expression levels due to the activity of the KLHL6 E3 ligase. See related article by Corcoran et al., p. 1653 (6) See related article by Meriranta et al. (7).


Subject(s)
Immunoconjugates , Humans , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology , CD79 Antigens , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, B-Cell/drug therapy
4.
Cell Rep Med ; 5(9): 101710, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39232496

ABSTRACT

Claudin18.2 has been recently recognized as a potential therapeutic target for gastric/gastroesophageal junction or pancreatic cancer. Here, we develop a Claudin18.2-directed antibody-drug conjugate (ADC), CMG901, with a potent microtubule-targeting agent MMAE (monomethyl auristatin E) and evaluate its preclinical profiles. In vitro studies show that CMG901 binds specifically to Claudin18.2 on the cell surface and kills tumor cells through direct cytotoxicity, antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and bystander killing activity. In vivo pharmacological studies show significant antitumor activity in patient-derived xenograft (PDX) models. Toxicity studies show that the major adverse effects related to CMG901 are reversible hematopoietic changes attributed to MMAE. The highest non-severely toxic dose (HNSTD) is 6 mg/kg in cynomolgus monkeys and 10 mg/kg in rats once every 3 weeks. CMG901's favorable preclinical profile supports its entry into the human clinical study. CMG901 is currently under phase 3 investigation in patients with advanced gastric/gastroesophageal junction adenocarcinoma expressing Claudin18.2 (NCT06346392).


Subject(s)
Claudins , Immunoconjugates , Animals , Female , Humans , Male , Mice , Rats , Antibody-Dependent Cell Cytotoxicity/drug effects , Cell Line, Tumor , Claudins/metabolism , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Macaca fascicularis , Neoplasms/drug therapy , Neoplasms/pathology , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays , Clinical Trials, Phase III as Topic
5.
Sci Rep ; 14(1): 21728, 2024 09 17.
Article in English | MEDLINE | ID: mdl-39289512

ABSTRACT

This study aimed to design a VEGFR-targeting peptide-drug conjugate with the ability to decrease tumor burden and suppress tumor angiogenesis, and to further evaluate the therapeutic effect of anti-PD-1 antibody in HCC therapy. A VEGFR-targeting peptide VEGF125 - 136 (QR) was conjugated with a lytic peptide (KLU) to form a peptide-drug conjugate QR-KLU. And the efficacy of QR-KLU in combination with anti-PD-1 antibody for HCC therapy in vivo and in vitro were evaluated. QR-KLU inhibited the proliferation and migration of mouse HCC cell line (Hepa1-6) cells under normoxic and hypoxic conditions in a dose-dependent manner. In the subcutaneous Hepa1-6 tumor model, QR-KLU combined with the anti-PD-1 antibody substantially inhibited tumor growth, promoted tumor necrosis, and prolonged the survival time of tumor-bearing mice. QR-KLU substantially inhibited hypoxia-induced expression of VEGF, promoted tumor vascular normalization, and increased cluster of differentiation 8+ (CD8+) T cell infiltration in the tumor. In addition, QR-KLU and anti-PD-1 antibody demonstrated a strong synergistic effect in promoting the activation of intratumoral CD8+ T cells, reducing the expression of immune-inhibitory factors, and increasing the expression of immune-stimulatory factors. This study proposed a novel approach for enhancing the efficacy of anti-PD-1 antibody using a VEGFR-targeting peptide-drug conjugate in HCC therapy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Programmed Cell Death 1 Receptor , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Animals , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Cell Line, Tumor , Receptors, Vascular Endothelial Growth Factor/metabolism , Cell Proliferation/drug effects , Humans , Peptides/pharmacology , Peptides/chemistry , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/immunology , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Immunoconjugates/chemistry
7.
Int J Mol Sci ; 25(17)2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39273642

ABSTRACT

The standard of care for advanced or metastatic urothelial carcinoma (mUC) was historically identified with platinum-based chemotherapy. Thanks to the advances in biological and genetic knowledge and technologies, new therapeutic agents have emerged in this setting recently: the immune checkpoint inhibitors and the fibroblast growth factor receptor inhibitors as the target therapy for patients harboring alterations in the fibroblast growth factor receptor (FGFR) pathway. However, chasing a tumor's tendency to recur and progress, a new class of agents has more recently entered the scene, with promising results. Antibody-drug conjugates (ADCs) are in fact the latest addition, with enfortumab vedotin being the first to receive accelerated approval by the U.S. Food and Drug Administration in December 2019, followed by sacituzumab govitecan. Many other ADCs are still under investigation. ADCs undoubtedly represent the new frontier, with the potential of transforming the management of mUC treatment in the future. Therefore, we reviewed the landscape of mUC treatment options, giving an insight into the molecular basis and mechanisms, and evaluating new therapeutic strategies in the perspective of more and more personalized treatments.


Subject(s)
Immunoconjugates , Humans , Immunoconjugates/therapeutic use , Neoplasm Metastasis , Immune Checkpoint Inhibitors/therapeutic use , Urologic Neoplasms/drug therapy , Urologic Neoplasms/pathology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy/methods , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Antibodies, Monoclonal/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Camptothecin/analogs & derivatives
8.
J Hematol Oncol ; 17(1): 88, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342286

ABSTRACT

Given its nearly ubiquitous expression on plasma cells and limited expression on essential normal tissue, the G protein-coupled receptor class C group 5 member D (GPRC5D) presents a promising opportunity for utilization as an immunotherapy target in multiple myeloma (MM). The therapeutic strategies targeting GPRC5D, such as bispecific antibodies (BsAbs), chimeric antigen receptor (CAR) T cells, and antibody-drug conjugates (ADCs), have been prominently emphasized in relapsed/refractory MM (R/R MM) in recent years. Further clinical trials are necessary to confirm the long-term efficacy of GPRC5D-targeting immunotherapies alone, explore their potentials co-targeting with other specific antigens, or investigate their combinations with existing treatments to overcome MM resistance. This review provides an overview of current research progress in GPRC5D, encompassing its biological characteristics and translational journey from laboratory to clinical application.


Subject(s)
Multiple Myeloma , Receptors, G-Protein-Coupled , Multiple Myeloma/drug therapy , Multiple Myeloma/therapy , Multiple Myeloma/immunology , Humans , Receptors, G-Protein-Coupled/antagonists & inhibitors , Immunoconjugates/therapeutic use , Antibodies, Bispecific/therapeutic use , Animals , Immunotherapy/methods , Molecular Targeted Therapy/methods
9.
Curr Oncol ; 31(9): 5694-5708, 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39330050

ABSTRACT

Sacituzumab Govitecan (SG) is an antibody-drug conjugate (ADC) comprised of an anti-Trop-2 IgG1 molecule conjugated to SN-38, the active metabolite of irinotecan, via a pH-sensitive hydrolysable linker. As a result of recent Canadian funding for SG in advanced hormone receptor (HR)-positive breast cancer and triple-negative breast cancer (TNBC), experience with using SG and managing adverse events (AEs) has grown. This review presents a summary of evidence and adverse event recommendations derived from Canadian experience, with SG use in metastatic TNBC for extrapolation and guidance in all indicated settings. SG is dosed at 10 mg/kg on day 1 and day 8 of a 21-day cycle. Compared to treatment of physicians' choice (TPC) the phase III ASCENT and TROPiCS-02 studies demonstrated favorable survival data in unresectable locally advanced or metastatic TNBC and HR-positive HER2 negative metastatic breast cancer, respectively. The most common AEs were neutropenia, diarrhea, nausea, fatigue, alopecia, and anemia. This review outlines AE management recommendations for SG based on clinical trial protocols and Canadian guidelines, incorporating treatment delay, dose reductions, and the use of prophylactic and supportive medications.


Subject(s)
Antibodies, Monoclonal, Humanized , Camptothecin , Immunoconjugates , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Canada , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Immunoconjugates/therapeutic use , Female , Neoplasm Metastasis , Antineoplastic Agents/therapeutic use
10.
J Med Chem ; 67(18): 15996-16001, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39231796

ABSTRACT

Herein, we discuss advancements in the field of a unique class of antibody-drug conjugates (ADCs) named molecular glue-antibody conjugate (MAC). ADCs traditionally employ cytotoxic agents as payloads, and this approach has been used in all approved ADCs to treat cancer. Complementary to this approach, proteolysis targeting chimera (PROTAC) degrader antibody conjugates (DACs) provide a unique opportunity to deliver these bifunctional agents to tumors by using antibodies as a delivery mechanism to overcome the bioavailability issues encountered by PROTAC payloads. Recently, a cereblon binding monovalent degrader called molecular glues has been used in new ADCs that we have coined the term molecular-glue antibody conjugates (MACs). In this article, we intend to review advancements made in the field of targeted delivery of cereblon-based molecular glue degraders.


Subject(s)
Immunoconjugates , Humans , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Animals , Proteolysis/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing
11.
Urologie ; 63(10): 1002-1010, 2024 Oct.
Article in German | MEDLINE | ID: mdl-39235511

ABSTRACT

BACKGROUND: Patients with locally advanced or metastatic urothelial carcinoma face a poor prognosis. Standard first-line treatment involves platinum-based combinations followed by avelumab maintenance therapy. Follow-up therapies include enfortumab vedotin, vinflunine, and taxanes. OBJECTIVE: To analyze new drug combinations in first-line and follow-up treatment for metastatic urothelial carcinoma concerning their clinical relevance, toxicities, and novel treatment sequences. MATERIALS AND METHODS: Analysis of new study data from EV-302/KN-A39 (enfortumab vedotin and pembrolizumab) and CheckMate-901 (nivolumab and gemcitabine-cisplatin) for untreated metastatic patients as well as TROPHY-U-01 (sacituzumab govitecan) and THOR (erdafitinib) for later lines. RESULTS: The new standard in first-line treatment for metastatic urothelial carcinoma is the combination of enfortumab vedotin and pembrolizumab. For cisplatin-eligible patients with contraindications to enfortumab vedotin, the combination of nivolumab and gemcitabine-cisplatin offers an alternative. Erdafitinib presents a new biomarker-based option in the follow-up treatment of metastatic urothelial carcinoma. CONCLUSION: These novel combinations are revolutionizing the treatment standard for metastatic urothelial carcinoma and necessitate a new approach to managing side effects.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Transitional Cell , Immunoconjugates , Humans , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/secondary , Carcinoma, Transitional Cell/pathology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/adverse effects , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology , Immunoconjugates/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Gemcitabine , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Deoxycytidine/administration & dosage , Neoplasm Metastasis , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Urologic Neoplasms/drug therapy , Urologic Neoplasms/pathology , Nivolumab/therapeutic use , Nivolumab/administration & dosage , Nivolumab/pharmacology , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Camptothecin/analogs & derivatives , Pyrazoles , Quinoxalines
12.
ESMO Open ; 9(9): 103699, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39255534

ABSTRACT

BACKGROUND: Trastuzumab deruxtecan (T-DXd) is approved for human epidermal growth factor receptor 2 (HER2)-positive and HER2-low advanced breast cancer (ABC). T-DXd has shown encouraging intracranial activity in HER2-positive ABC patients with stable or active brain metastases (BMs); however, its efficacy in patients with HER2-low ABC with BMs is not well established yet. METHODS: DEBBRAH is a single-arm, five-cohort, phase II study evaluating T-DXd in patients with central nervous system involvement from HER2-positive and HER2-low ABC. Here, we report results from patients with heavily pretreated HER2-low ABC and active BMs, enrolled in cohorts 2 (n = 6, asymptomatic untreated BMs) and 4 (n = 6, progressing BMs after local therapy). Patients received 5.4 mg/kg T-DXd intravenously once every 21 days. The primary endpoint was intracranial objective response rate per Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM) for both cohorts. RESULTS: Intracranial objective response rate per RANO-BM was 50.0% [3/6 patients; 95% confidence interval (CI) 11.8% to 88.2%] and 33.3% [2/6 patients; 95% CI 4.3% to 77.7%; P = 0.033 (one-sided)] in cohorts 2 and 4, respectively. All responders had partial responses. Median time to intracranial response was 2.3 months (range, 1.5-4.0 months) and median duration of intracranial response was 7.2 months (range, 2.8-16.8 months). Median progression-free survival per RECIST v.1.1. was 5.4 months (95% CI 4.1-10.0 months). Treatment-emergent adverse events occurred in all patients included (16.7% grade 3). Three patients (25.0%) had grade 1 interstitial lung disease/pneumonitis. CONCLUSIONS: T-DXd demonstrated promising intracranial activity in pretreated HER2-low ABC patients with active BMs. Further studies are needed to validate these results in larger cohorts. This trial is registered with ClinicalTrials.gov, NCT04420598.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Camptothecin , Receptor, ErbB-2 , Trastuzumab , Humans , Female , Trastuzumab/therapeutic use , Trastuzumab/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Brain Neoplasms/secondary , Brain Neoplasms/drug therapy , Middle Aged , Receptor, ErbB-2/metabolism , Aged , Adult , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Camptothecin/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology
13.
Sci Rep ; 14(1): 22357, 2024 09 27.
Article in English | MEDLINE | ID: mdl-39333227

ABSTRACT

Although antibody-drug conjugate (ADC) or immune checkpoint inhibitors (ICIs) alone fosters hope for the treatment of cancer, the effect of single drug treatment is limited and the safety profile of ADC and ICI therapy remains unclear. This meta-analysis aimed to examine the efficacy and safety of the combination of ADC and ICI therapy. This study type is a systematic review and meta-analysis. Literature retrieval was carried out through PubMed, Embase, Cochrane from inception to Jun. 5, 2024. Then, after data extraction, overall response rate (ORR) and adverse effects (AEs) were used to study its efficiency and safety. Publication bias was also calculated through Funnel plot, Begg's Test and Egger's test. Heterogeneity was investigated through subgroup and sensitivity analysis. The research protocol was registered with the PROSPERO (CRD42023375601). A total of 12 eligible clinical studies with 584 patients were included. The pooled ORR was 58% (95%CI 46%, 70%). Subgroup analysis showed an ORR of 77% (95%CI 63%, 91%) in classical Hodgkin lymphoma (cHL) and an ORR of 73% (95%CI 56%, 90%) in non-Hodgkin lymphoma (NHL). The most common AEs was peripheral neuropathy (38.0%). Meanwhile, AEs on skin (13.1-20.0%) and digestive system (9.0-36.0%) was hard be overlooked. ADC + ICI therapy may be recommended in cancer treatment, especially in cHL and NHL. However, strategies to manage toxicities warranted further exploration.


Subject(s)
Immune Checkpoint Inhibitors , Immunoconjugates , Neoplasms , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/adverse effects , Immunoconjugates/therapeutic use , Immunoconjugates/adverse effects , Neoplasms/drug therapy , Treatment Outcome , Hodgkin Disease/drug therapy
15.
Clin Cancer Res ; 30(15): 3179-3188, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39086310

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized , Antigens, Neoplasm , Camptothecin , Cell Adhesion Molecules , Immunoconjugates , Humans , Cell Adhesion Molecules/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Female , Male , Aged , Middle Aged , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Immunoconjugates/therapeutic use , Aged, 80 and over , Adult , Biomarkers, Tumor/metabolism , Urologic Neoplasms/drug therapy , Urologic Neoplasms/pathology , Urologic Neoplasms/mortality , Urologic Neoplasms/metabolism , Treatment Outcome , Neoplasm Staging
16.
PLoS One ; 19(8): e0308247, 2024.
Article in English | MEDLINE | ID: mdl-39088571

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Bendamustine Hydrochloride , Lymphoma, Large B-Cell, Diffuse , Rituximab , Systematic Reviews as Topic , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Rituximab/administration & dosage , Rituximab/therapeutic use , Bendamustine Hydrochloride/administration & dosage , Bendamustine Hydrochloride/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Immunoconjugates/therapeutic use , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Randomized Controlled Trials as Topic , Antibodies, Monoclonal
17.
Nat Commun ; 15(1): 6707, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112464

ABSTRACT

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 .


Subject(s)
Antibodies, Monoclonal, Humanized , Brain Neoplasms , Breast Neoplasms , Glioblastoma , Immunoconjugates , Neoplasm Recurrence, Local , Humans , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Brain Neoplasms/secondary , Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Middle Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Adult , Aged , Immunoconjugates/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Prospective Studies , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism
18.
J Exp Clin Cancer Res ; 43(1): 236, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39164784

ABSTRACT

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.


Subject(s)
Breast Neoplasms , DNA Repair , Drug Resistance, Neoplasm , Immunoconjugates , Receptor, ErbB-2 , Trastuzumab , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Animals , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice , Receptor, ErbB-2/metabolism , Cell Line, Tumor , Xenograft Model Antitumor Assays , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Camptothecin/therapeutic use , Drug Synergism
19.
Pharmacol Res ; 207: 107341, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39134188

ABSTRACT

Drug conjugates are obtained from tumor-located vectors connected to cytotoxic agents via linkers, which are designed to deliver hyper-toxic payloads directly to targeted cancer cells. These drug conjugates include antibody-drug conjugates (ADCs), peptide-drug conjugates (PDCs), small molecule-drug conjugates (SMDCs), nucleic acid aptamer-drug conjugates (ApDCs), and virus-like drug conjugate (VDCs), which show great therapeutic value in the clinic. Drug conjugates consist of a targeting carrier, a linker, and a payload. Payloads are key therapy components. Cytotoxic molecules and their derivatives derived from natural products are commonly used in the payload portion of conjugates. The ideal payload should have sufficient toxicity, stability, coupling sites, and the ability to be released under specific conditions to kill tumor cells. Microtubule protein inhibitors, DNA damage agents, and RNA inhibitors are common cytotoxic molecules. Among these conjugates, cytotoxic molecules of natural origin are summarized based on their mechanism of action, conformational relationships, and the discovery of new derivatives. This paper also mentions some cytotoxic molecules that have the potential to be payloads. It also summarizes the latest technologies and novel conjugates developed in recent years to overcome the shortcomings of ADCs, PDCs, SMDCs, ApDCs, and VDCs. In addition, this paper summarizes the clinical trials conducted on conjugates of these cytotoxic molecules over the last five years. It provides a reference for designing and developing safer and more efficient conjugates.


Subject(s)
Antineoplastic Agents , Biological Products , Immunoconjugates , Neoplasms , Humans , Neoplasms/drug therapy , Animals , Biological Products/therapeutic use , Biological Products/chemistry , Biological Products/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Immunoconjugates/therapeutic use , Immunoconjugates/chemistry , Immunoconjugates/pharmacology
20.
Gan To Kagaku Ryoho ; 51(7): 691-694, 2024 Jul.
Article in Japanese | MEDLINE | ID: mdl-39191682

ABSTRACT

Most recently, the antibody-drug conjugate, enfortumab vedotin, has been covered by insurance for the treatment of unresectable or metastatic urothelial carcinoma after chemotherapy and immune checkpoint inhibitors, and has significantly changed the pharmacotherapy for urothelial carcinoma in clinical practice. Recently, several reports demonstarated the efficacy and safety of enfortumab vedotin in real-world clinical practice, highlighting its current status and challenges. In addition, the combination of enfortumab vedotin and pembrolizumab for the treatment of untreated unresectable or metastatic urothelial carcinoma has received expedited approval by the FDA and is expected to become a key drug in urothelial carcinoma.


Subject(s)
Immunoconjugates , Humans , Immunoconjugates/therapeutic use , Urologic Neoplasms/drug therapy , Urologic Neoplasms/immunology , Urinary Bladder Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL