Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.328
Filter
1.
Nat Commun ; 15(1): 7077, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152118

ABSTRACT

Enoblituzumab, an immunotherapeutic agent targeting CD276, shows both safety and efficacy in activating T cells and oligodendrocyte-like cells against various cancers. Preclinical studies and mouse models suggest that therapies targeting CD276 may outperform PD1/PD-L1 blockade. However, data from mouse models indicate a significant non-responsive population to anti-CD276 treatment, with the mechanisms of resistance still unclear. In this study, we evaluate the activity of anti-CD276 antibodies in a chemically-induced murine model of head and neck squamous cell carcinoma. Using models of induced and orthotopic carcinogenesis, we identify ITGB6 as a key gene mediating differential responses to anti-CD276 treatment. Through single-cell RNA sequencing and gene-knockout mouse models, we find that ITGB6 regulates the expression of the tumor-associated chemokine CX3CL1, which recruits and activates PF4+ macrophages that express high levels of CX3CR1. Inhibition of the CX3CL1-CX3CR1 axis suppresses the infiltration and secretion of CXCL16 by PF4+ macrophages, thereby reinvigorating cytotoxic CXCR6+ CD8+ T cells and enhancing sensitivity to anti-CD276 treatment. Further investigations demonstrate that inhibiting ITGB6 restores sensitivity to PD1 antibodies in mice resistant to anti-PD1 treatment. In summary, our research reveals a resistance mechanism associated with immune checkpoint inhibitor therapy and identifies potential targets to overcome resistance in cancer treatment.


Subject(s)
B7 Antigens , Head and Neck Neoplasms , Mice, Knockout , Animals , Mice , B7 Antigens/metabolism , B7 Antigens/genetics , B7 Antigens/antagonists & inhibitors , Humans , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Macrophages/immunology , Macrophages/metabolism , Cell Line, Tumor , Mice, Inbred C57BL , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Disease Models, Animal , Female , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects
2.
Cancer Med ; 13(1): e6945, 2024 Jan.
Article in English | MEDLINE | ID: mdl-39102671

ABSTRACT

INTRODUCTION: Adaptive mutagenesis observed in colorectal cancer (CRC) cells upon exposure to EGFR inhibitors contributes to the development of resistance and recurrence. Multiple investigations have indicated a parallel between cancer cells and bacteria in terms of exhibiting adaptive mutagenesis. This phenomenon entails a transient and coordinated escalation of error-prone translesion synthesis polymerases (TLS polymerases), resulting in mutagenesis of a magnitude sufficient to drive the selection of resistant phenotypes. METHODS: In this study, we conducted a comprehensive pan-transcriptome analysis of the regulatory framework within CRC cells, with the objective of identifying potential transcriptome modules encompassing certain translesion polymerases and the associated transcription factors (TFs) that govern them. Our sampling strategy involved the collection of transcriptomic data from tumors treated with cetuximab, an EGFR inhibitor, untreated CRC tumors, and colorectal-derived cell lines, resulting in a diverse dataset. Subsequently, we identified co-regulated modules using weighted correlation network analysis with a minKMEtostay threshold set at 0.5 to minimize false-positive module identifications and mapped the modules to STRING annotations. Furthermore, we explored the putative TFs influencing these modules using KBoost, a kernel PCA regression model. RESULTS: Our analysis did not reveal a distinct transcriptional profile specific to cetuximab treatment. Moreover, we elucidated co-expression modules housing genes, for example, POLK, POLI, POLQ, REV1, POLN, and POLM. Specifically, POLK, POLI, and POLQ were assigned to the "blue" module, which also encompassed critical DNA damage response enzymes, for example. BRCA1, BRCA2, MSH6, and MSH2. To delineate the transcriptional control of this module, we investigated associated TFs, highlighting the roles of prominent cancer-associated TFs, such as CENPA, HNF1A, and E2F7. CONCLUSION: We found that translesion polymerases are co-regulated with DNA mismatch repair and cell cycle-associated factors. We did not, however, identified any networks specific to cetuximab treatment indicating that the response to EGFR inhibitors relates to a general stress response mechanism.


Subject(s)
Cetuximab , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Cetuximab/pharmacology , Cetuximab/therapeutic use , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Gene Regulatory Networks , Gene Expression Profiling , ErbB Receptors/metabolism , ErbB Receptors/genetics , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use
3.
Drug Resist Updat ; 76: 101118, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39094301

ABSTRACT

AIMS: Resistance to targeted therapy is one of the critical obstacles in cancer management. Resistance to trastuzumab frequently develops in the treatment for HER2+ cancers. The role of protein tyrosine phosphatases (PTPs) in trastuzumab resistance is not well understood. In this study, we aim to identify pivotal PTPs affecting trastuzumab resistance and devise a novel counteracting strategy. METHODS: Four public datasets were used to screen PTP candidates in relation to trastuzumab responsiveness in HER2+ breast cancer. Tyrosine kinase (TK) arrays were used to identify kinases that linked to protein tyrosine phosphate receptor type O (PTPRO)-enhanced trastuzumab sensitivity. The efficacy of small activating RNA (saRNA) in trastuzumab-conjugated silica nanoparticles was tested for PTPRO upregulation and resistance mitigation in cell models, a transgenic mouse model, and human cancer cell line-derived xenograft models. RESULTS: PTPRO was identified as the key PTP which influences trastuzumab responsiveness and patient survival. PTPRO de-phosphorated several TKs, including the previously overlooked substrate ERBB3, thereby inhibiting multiple oncogenic pathways associated with drug resistance. Notably, PTPRO, previously deemed "undruggable," was effectively upregulated by saRNA-loaded nanoparticles. The upregulated PTPRO simultaneously inhibited ERBB3, ERBB2, and downstream SRC signaling pathways, thereby counteracting trastuzumab resistance. CONCLUSIONS: Antibody-conjugated saRNA represents an innovative approach for targeting "undruggable" PTPs.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Nanoparticles , Receptor, ErbB-2 , Trastuzumab , Xenograft Model Antitumor Assays , Trastuzumab/pharmacology , Humans , Drug Resistance, Neoplasm/drug effects , Animals , Mice , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Cell Line, Tumor , Nanoparticles/chemistry , Mice, Transgenic , Antineoplastic Agents, Immunological/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Signal Transduction/drug effects
4.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125956

ABSTRACT

Cancer-specific monoclonal antibodies (CasMabs) that recognize cancer-specific antigens with in vivo antitumor efficacy are innovative therapeutic strategies for minimizing adverse effects. We previously established a cancer-specific anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody (mAb), H2Mab-250/H2CasMab-2. In flow cytometry and immunohistochemistry, H2Mab-250 reacted with HER2-positive breast cancer cells but did not show reactivity to normal epithelial cells. In contrast, a clinically approved anti-HER2 mAb, trastuzumab, strongly recognizes both breast cancer and normal epithelial cells in flow cytometry. The human IgG1 version of H2Mab-250 (H2Mab-250-hG1) possesses compatible in vivo antitumor effects against breast cancer xenografts to trastuzumab despite the lower affinity and effector activation than trastuzumab in vitro. This study compared the antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cellular cytotoxicity (CDC) between H2Mab-250-hG1 and trastuzumab. Both H2Mab-250-hG1 and trastuzumab showed ADCC activity against HER2-overexpressed Chinese hamster ovary -K1 and breast cancer cell lines (BT-474 and SK-BR-3) in the presence of human natural killer cells. Some tendency was observed where trastuzumab showed a more significant ADCC effect compared to H2Mab-250-hG1. Importantly, H2Mab-250-hG1 exhibited superior CDC activity in these cells compared to trastuzumab. Similar results were obtained in the mouse IgG2a types of both H2Mab-250 and trastuzumab. These results suggest the different contributions of ADCC and CDC activities to the antitumor effects of H2Mab-250-hG1 and trastuzumab, and indicate a future direction for the clinical development of H2Mab-250-hG1 against HER2-positive tumors.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Cricetulus , Receptor, ErbB-2 , Trastuzumab , Trastuzumab/pharmacology , Animals , Humans , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , CHO Cells , Cell Line, Tumor , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Antineoplastic Agents, Immunological/pharmacology , Antibodies, Monoclonal/pharmacology , Complement System Proteins/metabolism , Complement System Proteins/immunology , Mice , Cricetinae
5.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000238

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide. Therefore, the need for new therapeutic strategies is still a challenge. Surgery and chemotherapy represent the first-line interventions; nevertheless, the prognosis for metastatic CRC (mCRC) patients remains unacceptable. An important step towards targeted therapy came from the inhibition of the epidermal growth factor receptor (EGFR) pathway, by the anti-EGFR antibody, Cetuximab, or by specific tyrosine kinase inhibitors (TKI). Cetuximab, a mouse-human chimeric monoclonal antibody (mAb), binds to the extracellular domain of EGFR thus impairing EGFR-mediated signaling and reducing cell proliferation. TKI can affect the EGFR biochemical pathway at different steps along the signaling cascade. Apart from Cetuximab, other anti-EGFR mAbs have been developed, such as Panitumumab. Both antibodies have been approved for the treatment of KRAS-NRAS wild type mCRC, alone or in combination with chemotherapy. These antibodies display strong differences in activating the host immune system against CRC, due to their different immunoglobulin isotypes. Although anti-EGFR antibodies are efficient, drug resistance occurs with high frequency. Resistant tumor cell populations can either already be present before therapy or develop later by biochemical adaptations or new genomic mutations in the EGFR pathway. Numerous efforts have been made to improve the efficacy of the anti-EGFR mAbs or to find new agents that are able to block downstream EGFR signaling cascade molecules. Indeed, we examined the importance of analyzing the anti-EGFR antibody-drug conjugates (ADC) developed to overcome resistance and/or stimulate the tumor host's immunity against CRC growth. Also, patient-derived CRC organoid cultures represent a useful and feasible in vitro model to study tumor behavior and therapy response. Organoids can reflect tumor genetic heterogeneity found in the tissue of origin, representing a unique tool for personalized medicine. Thus, CRC-derived organoid cultures are a smart model for studying the tumor microenvironment and for the preclinical assay of anti-EGFR drugs.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm , ErbB Receptors , Organoids , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Organoids/metabolism , Organoids/drug effects , Drug Resistance, Neoplasm/drug effects , Animals , Cetuximab/pharmacology , Cetuximab/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Molecular Targeted Therapy/methods , Panitumumab/pharmacology , Panitumumab/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Signal Transduction/drug effects
6.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000504

ABSTRACT

HER2-targeted therapies, such as Trastuzumab (Tz), have significantly improved the clinical outcomes for patients with HER2+ breast cancer (BC). However, treatment resistance remains a major obstacle. To elucidate functional and metabolic changes associated with acquired resistance, we characterized protein profiles of BC Tz-responder spheroids (RSs) and non-responder spheroids (nRSs) by a proteomic approach. Three-dimensional cultures were generated from the HER2+ human mammary adenocarcinoma cell line BT-474 and a derived resistant cell line. Before and after a 15-day Tz treatment, samples of each condition were collected and analyzed by liquid chromatography-mass spectrometry. The analysis of differentially expressed proteins exhibited the deregulation of energetic metabolism and mitochondrial pathways. A down-regulation of carbohydrate metabolism and up-regulation of mitochondria organization proteins, the tricarboxylic acid cycle, and oxidative phosphorylation, were observed in nRSs. Of note, Complex I-related proteins were increased in this condition and the inhibition by metformin highlighted that their activity is necessary for nRS survival. Furthermore, a correlation analysis showed that overexpression of Complex I proteins NDUFA10 and NDUFS2 was associated with high clinical risk and worse survival for HER2+ BC patients. In conclusion, the non-responder phenotype identified here provides a signature of proteins and related pathways that could lead to therapeutic biomarker investigation.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Electron Transport Complex I , Proteomics , Receptor, ErbB-2 , Trastuzumab , Humans , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Electron Transport Complex I/metabolism , Proteomics/methods , Receptor, ErbB-2/metabolism , Cell Line, Tumor , Mitochondria/metabolism , Mitochondria/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/drug effects , Proteome/metabolism , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use
7.
Crit Rev Oncog ; 29(4): 43-54, 2024.
Article in English | MEDLINE | ID: mdl-38989737

ABSTRACT

Breast cancer (BC) is the most common cancer and the second leading cause of cancer-related deaths in women globally. Despite advancements in treatment strategies, many patients still develop challenging-to-treat metastatic disease. The development and progression of tumors are influenced by genetic/epigenetic changes within tumor cells and alterations in the tumor microenvironment (TME) through a dynamic communication. The TME comprises various elements, including immune, tumor, and stromal cells. Tumor cells at the core of the TME orchestrate complex signals that lead to tumor growth, survival, and resistance to treatment. Human epidermal growth factor receptor 2 (HER2) is overexpressed in a significant proportion of invasive breast cancers, influencing prognosis and prediction. Novel therapeutic approaches target HER2-positive breast cancers by leveraging HER2-targeted therapeuirtcs such as antibody-drug conjugates, monoclonal antibodies, and tyrosine kinase inhibitors. The TME in HER2-positive breast cancers also involves cancer-associated fibroblasts and cancer-associated adipocytes, which play critical roles in tumor progression and therapy resistance. The immune microenvironment also plays a significant role, with studies indicating its impact on outcomes in HER2-positive breast cancer. Trastuzumab, one of the first monoclonal antibodies targeting HER2, has shown promise in enhancing survival rates in HER2-overexpressing breast cancer. Integration of trastuzumab with chemotherapy has demonstrated significant enhancements in disease-free survival as well as overall survival rates during early breast cancer treatment. Trastuzumab functions by inhibiting HER2 signaling pathways, leading to cell cycle arrest and induction of apoptosis. Overall, understanding the complex interplay between HER2, the tumor microenvironment, and therapeutic interventions is essential for improving outcomes in HER2-positive BC.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Receptor, ErbB-2 , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Receptor, ErbB-2/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Female , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Animals
8.
Front Immunol ; 15: 1398508, 2024.
Article in English | MEDLINE | ID: mdl-38983860

ABSTRACT

Background: CD38 and CD47 are expressed in many hematologic malignancies, including multiple myeloma (MM), B-cell non-Hodgkin lymphoma (NHL), B-cell acute lymphoblastic leukemia (ALL), and B-cell chronic lymphocytic leukemia (CLL). Here, we evaluated the antitumor activities of CD38/CD47 bispecific antibodies (BsAbs). Methods: Five suitable anti-CD38 antibodies for co-targeting CD47 and CD38 BsAb were developed using a 2 + 2 "mAb-trap" platform. The activity characteristics of the CD38/CD47 BsAbs were evaluated using in vitro and in vivo systems. Results: Using hybridoma screening technology, we obtained nine suitable anti-CD38 antibodies. All anti-CD38 antibodies bind to CD38+ tumor cells and kill tumor cells via antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Five anti-CD38 antibodies (4A8, 12C10, 26B4, 35G5, and 65A7) were selected for designing CD38/CD47 BsAbs (IMM5605) using a "mAb-trap" platform. BsAbs had higher affinity and binding activity to the CD38 target than those to the CD47 target, decreasing the potential on-target potential and off-tumor effects. The CD38/CD47 BsAbs did not bind to RBCs and did not induce RBC agglutination; thus, BsAbs had much lower blood toxicity. The CD38/CD47 BsAbs had a greater ability to block the CD47/SIRPα signal in CD38+/CD47+ tumor cells than IMM01 (SIRPα Fc fusion protein). Through Fc domain engineering, CD38/CD47 BsAbs were shown to kill tumors more effectively by inducing ADCC and ADCP. IMM5605-26B4 had the strongest inhibitory effect on cellular CD38 enzymatic activity. IMM5605-12C10 had the strongest ability to directly induce the apoptosis of tumor cells. The anti-CD38 antibody 26B4 combined with the SIRPα-Fc fusion proteins showed strong antitumor effects, which were better than any of the mono-therapeutic agents used alone in the NCI-H929 cell xenograft model. The CD38/CD47 BsAbs exhibited strong antitumor effects; specifically, IMM5605-12C10 efficiently eradicated all established tumors in all mice. Conclusion: A panel of BsAbs targeting CD38 and CD47 developed based on the "mAb-tarp" platform showed potent tumor-killing ability in vitro and in vivo. As BsAbs had lower affinity for binding to CD47, higher affinity for binding to CD38, no affinity for binding to RBCs, and did not induce RBC agglutination, we concluded that CD38/CD47 BsAbs are safe and have a satisfactory tolerability profile.


Subject(s)
ADP-ribosyl Cyclase 1 , CD47 Antigen , Hematologic Neoplasms , CD47 Antigen/immunology , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/metabolism , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/immunology , ADP-ribosyl Cyclase 1/metabolism , Humans , Animals , Mice , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology , Cell Line, Tumor , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , Xenograft Model Antitumor Assays , Membrane Glycoproteins/immunology , Membrane Glycoproteins/antagonists & inhibitors , Antibody-Dependent Cell Cytotoxicity , Female , Antineoplastic Agents, Immunological/pharmacology
9.
ESMO Open ; 9(7): 103622, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39002179

ABSTRACT

BACKGROUND: PLAnning Treatment For Oesophago-gastric Cancer: a Randomised Maintenance Therapy Trial (PLATFORM) is an adaptive phase II study assessing the role of maintenance therapies in advanced oesophago-gastric (OG) adenocarcinoma. We evaluated the role of the anti-programmed death-ligand 1 (PD-L1) inhibitor durvalumab in these patients. PATIENTS AND METHODS: Patients with human epidermal growth factor receptor 2-negative locally advanced or metastatic OG adenocarcinoma with disease control or response to 18 weeks of platinum-based first-line chemotherapy were randomised to active surveillance or maintenance durvalumab. The primary endpoint was progression-free survival (PFS). Safety was assessed in all patients who had commenced surveillance visits or received at least one dose of durvalumab. Exploratory survival analyses according to PD-L1 Combined Positive Score (CPS) and immune (biomarker-positive) or angiogenesis dominant (biomarker-negative) tumour microenvironment (TME) phenotypes were conducted. RESULTS: Between March 2015 and April 2020, 205 patients were randomised to surveillance (n = 100) and durvalumab (n = 105). No significant differences were seen in PFS [hazard ratio (HR) 0.84, P = 0.13] and overall survival (OS; HR 0.98, P = 0.45) between surveillance and durvalumab. Five patients randomised to durvalumab demonstrated incremental radiological responses compared with none with surveillance. Treatment-related adverse events occurred in 77 (76.2%) durvalumab-assigned patients. A favourable effect in OS with durvalumab over surveillance in CPS ≥5 and immune biomarker-positive patients was observed compared with CPS <5 and biomarker-negative subgroups, respectively: CPS ≥5 versus <5: HR 0.63, 95% confidence interval (CI) 0.32-1.22 versus HR 0.93, 95% CI 0.44-1.96; biomarker-positive versus negative: HR 0.60, 95% CI 0.29-1.23 versus HR 0.84, 95% CI 0.42-1.65. CONCLUSION: Maintenance durvalumab does not improve PFS in patients with OG adenocarcinoma who respond to first-line chemotherapy but induced incremental radiological responses in a subset of patients. TME characterisation could refine patient selection for anti-PD-L1 therapy above PD-L1 CPS alone.


Subject(s)
Adenocarcinoma , Antibodies, Monoclonal , Esophageal Neoplasms , Stomach Neoplasms , Humans , Male , Female , Middle Aged , Stomach Neoplasms/drug therapy , Aged , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology , Adenocarcinoma/drug therapy , Esophageal Neoplasms/drug therapy , Adult , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Receptor, ErbB-2/metabolism , Aged, 80 and over , Progression-Free Survival
10.
J Cancer Res Ther ; 20(3): 984-992, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-39023607

ABSTRACT

AIM: Although people with HER2-positive breast cancer benefit from approved HER2-targeted therapy, acquiring resistance to the therapies occurs. Animal models can play a part in gaining a deep understanding of such a process and addressing questions concerning developing and improving immunotherapy approaches. MATERIALS AND METHODS: To develop such a model, we transfected murine 4T1 cells with the pCMV6-Neo-HER2 construct and evaluated HER2 expression and its effects on the established cell line behavior in vitro and in vivo. RESULTS: Data illustrated that human HER2 protein was expressed on isolated 4T1-HER2 clones in vitro and in vivo. Except for proliferation over 48 hours, such expression did not change 4T1-HER2 characteristics compared to 4T1 in vitro. Notwithstanding the reduction in proliferation, the rate of tumorigenicity was 90% in challenged mice and Herceptin therapy significantly decreased tumors' growth and metastasis compared to the control group. CONCLUSION: We describe a murine model for HER2-positive breast cancer not only helping shed light on the mechanisms by which the tumor evades antitumor immunity but also playing a key role in making breast cancer more sensitive to novel immunotherapy modalities.


Subject(s)
Breast Neoplasms , Disease Models, Animal , Receptor, ErbB-2 , Animals , Female , Humans , Mice , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Xenograft Model Antitumor Assays
11.
Med Sci (Paris) ; 40(6-7): 569-572, 2024.
Article in French | MEDLINE | ID: mdl-38986105

ABSTRACT

Title: L'immunocytokine FAP-IL2v: Un co-traitement efficace pour pallier la résistance au trastuzumab du cancer du sein HER2. Abstract: Dans le cadre de leur module d'analyse scientifique, des étudiants des promotions 2022-2023 et 2023-2024 des Master 2 « Immunologie Translationnelle et Biothérapies ¼ (ITB) et « Immunologie Intégrative et Systémique ¼ (I2S) (Mention Biologie Moléculaire et Cellulaire, Parcours Immunologie, Sorbonne Université) se sont penchés sur la littérature et ont pris la plume pour partager avec les lecteurs de m/s quelques-uns des faits marquants de l'actualité en immunologie. Voici une sélection de quelques-unes de ces nouvelles, illustrant la large palette des axes de recherche en cours sur les mécanismes physiopathologiques des maladies infectieuses, auto-immunes, inflammatoires et tumorales et sur le développement d'immunothérapies pour le traitement de ces pathologies. On y découvre ainsi de nouvelles avancées sur l'analyse transcriptomique du microenvironnement inflammatoire de pathologies autoimmunes, sur des aspects mécanistiques impliqués dans la survie des cellules cancéreuses et la réponse immunitaire anti-tumorale des cellules NK, l'interconnexion entre le système immunitaire et le système nerveux périphérique, le développement de nouvelles immunothérapies permettant de cibler préférentiellement le microenvironnement tumoral et la prise en charge des effets secondaires autoimmuns cardiaques induits par les immunothérapies. Toute l'équipe pédagogique remercie également chaleureusement les différents tuteurs, experts dans le domaine en lien avec les nouvelles, qui ont accompagné avec bienveillance et enthousiasme le travail de nos étudiants !


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Receptor, ErbB-2 , Trastuzumab , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Receptor, ErbB-2/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Trastuzumab/therapeutic use , Immunotherapy/methods , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Recombinant Fusion Proteins/therapeutic use , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
12.
Dokl Biochem Biophys ; 517(1): 285-290, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002014

ABSTRACT

The direct antitumor effect of bevacizumab (BEV) has long been debated. Evidence of the direct antitumor activities of drugs are mainly obtained from in vitro experiments, which are greatly affected by experimental conditions. In this study, we evaluated the effect of BEV-containing medium renewal on the results of in vitro cytotoxicity experiments in A549 and U251 cancer cells. We observed starkly different results between the experiments with and without BEV-containing medium renewal. Specifically, BEV inhibited the tumor cell growth in the timely replacement with a BEV-containing medium but promoted tumor cell growth without medium renewal. Meanwhile, compared with the control, a significant basic fibroblast growth factor (bFGF) accumulation in the supernatant was observed in the group without medium renewal but none in that with replaced medium. Furthermore, bFGF neutralization partially reversed the pro-proliferative effect of BEV in the medium non-renewed group, while exogenous bFGF attenuated the tumor cell growth inhibition of BEV in the medium-renewed group. Our data explain the controversy over the direct antitumor effect of BEV in different studies from the perspective of the compensatory autocrine cytokines in tumor cells.


Subject(s)
Bevacizumab , Cell Proliferation , Fibroblast Growth Factor 2 , Humans , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/metabolism , Bevacizumab/pharmacology , Cell Proliferation/drug effects , Cell Line, Tumor , Culture Media/chemistry , Culture Media/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , A549 Cells , Antineoplastic Agents, Immunological/pharmacology
13.
Oral Oncol ; 157: 106971, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39067262

ABSTRACT

BACKGROUND: Merkel cell carcinoma (MCC) is an uncommon skin cancer that in more than 90 % of cases develops within the head and neck region. MCC is an aggressive disease with a dismal prognosis before the advent of immunotherapy. Avelumab, an anti-PDL1 agent is approved since 2017 for the treatment of advanced MCC after demonstrating a high response rate and favorable impact in survival. METHODS: Next generation sequencing (NGS) of the primary tumor biopsy from initial diagnosis (Foundation One CDx, Roche Diagnostics) as well as from plasma samples (Foundation One Liquid, Roche Diagnostics) obtained before and during treatment with avelumab were performed. RESULTS: We present the case of a patient with a metastatic MCC developing in the left parotid gland / pre-auricular area in an 80-year-old male with a long-lasting history of high UV exposure. After two cycles of avelumab 10 mg/kg/q2wk a near complete metabolic response and a major radiological response occurred in parallel to a brisk reduction in the ctDNA tumor fraction as well as variant-allele frequencies (VAFs) of all the mutations detected before the start of avelumab. CONCLUSION: Avelumab may achieve rapid and major responses in metastatic MCC. Our study demonstrates that ctDNA mirrors radiological responses and may serve as an ideal companion for diagnosis and disease monitoring in MCC.


Subject(s)
Antibodies, Monoclonal, Humanized , Carcinoma, Merkel Cell , Circulating Tumor DNA , Head and Neck Neoplasms , Skin Neoplasms , Humans , Carcinoma, Merkel Cell/drug therapy , Male , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Aged, 80 and over , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/genetics , Circulating Tumor DNA/genetics , Circulating Tumor DNA/blood , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology
14.
Cancer Biol Ther ; 25(1): 2373497, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38967961

ABSTRACT

Despite advances in targeted therapies, primary and acquired resistance make the treatment of colorectal cancer (CRC) a pressing issue to be resolved. According to reports, the development of CRC is linked to miRNA dysregulation. Multiple studies have demonstrated that miR-135b-5p has an aberrant expression level between CRC tissues and adjacent tissues. However, it is unclear whether there is a correlation between miR-135b-5p and cetuximab (CTx) resistance in CRC. Use the GEO database to measure miR-135b-5p expression in CRC. Additionally, RT-qPCR was applied to ascertain the production level of miR-135b-5p in three human CRC cells and NCM460 cells. The capacity of cells to migrate and invade was examined utilizing the wound-healing and transwell assays, while the CCK-8 assay served for evaluating cell viability, as well as colony formation assays for proliferation. The expected target protein of miR-135b-5p in CRC cell cetuximab resistance has been investigated using western blot. Suppression of miR-135b-5p could increase the CTx sensitivity of CTx-resistant CRC cells, as manifested by the attenuation of proliferation, migration, and invasion ability. Mechanistic studies revealed miR-135b-5p regulates the epithelial-to-mesenchymal transition (EMT) process and Wnt/ß-catenin signaling pathway through downgulating FOXN3. In short, knockdowning miR-135b-5p could increase FOXN3 expression in CRC cells, promote the EMT process, and simultaneously activate the Wnt/ß-catenin signaling pathway to elevate CTx resistance in CRC cells.


Subject(s)
Cetuximab , Colorectal Neoplasms , Drug Resistance, Neoplasm , Forkhead Transcription Factors , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Cetuximab/pharmacology , Cetuximab/therapeutic use , Drug Resistance, Neoplasm/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Cell Proliferation/drug effects , Cell Movement , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Wnt Signaling Pathway/drug effects , Cell Cycle Proteins
15.
Cancer Lett ; 598: 217103, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38969162

ABSTRACT

Cetuximab in combination with FOLFIRI/FOLFOX is the standard first-line treatment for patients with RAS wild-type metastatic colorectal cancer (mCRC). However, some patients experience rapid tumor progression after treatment with cetuximab (primary resistance). Our previous research identified a gene mutation, REV1 p.R704Q, which may be a key biomarker for primary cetuximab resistance. This study aimed to study the mechanism of cetuximab resistance caused by REV1 p.R704Q mutation and reveal a novel mechanism to induce cetuximab resistance. Sanger sequencing and multivariate clinical prognostic analysis of 208 patients with mCRC showed that REV1 p.R704Q mutation is an independent risk factor for tumor progression after treatment with cetuximab in patients with RAS wild-type mCRC (Hazard ratio = 2.481, 95 % Confidence interval: 1.389-4.431, P = 0.002). The sensitivity of REV1 p.R704Q mutant cell lines to cetuximab decreased in vitro Cell Counting Kit-8 assay and in vivo subcutaneous tumor model. In vitro, we observed that decreased stability and accelerated degradation of REV1 mutant protein results in REV1 dysfunction, which activated autophagy and mediated cetuximab resistance. These findings suggested that REV1 p.R704Q mutation could predict cetuximab primary resistance in mCRC. REV1 p.R704Q mutation caused decreased stability and degradation of REV1 protein, as well as dysfunction of p.R704Q protein. REV1 p.R704Q mutation activates autophagy and mediates cetuximab resistance; further, inhibition of autophagy could reverse cetuximab resistance.


Subject(s)
Autophagy , Cetuximab , Colorectal Neoplasms , Drug Resistance, Neoplasm , Mutation , Humans , Cetuximab/pharmacology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Autophagy/drug effects , Autophagy/genetics , Male , Female , Animals , Mice , Middle Aged , Cell Line, Tumor , Xenograft Model Antitumor Assays , Aged , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Mice, Nude , Prognosis
16.
Trends Cancer ; 10(8): 708-732, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38987076

ABSTRACT

Over the past 30 years the incorporation of monoclonal antibody (mAb) treatments into the management of hematologic malignancies has led to significant improvements in patient outcomes. The key limitation of mAb treatments is the necessity for target antigen presentation on major histocompatibility complex (MHC) and costimulatory molecules to elicit a cytotoxic immune response. With the advent of bispecific antibodies (BsAbs), these limitations can be overcome through direct stimulation of cytotoxic T cells, thus limiting tumor cell evasion. BsAbs are rapidly being incorporated into treatment regimens for hematologic malignancies, and there are now seven FDA-approved treatments in this class, six of which have been approved in the past year. In this review we describe the function, complications, and clinical trial data available for CD3 BsAbs in the treatment of lymphoma, myeloma, and leukemia.


Subject(s)
Antibodies, Bispecific , CD3 Complex , Hematologic Neoplasms , Humans , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , CD3 Complex/immunology , CD3 Complex/antagonists & inhibitors , Hematologic Neoplasms/immunology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/therapy , Clinical Trials as Topic , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Multiple Myeloma/immunology , Multiple Myeloma/drug therapy , Multiple Myeloma/therapy , Lymphoma/immunology , Lymphoma/drug therapy , Lymphoma/therapy , Animals , Leukemia/immunology , Leukemia/drug therapy , Leukemia/therapy
17.
Int Immunopharmacol ; 138: 112612, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38968862

ABSTRACT

Cholangiocarcinoma (CCA) is an aggressive and fatal cancer. The prognosis is very poor and no optimal chemotherapy has been established. Human epidermal growth factor receptor 2 (HER2, neu, and erbB2) is highly-expressed in breast cancer and is expressed in many other tumors but poorly expressed in CCA. The anti-HER2 antibody, trastuzumab, has been used for the treatment of HER2-positive breast and gastric cancer. In this study, we examined the surface expression of HER2 on seven Thai liver-fluke-associated CCA cell lines by flow cytometry, and found all of these CCA cells were weakly positive for HER2. MTT assay revealed that trastuzumab directly suppressed the growth of CCA. By using FcR-bearing recombinant Jurkat T-cell-expressing firefly luciferase gene under the control of NFAT response elements, we defined the activities of antibody-dependent cytotoxicity (ADCC) and antibody-dependent cell phagocytosis (ADCP). ADCC was confirmed by using expanded NK cells. ADCP was confirmed by using mouse peritoneal macrophages and human monocyte-derived macrophages as effector cells. Rabbit serum was administered to test the complement-dependent cytotoxicity (CDC) activity of trastuzumab. Finally, we evaluated the efficacy of trastuzumab in in vivo patient-derived cell xenograft and patient-derived xenograft (PDX) models. Our results showed that a distinct population of CCA (liver-fluke-associated CCA) expressed HER2. Trastuzumab demonstrated a potent inhibitory effect on even HER2 weakly positive CCA both in vitro and in vivo via multiple mechanisms. Thus, HER2 is a promising target in anti-CCA therapy, and trastuzumab can be considered a promising antibody immunotherapy agent for the treatment of CCA.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents, Immunological , Bile Duct Neoplasms , Cholangiocarcinoma , Trastuzumab , Animals , Female , Humans , Male , Mice , Rabbits , Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Agents, Immunological/pharmacology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/immunology , Cell Line, Tumor , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/immunology , Jurkat Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Phagocytosis/drug effects , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Xenograft Model Antitumor Assays
18.
Int Immunopharmacol ; 138: 112609, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38971103

ABSTRACT

T-cell-engaging bispecific antibody (TCB) therapies have emerged as a promising immunotherapeutic approach, effectively redirecting effector T cells to selectively eliminate tumor cells. The therapeutic potential of TCBs has been well recognized, particularly with the approval of multiple TCBs in recent years for the treatment of hematologic malignancies as well as some solid tumors. However, TCBs encounter multiple challenges in treating solid tumors, such as on-target off-tumor toxicity, cytokine release syndrome (CRS), and T cell dysfunction within the immunosuppressive tumor microenvironment, all of which may impact their therapeutic efficacy. In this review, we summarize clinical data on TCBs for solid tumor treatment, highlight the challenges faced, and discuss potential solutions based on emerging strategies from current clinical and preclinical research. These solutions include TCB structural optimization, target selection, and combination strategies. This comprehensive analysis aims to guide the development of TCBs from design to clinical application, addressing the evolving landscape of cancer immunotherapy.


Subject(s)
Antibodies, Bispecific , Immunotherapy , Neoplasms , T-Lymphocytes , Tumor Microenvironment , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/pharmacology , Humans , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Immunotherapy/methods , Tumor Microenvironment/immunology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology
19.
Front Immunol ; 15: 1400177, 2024.
Article in English | MEDLINE | ID: mdl-38953027

ABSTRACT

Background: Chimeric antigen receptor T (CAR-T) cell therapies have achieved remarkable success in the treatment of hematological tumors. However, given the distinct features of solid tumors, particularly heterogeneity, metabolic aggressiveness, and fewer immune cells in tumor microenvironment (TME), the practical utility of CAR-T cells for solid tumors remains as a challenging issue. Meanwhile, although anti-PD-1 monoclonal antibody (mAb) has shown clinical efficacy, most mAbs also show limited clinical benefits for solid tumors due mainly to the issues associated with the lack of immune cells in TME. Thus, the infiltration of targeted immunological active cells into TME could generate synergistic efficacy for mAbs. Methods: We present a combinational strategy for solid tumor treatment, which combines armored-T cells to express Fc-gamma receptor I (FcγRI) fragment on the surfaces for targeting various tumors with therapeutically useful mAbs. Choosing CD20 and HER-2 as the targets, we characterized the in vitro and in vivo efficacy and latent mechanism of the combination drug by using flow cytometry, ELISA and other methods. Results: The combination and preprocessing of armored T-cells with corresponding antibody of Rituximab and Pertuzumab exerted profound anti-tumor effects, which is demonstrated to be mediated by synergistically produced antibody-dependent cellular cytotoxicity (ADCC) effects. Meanwhile, mAb was able to carry armored-T cell by preprocessing for the infiltration to TME in cell derived xenograft (CDX) model. Conclusions: This combination strategy showed a significant increase of safety profiles from the reduction of antibody doses. More importantly, the present strategy could be a versatile tool for a broad spectrum of cancer treatment, with a simple pairing of engineered T cells and a conventional antibody.


Subject(s)
Neoplasms , Receptors, IgG , T-Lymphocytes , Tumor Microenvironment , Receptors, IgG/immunology , Receptors, IgG/metabolism , Humans , Animals , Mice , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Xenograft Model Antitumor Assays , Immunotherapy, Adoptive/methods , Receptor, ErbB-2/immunology , Receptor, ErbB-2/antagonists & inhibitors , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Female , Antigens, CD20/immunology
20.
Expert Opin Investig Drugs ; 33(8): 851-865, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38967422

ABSTRACT

INTRODUCTION: Antibody-drug conjugates (ADCs) represent a revolutionary approach in the systemic treatment for both solid and hematologic tumors. Constituted by an antibody, a cytotoxic payload, and a linker, ADCs aim to selectively deliver cytotoxic agents to tumors while sparing normal tissues. Various ADCs have been tested and approved for multiple solid tumors so far, but if there is one that had a major impact on clinical practice, this is Trastuzumab-deruxtecan (T-DXd). Notably, T-DXd was approved for HER2-positive and HER2-low metastatic breast cancer (MBC), HER2-positive gastric cancer (GC), HER2-mutant non-small cell lung cancer (NSCLC) and HER2 3+ solid tumors. Moreover, it received Breakthrough Therapy Designation for HER2-positive colorectal cancer (CRC). AREAS COVERED: We review preclinical and clinical data of T-DXd, focusing on early-phase ongoing trials exploring combination therapies to enhance the activity of T-DXd in HER2-expressing solid tumors. EXPERT OPINION: The clinical use of T-DXd still raises questions about selection of patients, treatment duration, prioritization over other approved ADCs, and management of resistance. Concerns regarding the toxicity of T-DXd remain, particularly with combinations involving potentially toxic drugs. Advancements in biomarker identification and combination therapies offer promising avenues to enhance efficacy and overcome resistance to T-DXd, ultimately improving outcomes for patients with cancer.


Subject(s)
Camptothecin , Drug Development , Immunoconjugates , Receptor, ErbB-2 , Trastuzumab , Humans , Immunoconjugates/pharmacology , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Trastuzumab/administration & dosage , Animals , Camptothecin/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/administration & dosage , Camptothecin/adverse effects , Neoplasms/drug therapy , Neoplasms/pathology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL