1
|
Zhang M, Yang Q, Lou J, Hu Y, Shi Y. A new strategy to HER2-specific antibody discovery through artificial intelligence-powered phage display screening based on the Trastuzumab framework. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167772. [PMID: 40056877 DOI: 10.1016/j.bbadis.2025.167772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a recognized drug target, and it serves as a critical target for various cancer treatments, necessitating the discovery of more antibodies for therapeutic and detection purposes. Here, we have developed an innovative workflow for antibody generation through Artificial Intelligence-powered Phage Display Screening (AIPDS). This workflow integrates artificial intelligence-driven antibody CDRH3 sequence design, high-throughput DNA synthesis and phage display screening. We applied AIPDS workflow to generate promising antibodies against the human epidermal growth factor receptor 2 (HER2), offering a template for streamlined antibody generation. Seven novel antibodies stood out, demonstrating promising efficacy in various functional assays. Notably, DYHER2-02 demonstrates strong performance across all experimental tests. In summary, our study introduces a novel methodology to generate new antibody variants of an existing antibody using an AI-assisted phage display approach. These new antibody variants hold potential applications in research, diagnosis, and therapeutic applications.
Collapse
Affiliation(s)
- Mancang Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Qiangzhen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Jiangrong Lou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Yang Hu
- United Research Center for Next Generation DNA Synthesis of SJTU-Dynegene, Shanghai 201108, People's Republic of China
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China; Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China.
| |
Collapse
|
2
|
Ghosh M, Lingaraju SM, C R K, Balaram G, Kodandapani R, E V, K V, N S, H D, Patil S, Thungappa SC, Bhattacharjee S, P S S, Dasgupta R, Naseer M, B J S, Rao V, Ramaswamy V, Naik R, Babu G, Ravichandran A, Bahadur U, Murugan K, B M, Reddy L, Basavalinga S A. Comprehensive genomic profiling reveals a unique genomic landscape in solid tumors in an Indian cancer cohort of 1000 patients: a single institutional experience. Sci Rep 2025; 15:12455. [PMID: 40216820 PMCID: PMC11992052 DOI: 10.1038/s41598-025-94762-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
The use of Comprehensive Genomic Profiling (CGP) in clinical practice to detect broad-spectrum therapeutic, prognostic, and predictive biomarkers, including tumor mutational burden (TMB), microsatellite instability (MSI), somatic BRCA (sBRCA) and other homologous recombination repair genes (HRRs) provides a more cost-efficient and tissue-preserving approach than serial single-biomarker analysis. A total of 1000 biopsy-proven cancer patients at the HCG cancer center were profiled in an IRB-approved prospective study. The findings were discussed in the multidisciplinary molecular tumor board (MTB), and recommendations were documented in electronic medical records (EMRs) for clinical management and follow-up. A total of 1747 genomic alterations were detected (mean 1.7 mutations/sample), with 80% of patients having genetic alterations with therapeutic and prognostic implications (Tier I-32%, Tier II-50%). CGP revealed a greater number of druggable genes (47%) than did small panels (14%). Tumor-agnostic markers for immunotherapy (IO) were observed in 16% of the current cohort, based on which IO was initiated. In 13.5% of the cohort, alterations in the HRR pathway including sBRCA (5.5%) were detected providing an option for treatment with platinum or PARP inhibitors. Other significant alterations included those in EGFR, KRAS/BRAF, PIK3CA, cKIT, PDGFRA, ARID1A, ARID2, and FGFR. RNA sequencing revealed 55 + RNA alterations, including those in TMPRSS-ERG, RPS6KB1-VMP1, EML4-ALK, NTRK, PDGFRA and EWSR. Clinical outcome data were available via EMR for 618 patients (62%), out of whom 419 patients had druggable mutations (67%; 95% CI 88.9-93.9%) and 39 patients had 1 or more mutations with prognostic implications. However, only 200 patients (44%; 95% CI 39.1-48.1%) were included in the MTB discussion. Based on genomics reports, the treatment regimen was changed for 137 and 61 patients with and without clinical inputs from the MTB, respectively. The overall change in therapy based on CGP in the clinical cohort was 43%, which was greater in patients enrolled for MTB than in patients who had not undergone MTB. At the interim analysis, with a median follow-up of 18 months (range 12-24 months) after the change in therapy as per genomics report, 97 patients (71%) were found to be alive thus establishing the importance of CGP and MTB in personalized genomics-driven treatment.
Collapse
Affiliation(s)
- Mithua Ghosh
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India.
| | - Sheela Mysore Lingaraju
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Krishna C R
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Gautam Balaram
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Ramya Kodandapani
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Vijay E
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Vijay K
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Suhas N
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Devika H
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Shekar Patil
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | | | | | - Sridhar P S
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Roshni Dasgupta
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Mohammed Naseer
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Srinivas B J
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Vishal Rao
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Veena Ramaswamy
- Triesta Sciences, A Unit of HeathCare Global Enterprises Limited, HCG Towers, Tower 1, #8P Kalinga Rao Road, Sampangiram Nagar, Bangalore, Karnataka, 560027, India
| | - Radheshyam Naik
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Govind Babu
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | | | | | - Krithika Murugan
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Mahesh B
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | - Lohith Reddy
- HealthCare Global Enterprises Limited, Bangalore, Karnataka, 560027, India
| | | |
Collapse
|
3
|
Aroonthongsawat P, Manocheewa S, Srisawat C, Punnakitikashem P, Suwanwong Y. Enhancement of the in vitro anti-leukemic effect of the histone deacetylase inhibitor romidepsin using Poly-(D, L-lactide-co-glycolide) nanoparticles as a drug carrier. Eur J Pharm Sci 2025; 207:107043. [PMID: 39952370 DOI: 10.1016/j.ejps.2025.107043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
The goal of this work is to develop a delivery system for histone deacetylase inhibitor (HDACi) romidepsin (ROM) using Poly(D, L-lactide-co-glycolide) as a carrier and evaluate its anti-leukemic effects. Romidepsin-loaded nanoparticles (ROM NPs) required for this purpose were fabricated using a single emulsion-solvent evaporation technique. Their physical characteristics and in vitro drug release profiles were studied, alongside biocompatibility and hemocompatibility assessments. Cell viability assays and Annexin V/Propidium Iodide (PI) staining were conducted to evaluate the anti-leukemic and apoptosis induction efficiency of ROM NPs in vitro. ROM NPs displayed a spherical shape with an average hydrodynamic size of about 149.7 ± 8.4 nm, a PDI of 0.11 ± 0.03, and a zeta potential of -25.27 ± 2.12 mV. The nanoparticles demonstrated a high encapsulation efficiency of ROM (∼93 %) and these nanoparticles effectively entered acute leukemia cells, including U937 and Jurkat. ROM NPs also exhibited a prolonged biphasic release pattern, specifically, the initial burst release phase occurred within the first 24 h, followed by a slower, sustained release. Additionally, they showed no hematological or biological toxicity, indicating their potential use for the delivery of anti-cancer drugs through the circulatory system. In tests on acute leukemia cell lines, ROM NPs showed significantly stronger anti-leukemic effects and induced apoptosis to a greater extent compared to free ROM. In summary, ROM NPs represent a promising therapy option for leukemia according to their enhanced anti-leukemic effects. Further modification of this strategy could be performed to enable target specificity, hence minimizing damage to normal cells.
Collapse
Affiliation(s)
- Pinyadapat Aroonthongsawat
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Siriphan Manocheewa
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chatchawan Srisawat
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Bangkok 10700, Thailand; Siriraj Center of Research Excellence in Theranostic Nanomedicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Primana Punnakitikashem
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Bangkok 10700, Thailand; Siriraj Center of Research Excellence in Theranostic Nanomedicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Yaneenart Suwanwong
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Center of Excellence for Biosensors and Bioengineering (CEBB), Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
4
|
Ko HC, Strickland KC, Jaggessarsingh D, Dillard A, Green M, Newburn E, Sellaro T, Klein JL, Caveney B, Eisenberg M, Severson EA, Ramkissoon S, Previs RA, Toumeh A. From tissue-specific to tissue-agnostic: HER2 overexpression and the rise of antibody-drug conjugates. Front Oncol 2025; 15:1565872. [PMID: 40098704 PMCID: PMC11911164 DOI: 10.3389/fonc.2025.1565872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
The growing importance of HER2 expression as a biomarker across multiple cancers is largely driven by advances in HER2-directed antibody-drug conjugates. The recent approval of trastuzumab deruxtecan (T-DXd) as a tumor-agnostic therapy has revolutionized treatment strategies for HER2-overexpressed tumors beyond breast, gastric, and colorectal cancers (CRC). This mini-review explores the evolving role of assessing HER2 overexpression in pan-solid tumors, following the recent approval of T-DXd as a tumor-agnostic therapy. It examines how HER2 scoring criteria for pan-tumor indications rely on immunohistochemistry (IHC) assessment, which may be prone to subjective interpretation and interobserver variability, and how these criteria differ from those used in breast, gastric, and CRC tumors. We also address the potential for NGS approaches to identify ERBB2 copy number gain (CNG) and the utility of artificial intelligence (AI) algorithms to enhance the consistency and accuracy of HER2 score interpretation for T-DXd treatment eligibility in solid tumors.
Collapse
Affiliation(s)
| | - Kyle C. Strickland
- Labcorp, Durham, NC, United States
- Department of Pathology, Duke University Medical Center, Duke Cancer Institute, Durham, NC, United States
| | | | | | | | | | | | | | | | | | | | - Shakti Ramkissoon
- Labcorp, Durham, NC, United States
- Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Rebecca A. Previs
- Labcorp, Durham, NC, United States
- Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, Duke University Medical Center, Duke Cancer Institute, Durham, NC, United States
| | - Anis Toumeh
- Hospital Corporation of America (HCA) Midwest Oncology Associates, Overland Park, KS, United States
| |
Collapse
|
5
|
Sang D, Su Y, Zhang Y, Guan Y, Fan S, Zhang J, Zheng L, Wang Y, Guo Y, Lei Z, Li M, Yuan P. Efficacy and safety of trastuzumab deruxtecan in Chinese patients with HER2-positive and HER2-low advanced breast cancer: a multicenter, observational, real-world study. Ther Adv Med Oncol 2025; 17:17588359251318853. [PMID: 40034603 PMCID: PMC11873904 DOI: 10.1177/17588359251318853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
Objective Limited real-world efficacy and safety data exist regarding the use of trastuzumab deruxtecan (T-DXd) in the Chinese population with human epidermal growth factor receptor (HER2)-positive and HER2-low advanced breast cancer (BC). This multicenter, observational, real-world study aimed to evaluate the efficacy and safety of T-DXd for the treatment of Chinese patients with HER2-positive and HER2-low advanced BC. Methods The medical records of 61 patients were collected from The Second Hospital of Dalian Medical University, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing Jingxin Hospital, and Cancer Hospital of the Chinese Academy of Medical Sciences. The primary endpoint of the study was progression-free survival (PFS), and the secondary endpoints were overall survival (OS), objective response rate (ORR), disease control rate (DCR), time to response (TTR), and safety. PFS and OS were analyzed using the Kaplan-Meier method and log-rank test. Results The primary endpoint, PFS was 10.51 months (95% confidence interval (CI), 3.02-NE) in the HER2-low group and 10.18 months (95% CI, 3.88-NE) in the HER2-positive group. Regarding the secondary endpoints in the HER2-low and HER2-positive groups, OS data were immature, ORR rates were 37.93% and 62.50%, DCR rates were 79.31% and 87.50%, and the median TTR rates were 1.28 and 1.31 months, respectively. In the subgroup analysis, front-line treatment with T-DXd was associated with increased beneficial effects. The primary adverse events (AEs) related to T-DXd treatment were gastrointestinal reactions and bone marrow suppression, which were predominantly grades 1-2, with no severe grade 4/5 AEs reported, only one patient developed infectious pneumonia. Conclusion This study was the first multicenter, real-world study of T-DXd for advanced BC in China. The findings demonstrated that T-DXd may be an effective antitumor treatment with controllable adverse reactions in patients with advanced BC irrespective of HER2 expression levels.
Collapse
Affiliation(s)
- Die Sang
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, China
| | - Yanfang Su
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, China
| | - Yurong Zhang
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, China
| | - Yanfeng Guan
- Department of Oncology, Beijing Jingxin Hospital, Beijing, China
| | - Shanmin Fan
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, China
| | - Jintao Zhang
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, China
| | - Lijun Zheng
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, China
| | - Yanling Wang
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, China
| | - Ying Guo
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, China
| | - Zixuan Lei
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Man Li
- Department of Oncology, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, 116023, China
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| |
Collapse
|
6
|
Gu H, Zhu T, Ding J, Yang Z, Qi S, Guo G. Real-World Analysis of the Efficacy and Adverse Events of T-DM1 in Chinese Patients With HER2-Positive Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:201-210. [PMID: 40008213 PMCID: PMC11853105 DOI: 10.2147/bctt.s503150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/25/2025] [Indexed: 02/27/2025]
Abstract
Purpose This study efforts to explore the association of adverse events (AEs) with efficacy in HER2-positive breast cancer patients treated with TDM1. Methods and Materials This retrospective study included women diagnosed with HER2+ BC treated with TDM1 from January 2012 to December 2023. Event-free survival (EFS) was the endpoint. Tumour response was assessed by disease control rate (DCR) and objective response rate (ORR). The chi-squared test, analysis of variance (ANOVA), Cox proportional hazards regression and Kaplan-Meier survival analysis was employed to evaluate the association of AEs with tumour efficacy. Results A total of 48 women with a median age of 52 years (median follow-up 8.4 months) were included in the study. Among them, 33 patients (68.8%) experienced adverse events, including platelet depletion and liver function abnormalities, 3 patients (6.3%) discontinued TDM1 due to severe platelet depletion. The overall objective response rate (ORR) was 25.0% and the disease control rate (DCR) was 43.8%. Using the Chi-squared test, we found a statistically significant difference in ORR and DCR between patients who developed a platelet reduction and those who did not. DCR was significantly higher in patients with liver dysfunction than in those without. ANOVA showed that exposure to hepatic dysfunction and platelet reduction, lines of therapy, and treatment course were associated with EFS. In the Kaplan-Meier survival analysis, both liver dysfunction and platelet reduction were correlated with significantly longer EFS (p=0.033 and p=0.038, respectively). Conclusion This retrospective study demonstrated that AEs were associated with tumour efficacy in patients with HER2+ BC treated with TDM1.
Collapse
Affiliation(s)
- Huayan Gu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Teng Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - JiaLing Ding
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Zhi Yang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Shuangyi Qi
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Guilong Guo
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| |
Collapse
|
7
|
Ford AL, Taft CW, Sprague-Getsy AM, Carlson GC, Mate NA, Sieburg MA, Chisholm JD, Hougland JL. A Modular Customizable Ligand-Conjugate (LC) System Targeting Ghrelin O-Acyltransferase. Biomolecules 2025; 15:204. [PMID: 40001510 PMCID: PMC11852496 DOI: 10.3390/biom15020204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/05/2025] [Accepted: 01/08/2025] [Indexed: 02/27/2025] Open
Abstract
Ghrelin is a 28 amino acid peptide hormone that impacts a wide range of biological processes, including appetite regulation, glucose metabolism, growth hormone regulation, and cognitive function. To bind and activate its cognate receptor, ghrelin must be acylated on a serine residue in a post-translational modification performed by ghrelin O-acyltransferase (GOAT). GOAT is a membrane-bound O-acyltransferase (MBOAT) responsible for the catalysis of the addition of an octanoyl fatty acid to the third serine of desacyl ghrelin. Beyond its canonical role for ghrelin maturation in endocrine cells within the stomach, GOAT was recently reported to be overexpressed in prostate cancer (PCa) cells and detected at increased levels in the serum and urine of PCa patients. This suggests GOAT can serve as a potential route for the detection and therapeutic targeting of PCa and other diseases that exhibit GOAT overexpression. Building upon a ghrelin mimetic peptide with nanomolar affinity for GOAT, we developed an antibody-conjugate-inspired system for customizable ligand-conjugate (LC) synthesis allowing for the attachment of a wide range of cargoes. The developed synthetic scheme allows for the easy synthesis of the desired LCs and demonstrates that our ligand system tolerates an extensive palette of cargoes while maintaining nanomolar affinity against GOAT.
Collapse
Affiliation(s)
- Amber L. Ford
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Caine W. Taft
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Andrea M. Sprague-Getsy
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Gracie C. Carlson
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Nilamber A. Mate
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Michelle A. Sieburg
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - John D. Chisholm
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
- BioInspired Syracuse, Syracuse University, Syracuse, NY 13244, USA
| | - James L. Hougland
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
- BioInspired Syracuse, Syracuse University, Syracuse, NY 13244, USA
- Department of Biology, Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
8
|
Elshazly AM, Elzahed AA, Gewirtz DA. Evidence for cytoprotective autophagy in response to HER2-targeted monoclonal antibodies. J Pharmacol Exp Ther 2025; 392:100007. [PMID: 39892993 DOI: 10.1124/jpet.123.002048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/03/2024] [Accepted: 02/11/2024] [Indexed: 03/09/2024] Open
Abstract
The advent of HER2-targeted monoclonal antibodies such as trastuzumab has significantly improved the clinical outcomes for patients with breast cancer overexpressing HER2 and, more recently, also for gastric cancers. However, the development of resistance, as is frequently the case for other antineoplastic modalities, constrains their clinical efficacy. Multiple molecular mechanisms and signaling pathways have been investigated for their potential involvement in the development of resistance to HER2-targeted therapies, among which is autophagy. Autophagy is an inherent cellular mechanism whereby cytoplasmic components are selectively degraded to maintain cellular homeostasis via the generation of energy and metabolic intermediates. Although the cytoprotective form of autophagy is thought to predominate, other forms of autophagy have also been identified in response to chemotherapeutic agents in various tumor models; these include cytotoxic, cytostatic, and nonprotective functional forms of autophagy. In this review, we provide an overview of the autophagic machinery induced in response to HER2-targeted monoclonal antibodies, with a focus on trastuzumab and trastuzumab-emtansine, in an effort to determine whether autophagy targeting or modulation could be translated clinically to increase their effectiveness and/or overcome the development of resistance. SIGNIFICANCE STATEMENT: This manuscript is one in a series of papers that interrogate the role(s) of the autophagy induced in response to antineoplastic agents in various cancer models. This series of papers was developed in an effort to establish whether autophagy targeting or modulation is likely to be an effective adjuvant strategy to increase the efficacy of cancer chemotherapeutic agents. This review explores the relationship between the autophagic machinery and HER2-targeted therapies.
Collapse
Affiliation(s)
- Ahmed M Elshazly
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Aya A Elzahed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
9
|
Mihaylova R, Momekova D, Elincheva V, Momekov G. Immunoconjugates as an Efficient Platform for Drug Delivery: A Resurgence of Natural Products in Targeted Antitumor Therapy. Pharmaceuticals (Basel) 2024; 17:1701. [PMID: 39770542 PMCID: PMC11677665 DOI: 10.3390/ph17121701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
The present review provides a detailed and comprehensive discussion on antibody-drug conjugates (ADCs) as an evolving new modality in the current therapeutic landscape of malignant diseases. The principle concepts of targeted delivery of highly toxic agents forsaken as stand-alone drugs are examined in detail, along with the biochemical and technological tools for their successful implementation. An extensive analysis of ADCs' major components is conducted in parallel with their function and impact on the stability, efficacy, safety, and resistance profiles of the immunoconjugates. The scope of the article covers the major classes of currently validated natural compounds used as payloads, with an emphasis on their structural and mechanistic features, natural origin, and distribution. Future perspectives in ADCs' design are thoroughly explored, addressing their inherent or emerging challenges and limitations. The survey also provides a comprehensive overview of the molecular rationale for active tumor targeting of ADC-based platforms, exploring the cellular biology and clinical relevance of validated tumor markers used as a "homing" mechanism in both hematological and solid tumor malignancies.
Collapse
Affiliation(s)
- Rositsa Mihaylova
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Denitsa Momekova
- Department “Pharmaceutical Technology and Biopharmaceutics”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria;
| | - Viktoria Elincheva
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Georgi Momekov
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| |
Collapse
|
10
|
Gadaleta-Caldarola G, Lanotte L, Santoro AN, Pinto A, Gadaleta-Caldarola A, Giacomelli L, Fedele P. Predictive Factors of Antibody-Drug Conjugate Treatment in Metastatic Breast Cancer: A Narrative Review. Cancers (Basel) 2024; 16:4082. [PMID: 39682268 DOI: 10.3390/cancers16234082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have revolutionized the treatment landscape for metastatic breast cancer, offering targeted delivery of cytotoxic agents with improved efficacy and tolerability compared to conventional chemotherapy. This narrative review explores key predictive factors influencing the efficacy of ADCs, focusing on HER2-targeted therapies, such as trastuzumab emtansine and trastuzumab deruxtecan, as well as sacituzumab govitecan for triple-negative breast cancer. HER2 expression, TROP-2 levels, hormone receptor status, and the tumor microenvironment emerge as critical biomarkers for patient selection and therapeutic outcomes. Additionally, we discuss resistance mechanisms, such as antigen loss, impaired drug internalization, and the role of circulating tumor DNA in predicting ADC response. Finally, future perspectives on the sequential use of ADCs and potential combination therapies are highlighted, along with emerging agents targeting alternative antigens like HER3 and LIV-1. Overall, identifying predictive biomarkers and overcoming resistance mechanisms are essential for optimizing the use of ADCs in metastatic breast cancer, thereby improving patient outcomes.
Collapse
Affiliation(s)
| | - Laura Lanotte
- Oncology Unit, "Mons. A. R. Dimiccoli" Hospital, 70051 Barletta, Italy
| | | | - Antonello Pinto
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| | | | | | - Palma Fedele
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| |
Collapse
|
11
|
Goldberg SD, Satomaa T, Aina O, Aitio O, Burke K, Dudkin V, Geist B, Irrechukwu O, Hänninen AL, Heiskanen A, Helin J, Hiltunen JO, Kinyamu-Akunda J, Klein DM, Kohli N, Kotiranta T, Lähteenmäki T, Niemelä R, Pitkänen V, Pynnönen H, Rittase W, Wiley K, Zhou J, Saarinen J. Trastuzumab-MMAU Antibody-Auristatin Conjugates: Valine-Glucoserine Linker with Stabilized Maleimide Conjugation Improves In Vivo Efficacy and Tolerability. Mol Cancer Ther 2024; 23:1530-1543. [PMID: 38324296 DOI: 10.1158/1535-7163.mct-23-0591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/16/2023] [Accepted: 02/01/2024] [Indexed: 02/08/2024]
Abstract
Antibody-drug conjugates (ADC) have shown impressive clinical activity with approval of many agents in hematologic and solid tumors. However, challenges remain with both efficacy and safety of ADCs. This study describes novel trastuzumab-auristatin conjugates with the hydrophilic monomethylauristatin E (MMAE) prodrug MMAU, and optimization of a glycopeptide linker leading to a wider therapeutic window. Trastuzumab was conjugated with auristatin payloads via a series of linkers using a stabilized maleimide handle. The ADCs were characterized in vitro and their relative in vivo antitumor efficacies were assessed in HER2+ xenograft models. Relative linker stabilities and the mechanism of linker cleavage were studied using in vitro assays. Toxicity and toxicokinetics of the best performing ADC were evaluated in cynomolgus monkey (cyno). The trastuzumab-MMAU ADC with stabilized glycopeptide linker showed maleimide stabilization and higher resistance to cleavage by serum and lysosomal enzymes compared with a valine-citrulline conjugated trastuzumab ADC (trastuzumab-vc-MMAE). A single dose of 1 or 2 mg/kg of trastuzumab-MMAU at drug-to-antibody ratios (DAR) of eight and four respectively resulted in xenograft tumor growth inhibition, with superior efficacy to trastuzumab-vc-MMAE. Trastuzumab-MMAUDAR4 was tolerated at doses up to 12 mg/kg in cyno, which represents 2- to 4-fold higher dose than that observed with vedotin ADCs, and had increased terminal half-life and exposure. The optimized trastuzumab-MMAU ADC showed potent antitumor activity and was well tolerated with excellent pharmacokinetics in nonhuman primates, leading to a superior preclinical therapeutic window. The data support potential utility of trastuzumab-MMAU for treatment of HER2+ tumors.
Collapse
Affiliation(s)
| | | | - Olulanu Aina
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | | | - Krista Burke
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | - Vadim Dudkin
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | - Brian Geist
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | | | | | | | | | | | | | | | - Neeraj Kohli
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | | | | | | | | | | | | | | | - Junguo Zhou
- Janssen Pharmaceuticals, Spring House, Pennsylvania
| | | |
Collapse
|
12
|
Saha N, Lee SG, Brockmann EC, de la Cruz MJ, Goldgur Y, Mendoza RP, Stanchina ED, Love TM, Marvald J, Xu Y, Xu K, Himanen JP, Lamminmäki U, Veach D, Nikolov DB. Fully human monoclonal antibody targeting the cysteine-rich substrate-interacting region of ADAM17 on cancer cells. Biomed Pharmacother 2024; 180:117605. [PMID: 39461016 PMCID: PMC11787792 DOI: 10.1016/j.biopha.2024.117605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/05/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
ADAM17 sheds EGFR/erbB ligands and triggers oncogenic pathways that lead to the progression of solid tumors. We targeted the ADAM17 disintegrin and cysteine rich domain region (D+C) to generate a panel of single-chain antibody fragments (scFvs) that selectively bind to the D or C domains of ADAM17, but not of ADAM10 or ADAM19. From the panel, we selected one scFv, referred to as C12, based on its high binding affinity towards the target, and re-formatted it to a full IgG for further studies. High-resolution cryo-electron microscopy studies documented that the mAb binds to the ADAM17 C-domain that in ADAM proteases, notably ADAM10 and ADAM17, is known to impart substrate-specificity. The C12 mAb significantly inhibited EGFR phosphorylation in cancer cell lines by hindering the cleavage of EGFR ligands tethered to the cell surface. This inhibition provides a mechanism for potential anti-tumor effects, and indeed C12 diminished the viability of a variety of EGFR-expressing cancer cell lines. Cell-based ELISA studies revealed that C12 preferentially bound to activated ADAM17 present on tumor cells, as compared to the autoinhibited ADAM17 that is the predominant form on HEK293 and other non-tumor cells. C12 also exhibited tumor growth inhibition in an ovarian cancer xenograft mouse model. Consistent with its selective tumor cell binding in vitro, radioimmuno PET (positron emission tomography) imaging with 89Zr-DFO-C12 in mouse xenograft models confirmed tumoral accumulation of the C12 mAb.
Collapse
Affiliation(s)
- Nayanendu Saha
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| | - Sang Gyu Lee
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | | | - M Jason de la Cruz
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Yehuda Goldgur
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Rachelle P Mendoza
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Elisa de Stanchina
- Antitumor Assessment Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Tanzy M Love
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - Josh Marvald
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - Yan Xu
- Texas Therapeutic Institute, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Kai Xu
- Texas Therapeutic Institute, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Juha P Himanen
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Urpo Lamminmäki
- Department of Life Technologies, University of Turku, Turku, Finland
| | - Darren Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Dimitar B Nikolov
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
13
|
Gao X, Bu T, Wang W, Xu Y. Comparative Analysis and Future Prospects of Human Epidermal Growth Factor Receptor 2 (HER2) and Trophoblast Cell-Surface Antigen 2 (Trop-2) Targeted Antibody-Drug Conjugates in Breast Cancer Treatment. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:621-630. [PMID: 39310781 PMCID: PMC11416103 DOI: 10.2147/bctt.s480796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024]
Abstract
Breast cancer remains the most prevalent malignancy among women globally, presenting significant challenges in therapeutic strategies due to tumor heterogeneity, drug resistance, and adverse side effects. Recent advances in targeted therapies, particularly antibody-drug conjugates (ADCs), have shown promise in addressing these challenges by selectively targeting tumor cells while sparing normal tissues. This study provides a comprehensive analysis of two innovative ADCs targeting HER2 and Trop-2, which are critical markers in various breast cancer subtypes. These conjugates combine potent cytotoxic drugs with specific antibodies, leveraging the antigens' differential expression to enhance therapeutic efficacy and reduce systemic toxicity. Our comparative analysis highlights the clinical applications, efficacy, and safety profiles of these ADCs, drawing on data from recent clinical trials. In addition, the paper discusses the potential of these ADCs in treating other types of cancers where HER2 and Trop-2 are expressed, as well as the toxicity risks associated with targeting these antigens in normal cells. Additionally, the paper discusses novel synthetic drugs that show potential in preclinical models, focusing on their mechanisms of action and therapeutic advantages over traditional chemotherapy. The findings underscore the transformative impact of targeted ADCs in breast cancer treatment, noting significant advancements in patient outcomes and management of side effects. However, ongoing issues such as resistance mechanisms and long-term safety remain challenges. The conclusion offers a forward-looking perspective on potential improvements and the future trajectory of ADC research. This study not only elucidates the current landscape of ADCs in breast cancer but also sets the stage for the next generation of oncological therapeutics. This study not only elucidates the current landscape of ADCs in breast cancer but also sets the stage for the next generation of oncological therapeutics, with particular attention to their broader applications and associated risks.
Collapse
Affiliation(s)
- Xiaojuan Gao
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, Gansu Province, People’s Republic of China
| | - Tiansheng Bu
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, Gansu Province, People’s Republic of China
| | - Wenying Wang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, Gansu Province, People’s Republic of China
| | - Ying Xu
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, Gansu Province, People’s Republic of China
| |
Collapse
|
14
|
Panaampon J, Sungwan P, Fujikawa S, Sampattavanich S, Jirawatnotai S, Okada S. Trastuzumab, a monoclonal anti-HER2 antibody modulates cytotoxicity against cholangiocarcinoma via multiple mechanisms. Int Immunopharmacol 2024; 138:112612. [PMID: 38968862 DOI: 10.1016/j.intimp.2024.112612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024]
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.
Collapse
Affiliation(s)
- Jutatip Panaampon
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan; Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Prin Sungwan
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Sawako Fujikawa
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Somponnat Sampattavanich
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Siwanon Jirawatnotai
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan; Institute of Industrial Nanomaterials, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-8555, Japan.
| |
Collapse
|
15
|
Papa F, Grinda T, Rassy E, Cheickh-Hussin R, Ribeiro J, Antonuzzo L, Pistilli B. Long road towards effective HER3 targeting in breast cancer. Cancer Treat Rev 2024; 129:102786. [PMID: 38885540 DOI: 10.1016/j.ctrv.2024.102786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/25/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer is a heterogeneous disease, encompassing multiple different subtypes. Thanks to the increasing knowledge of the diverse biological features of each subtype, most patients receive personalized treatment based on known biomarkers. However, the role of some biomarkers in breast cancer evolution is still unknown, and their potential use as a therapeutic target is still underexplored. HER3 is a member of the human epidermal growth factors receptor family, overexpressed in 50%-70% of breast cancers. HER3 plays a key role in cancer progression, metastasis development, and drug resistance across all the breast cancer subtypes. Owing to its critical role in cancer progression, many HER3-targeting therapies have been developed over the past decade with conflicting findings. Next-generation antibody-drug conjugates have recently shown promising results in solid tumors expressing HER3, including breast cancer. In this review, we discuss the HER3 role in the pathogenesis of breast cancer and its relevance across all subtypes. We also explore the new anti-HER3 treatment strategies, calling into question the significance of HER3 detection as crucial information in breast cancer treatment.
Collapse
Affiliation(s)
- Francesca Papa
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Department of Medical Oncology, Florence University, Italy
| | - Thomas Grinda
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | | | - Joana Ribeiro
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | | | - Barbara Pistilli
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; INSERM U1279, Gustave Roussy, Villejuif, France.
| |
Collapse
|
16
|
Jain S, Griffith JI, Porath KA, Rathi S, Le J, Pasa TI, Decker PA, Gupta SK, Hu Z, Carlson BL, Bakken K, Burgenske DM, Feldsien TM, Lefebvre DR, Vaubel RA, Eckel-Passow JE, Reilly EB, Elmquist WF, Sarkaria JN. Bystander Effects, Pharmacokinetics, and Linker-Payload Stability of EGFR-Targeting Antibody-Drug Conjugates Losatuxizumab Vedotin and Depatux-M in Glioblastoma Models. Clin Cancer Res 2024; 30:3287-3297. [PMID: 38743766 PMCID: PMC11292202 DOI: 10.1158/1078-0432.ccr-24-0426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/05/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
PURPOSE Antibody-drug conjugates (ADC) are targeted therapies with robust efficacy in solid cancers, and there is intense interest in using EGFR-specific ADCs to target EGFR-amplified glioblastoma (GBM). Given GBM's molecular heterogeneity, the bystander activity of ADCs may be important for determining treatment efficacy. In this study, the activity and toxicity of two EGFR-targeted ADCs with similar auristatin toxins, Losatuxizumab vedotin (ABBV-221) and Depatuxizumab mafodotin (Depatux-M), were compared in GBM patient-derived xenografts (PDX) and normal murine brain following direct infusion by convection-enhanced delivery (CED). EXPERIMENTAL DESIGN EGFRviii-amplified and non-amplified GBM PDXs were used to determine in vitro cytotoxicity, in vivo efficacy, and bystander activities of ABBV-221 and Depatux-M. Nontumor-bearing mice were used to evaluate the pharmacokinetics (PK) and toxicity of ADCs using LC-MS/MS and immunohistochemistry. RESULTS CED improved intracranial efficacy of Depatux-M and ABBV-221 in three EGFRviii-amplified GBM PDX models (Median survival: 125 to >300 days vs. 20-49 days with isotype control AB095). Both ADCs had comparable in vitro and in vivo efficacy. However, neuronal toxicity and CD68+ microglia/macrophage infiltration were significantly higher in brains infused with ABBV-221 with the cell-permeable monomethyl auristatin E (MMAE), compared with Depatux-M with the cell-impermeant monomethyl auristatin F. CED infusion of ABBV-221 into the brain or incubation of ABBV-221 with normal brain homogenate resulted in a significant release of MMAE, consistent with linker instability in the brain microenvironment. CONCLUSIONS EGFR-targeting ADCs are promising therapeutic options for GBM when delivered intratumorally by CED. However, the linker and payload for the ADC must be carefully considered to maximize the therapeutic window.
Collapse
Affiliation(s)
- Sonia Jain
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Jessica I. Griffith
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota.
| | - Kendra A. Porath
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Sneha Rathi
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota.
| | - Jiayan Le
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota.
| | - Tugce I. Pasa
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.
| | - Paul A. Decker
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota.
| | - Shiv K. Gupta
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Zeng Hu
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Brett L. Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Katrina Bakken
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | | | | | | | - Rachael A. Vaubel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.
| | | | | | - William F. Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota.
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
17
|
Cai A, Chen Y, Wang LS, Cusick JK, Shi Y. Depicting Biomarkers for HER2-Inhibitor Resistance: Implication for Therapy in HER2-Positive Breast Cancer. Cancers (Basel) 2024; 16:2635. [PMID: 39123362 PMCID: PMC11311605 DOI: 10.3390/cancers16152635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
HER2 (human epidermal growth factor receptor 2) is highly expressed in a variety of cancers, including breast, lung, gastric, and pancreatic cancers. Its amplification is linked to poor clinical outcomes. At the genetic level, HER2 is encoded by the ERBB2 gene (v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2), which is frequently mutated or amplified in cancers, thus spurring extensive research into HER2 modulation and inhibition as viable anti-cancer strategies. An impressive body of FDA-approved drugs, including anti-HER2 monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), and HER2-tyrosine kinase inhibitors (TKIs), have demonstrated success in enhancing overall survival (OS) and disease progression-free survival (PFS). Yet, drug resistance remains a persistent challenge and raises the risks of metastatic potential and tumor relapse. Research into alternative therapeutic options for HER2+ breast cancer therefore proves critical for adapting to this ever-evolving landscape. This review highlights current HER2-targeted therapies, discusses predictive biomarkers for drug resistance, and introduces promising emergent therapies-especially combination therapies-that are aimed at overcoming drug resistance in the context of HER2+ breast cancer.
Collapse
Affiliation(s)
- Alvan Cai
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yuan Chen
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany;
| | - Lily S. Wang
- University of California, Berkeley, CA 94720, USA;
| | - John K. Cusick
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yihui Shi
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
- California Pacific Medical Center Research Institute, Sutter Bay Hospitals, San Francisco, CA 94107, USA
| |
Collapse
|
18
|
Chen YS, Jin E, Day PJ. Use of Drug Sensitisers to Improve Therapeutic Index in Cancer. Pharmaceutics 2024; 16:928. [PMID: 39065625 PMCID: PMC11279903 DOI: 10.3390/pharmaceutics16070928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The clinical management of malignant tumours is challenging, often leading to severe adverse effects and death. Drug resistance (DR) antagonises the effectiveness of treatments, and increasing drug dosage can worsen the therapeutic index (TI). Current efforts to overcome DR predominantly involve the use of drug combinations, including applying multiple anti-cancerous drugs, employing drug sensitisers, which are chemical agents that enhance pharmacokinetics (PK), including the targeting of cellular pathways and regulating pertinent membrane transporters. While combining multiple compounds may lead to drug-drug interactions (DDI) or polypharmacy effect, the use of drug sensitisers permits rapid attainment of effective treatment dosages at the disease site to prevent early DR and minimise side effects and will reduce the chance of DDI as lower drug doses are required. This review highlights the essential use of TI in evaluating drug dosage for cancer treatment and discusses the lack of a unified standard for TI within the field. Commonly used benefit-risk assessment criteria are summarised, and the critical exploration of the current use of TI in the pharmaceutical industrial sector is included. Specifically, this review leads to the discussion of drug sensitisers to facilitate improved ratios of effective dose to toxic dose directly in humans. The combination of drug and sensitiser molecules might see additional benefits to rekindle those drugs that failed late-stage clinical trials by the removal of detrimental off-target activities through the use of lower drug doses. Drug combinations and employing drug sensitisers are potential means to combat DR. The evolution of drug combinations and polypharmacy on TI are reviewed. Notably, the novel binary weapon approach is introduced as a new opportunity to improve TI. This review emphasises the urgent need for a criterion to systematically evaluate drug safety and efficiency for practical implementation in the field.
Collapse
Affiliation(s)
- Yu-Shan Chen
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (Y.-S.C.); (E.J.)
| | - Enhui Jin
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (Y.-S.C.); (E.J.)
| | - Philip J. Day
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (Y.-S.C.); (E.J.)
- Department of Medicine, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
19
|
Kınıkoğlu O, Odabas H, Altıntaş YE, Yıldız A, Çakan B, Akdağ G, Yıldırım S, Bal H, Kaya T, Tünbekici S, Işık D, Başoğlu T, Yıldırım ME, Turan N. Combining Endocrine Therapy with Trastuzumab Emtansine Improves Progression-Free Survival and Overall Survival in HER2-Positive, Hormone Receptor-Positive Metastatic Breast Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:951. [PMID: 38929568 PMCID: PMC11205527 DOI: 10.3390/medicina60060951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: Patients with human epidermal growth factor receptor 2 (HER2) -positive, hormone receptor-positive (HR-positive) metastatic breast cancer (MBC) usually undergo trastuzumab emtansine (T-DM1) therapy in subsequent lines. Combining endocrine therapy (ET) with T-DM1 can improve treatment outcomes in this subtype. Therefore, this study aimed to investigate the benefits of using T-DM1 with ET in HER2-positive and HR-positive MBC. This study was the first to investigate the benefits of combining ET with T-DM1. Material and Methods: This study analyzed the medical records of patients with HER2-positive and HR-positive MBC who were treated with T-DM1 from June 2010 to December 2021. The patients were divided into groups based on whether they received concomitant ET with T-DM1. The primary endpoint was to determine the progression-free survival (PFS), while the secondary endpoints were overall survival (OS), objective response rate, and safety of the treatment. Results: Our analysis examined 88 patients, of whom 32 (36.4%) were treated with T-DM1 in combination with ET. The combination therapy showed a significant improvement in median PFS (15.4 vs. 6.4 months; p = 0.00004) and median OS (35.0 vs. 23.1 months; p = 0.026) compared to T-DM1 alone. The ORR was also higher in the combination group (65.6% vs. 29.3%; p = 0.026). Patients treated with pertuzumab priorly had reduced median PFS on T-DM1 compared to those who were not treated with pertuzumab (11.7 vs. 5.4 months, respectively; p < 0.01). T-DM1 demonstrated better median PFS in HER2 3+ patients compared to HER2 2+ patients, with an amplification ratio of >2.0 (10.8 vs 5.8 months, respectively; p = 0.049). The safety profiles were consistent with previous T-DM1 studies. Conclusions: The combination of T-DM1 with ET can significantly improve PFS and OS in patients with HER2-positive and HR-positive MBC. Our study suggests that prior pertuzumab treatment plus trastuzumab treatment might decrease T-DM1 efficacy.
Collapse
Affiliation(s)
- Oğuzcan Kınıkoğlu
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Hatice Odabas
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Yunus Emre Altıntaş
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Anıl Yıldız
- Department of Medical Oncology, Istanbul University Oncology Institute, Istanbul 34093, Turkey;
| | - Burçin Çakan
- Department of Medical Oncology, Bağcılar Research and Training Hospital, Istanbul 34212, Turkey;
| | - Goncagül Akdağ
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Sedat Yıldırım
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Hamit Bal
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Tuğba Kaya
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Salih Tünbekici
- Department of Medical Oncology, Ege University Faculty of Medicine, Izmir 35100, Turkey;
| | - Deniz Işık
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Tuğba Başoğlu
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Mahmut Emre Yıldırım
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| | - Nedim Turan
- Department of Medical Oncology, Health Science University, Kartal Dr. Lütfi Kirdar City Hospital, Istanbul 34865, Turkey; (H.O.); (Y.E.A.); (G.A.); (S.Y.); (H.B.); (T.K.); (D.I.); (T.B.); (M.E.Y.); (N.T.)
| |
Collapse
|
20
|
Borstnar S, Bozovic-Spasojevic I, Cvetanovic A, Plavetic ND, Konsoulova A, Matos E, Popovic L, Popovska S, Tomic S, Vrdoljak E. Advancing HER2-low breast cancer management: enhancing diagnosis and treatment strategies. Radiol Oncol 2024; 58:258-267. [PMID: 38860691 PMCID: PMC11165977 DOI: 10.2478/raon-2024-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/14/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Recent evidence brought by novel anti-human epidermal growth factor receptor 2 (HER2) antibody-drug conjugates is leading to significant changes in HER2-negative breast cancer (BC) best practices. A new targetable category termed 'HER2-low' has been identified in tumors previously classified as 'HER2-negative'. Daily practice in pathology and medical oncology is expected to align to current recommendations, but patient access to novel anticancer drugs across geographies might be impeded due to local challenges. MATERIALS AND METHODS An expert meeting involving ten regional pathology and oncology opinion leaders experienced in BC management in four Central and Eastern Europe (CEE) countries (Bulgaria, Croatia, Serbia, Slovenia) was held. Herein we summarized the current situation of HER2-low metastatic BC (mBC), local challenges, and action plans to prevent delays in patient access to testing and treatment based on expert opinion. RESULTS Gaps and differences at multiple levels were identified across the four countries. These included variability in the local HER2-low epidemiology data, certification of pathology laboratories and quality control, and reimbursement conditions of testing and anticancer drugs for HER2-negative mBC. While clinical decisions were aligned to international guidelines in use, optimal access to testing and innovative treatment was restricted due to significant delays in reimbursement or limitative reimbursement conditions. CONCLUSIONS Preventing delays in HER2-low mBC patient access to diagnosis and novel treatments is crucial to optimize outcomes. Multidisciplinary joint efforts and pro-active discussions between clinicians and decision makers are needed to improve care of HER2-low mBC patients in CEE countries.
Collapse
Affiliation(s)
| | | | - Ana Cvetanovic
- Department of Oncology, Medical Faculty University of Niš; Clinic of Oncology, University Clinical Centre Niš, Serbia
| | | | | | | | - Lazar Popovic
- Oncology Institute of Vojvodina, Faculty of Medicine, University Novi Sad, Novi Sad, Serbia
| | | | - Snjezana Tomic
- University Hospital of Split, University of Split - School of Medicine, Croatia
| | - Eduard Vrdoljak
- University Hospital of Split, University of Split - School of Medicine, Croatia
| |
Collapse
|
21
|
Calopiz MC, Linderman JJ, Thurber GM. Optimizing Solid Tumor Treatment with Antibody-drug Conjugates Using Agent-Based Modeling: Considering the Role of a Carrier Dose and Payload Class. Pharm Res 2024; 41:1109-1120. [PMID: 38806889 DOI: 10.1007/s11095-024-03715-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
INTRODUCTION Antibody-drug conjugates (ADCs) show significant clinical efficacy in the treatment of solid tumors, but a major limitation to their success is poor intratumoral distribution. Adding a carrier dose improves both distribution and overall drug efficacy of ADCs, but the optimal carrier dose has not been outlined for different payload classes. OBJECTIVE In this work, we study two carrier dose regimens: 1) matching payload potency to cellular delivery but potentially not reaching cells farther away from blood vessels, or 2) dosing to tumor saturation but risking a reduction in cell killing from a lower amount of payload delivered per cell. METHODS We use a validated computational model to test four different payloads conjugated to trastuzumab to determine the optimal carrier dose as a function of target expression, ADC dose, and payload potency. RESULTS We find that dosing to tumor saturation is more efficacious than matching payload potency to cellular delivery for all payloads because the increase in the number of cells targeted by the ADC outweighs the loss in cell killing on targeted cells. An important exception exists if the carrier dose reduces the payload uptake per cell to the point where all cell killing is lost. Likewise, receptor downregulation can mitigate the benefits of a carrier dose. CONCLUSIONS Because tumor saturation and in vitro potency can be measured early in ADC design, these results provide insight into maximizing ADC efficacy and demonstrate the benefits of using simulation to guide ADC design.
Collapse
Affiliation(s)
- Melissa C Calopiz
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
22
|
Lynch SM, Heeran AB, Burke C, Lynam-Lennon N, Eustace AJ, Dean K, Robson T, Rahman A, Marcone S. Precision Oncology, Artificial Intelligence, and Novel Therapeutic Advancements in the Diagnosis, Prevention, and Treatment of Cancer: Highlights from the 59th Irish Association for Cancer Research (IACR) Annual Conference. Cancers (Basel) 2024; 16:1989. [PMID: 38893110 PMCID: PMC11171401 DOI: 10.3390/cancers16111989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Advancements in oncology, especially with the era of precision oncology, is resulting in a paradigm shift in cancer care. Indeed, innovative technologies, such as artificial intelligence, are paving the way towards enhanced diagnosis, prevention, and personalised treatments as well as novel drug discoveries. Despite excellent progress, the emergence of resistant cancers has curtailed both the pace and extent to which we can advance. By combining both their understanding of the fundamental biological mechanisms and technological advancements such as artificial intelligence and data science, cancer researchers are now beginning to address this. Together, this will revolutionise cancer care, by enhancing molecular interventions that may aid cancer prevention, inform clinical decision making, and accelerate the development of novel therapeutic drugs. Here, we will discuss the advances and approaches in both artificial intelligence and precision oncology, presented at the 59th Irish Association for Cancer Research annual conference.
Collapse
Affiliation(s)
- Seodhna M. Lynch
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Londonderry BT47 6SB, UK;
| | - Aisling B. Heeran
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (A.B.H.); (N.L.-L.); (S.M.)
| | - Caoimbhe Burke
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, D04 C1P1 Dublin, Ireland;
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (A.B.H.); (N.L.-L.); (S.M.)
| | - Alex J. Eustace
- Life Sciences Institute, Dublin City University, D09 NR58 Dublin, Ireland;
| | - Kellie Dean
- School of Biochemistry and Cell Biology, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland;
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Arman Rahman
- UCD School of Medicine, UCD Conway Institute, University College Dublin, Belfield, D04 C1P1 Dublin, Ireland;
| | - Simone Marcone
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (A.B.H.); (N.L.-L.); (S.M.)
| |
Collapse
|
23
|
Aupperle-Lellbach H, Kehl A, de Brot S, van der Weyden L. Clinical Use of Molecular Biomarkers in Canine and Feline Oncology: Current and Future. Vet Sci 2024; 11:199. [PMID: 38787171 PMCID: PMC11126050 DOI: 10.3390/vetsci11050199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Molecular biomarkers are central to personalised medicine for human cancer patients. It is gaining traction as part of standard veterinary clinical practice for dogs and cats with cancer. Molecular biomarkers can be somatic or germline genomic alterations and can be ascertained from tissues or body fluids using various techniques. This review discusses how these genomic alterations can be determined and the findings used in clinical settings as diagnostic, prognostic, predictive, and screening biomarkers. We showcase the somatic and germline genomic alterations currently available to date for testing dogs and cats in a clinical setting, discussing their utility in each biomarker class. We also look at some emerging molecular biomarkers that are promising for clinical use. Finally, we discuss the hurdles that need to be overcome in going 'bench to bedside', i.e., the translation from discovery of genomic alterations to adoption by veterinary clinicians. As we understand more of the genomics underlying canine and feline tumours, molecular biomarkers will undoubtedly become a mainstay in delivering precision veterinary care to dogs and cats with cancer.
Collapse
Affiliation(s)
- Heike Aupperle-Lellbach
- Laboklin GmbH&Co.KG, Steubenstr. 4, 97688 Bad Kissingen, Germany; (H.A.-L.); (A.K.)
- School of Medicine, Institute of Pathology, Technical University of Munich, Trogerstr. 18, 80333 München, Germany
| | - Alexandra Kehl
- Laboklin GmbH&Co.KG, Steubenstr. 4, 97688 Bad Kissingen, Germany; (H.A.-L.); (A.K.)
- School of Medicine, Institute of Pathology, Technical University of Munich, Trogerstr. 18, 80333 München, Germany
| | - Simone de Brot
- Institute of Animal Pathology, COMPATH, University of Bern, 3012 Bern, Switzerland;
| | | |
Collapse
|
24
|
Cecco S, Puligheddu S, Fusaroli M, Gerratana L, Yan M, Zamagni C, De Ponti F, Raschi E. Emerging Toxicities of Antibody-Drug Conjugates for Breast Cancer: Clinical Prioritization of Adverse Events from the FDA Adverse Event Reporting System. Target Oncol 2024; 19:435-445. [PMID: 38696126 PMCID: PMC11111510 DOI: 10.1007/s11523-024-01058-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) are gaining widespread use in the treatment of breast cancer, although toxicity remains an underexplored issue in the real-world clinical setting. Individual case safety reports collected in large pharmacovigilance databases can advance our knowledge on their safety profile in routine clinical practice. OBJECTIVE We prioritized adverse events (AEs) reported with ADCs approved for breast cancer using the Food and Drug Administration Adverse Event Reporting System (FAERS). METHODS We assessed clinical priority of AEs reported in FAERS (February 2013-March 2022) for trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), and sacituzumab govitecan (SG) by attributing a score to each AE disproportionally reported with ADCs. Four criteria were assessed: clinical relevance, reporting rate, reported case fatality rate, and stability of disproportionality signals (consistency of the reporting odds ratio across multiple analyses using three different comparators). RESULTS We retained 6589 reports (77.4% referring to T-DM1 as suspect), and 572 AEs generated a disproportionality signal in at least one analysis. The majority of these AEs (62%) were classified as moderate clinical priorities (e.g., interstitial lung disease with T-DXd, thrombocytopenia, peripheral neuropathy with T-DM1, febrile neutropenia, and large intestine perforation with SG). Three AEs emerged as high clinical priorities (6 points): septic shock and neutropenic colitis with SG (N = 8 and 13, with median onset 13 and 10 days, respectively), without co-reported immunosuppressive agents; and pulmonary embolism with T-DM1 (N = 31, median onset 109 days, 52% with reported metastasis). CONCLUSION The heterogeneous spectrum of post-marketing toxicities for ADCs used in breast cancer, as emerging from the FAERS, is largely in line with preapproval evidence. Although causality cannot be proved, we call for increased awareness by oncologists on potential serious unexpected reactions, including early onset of septic shock and neutropenic colitis with SG, and late emergence of pulmonary embolism with T-DM1.
Collapse
Affiliation(s)
- Sara Cecco
- Hospital Pharmacy Unit-CRO Aviano, National Cancer Institute, IRCCS, 33081, Aviano, PN, Italy.
| | - Stefano Puligheddu
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Michele Fusaroli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Lorenzo Gerratana
- Department of Medical Oncology-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
- Toxicology Counseling Center of Hunan Province, Changsha, China
| | - Claudio Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fabrizio De Ponti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Emanuel Raschi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
25
|
Bornstein-Quevedo L, de Anda-González J, Lara-Torres CO, Flores-Gutiérrez JP, Dorantes-Heredia R, Bautista-Piña V, Zaragoza-Vargas P, Alcaraz-Wong A, Soto-Sañudo AK, Mendoza-Ramírez S, Salamanca-García M, Loyola-Rodríguez G, Gómez-Macías GS, Murguia-Perez M, De Luna-Sánchez M, Villalobos-Valencia R, Talamantes E, Arce-Salinas C. Navigating HER2-Low Testing in Invasive Breast Cancer: Update Recommendations for Pathologists. J Pers Med 2024; 14:467. [PMID: 38793049 PMCID: PMC11122297 DOI: 10.3390/jpm14050467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 05/26/2024] Open
Abstract
The article discusses the importance of accurately distinguishing HER2-low from HER2-negative breast cancer, as novel ADCs have demonstrated activity in a large population of patients with HER2-low-expressing BC. While current guidelines recommend a dichotomous classification of HER2 as either positive or negative, the emergence of the HER2-low concept calls for standardization of HER2 testing in breast cancer, using currently available assays to better discriminate HER2 levels. This review covers the evolution and latest updates of the ASCO/CAP guidelines relevant to this important biomarker in breast cancer, including still-evolving concepts such as HER2 low, HER2 heterogeneity, and HER2 evolution. Our group presents the latest Mexican recommendations for HER2 status evaluation in breast cancer, considering the ASCO/CAP guidelines and introducing the HER2-low concept. In the era of personalized medicine, accurate HER2 status assessment remains one of the most important biomarkers in breast cancer, and the commitment of Mexican pathologists to theragnostic biomarker quality is crucial for providing the most efficient care in oncology.
Collapse
Affiliation(s)
| | - Jazmín de Anda-González
- Department of Pathology, Hospital de Oncología CMN Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | | | - Juan Pablo Flores-Gutiérrez
- Department of Pathology, University Hospital Dr. José Eleuterio González, Monterrey 64460, Mexico; (J.P.F.-G.); (G.S.G.-M.)
| | | | | | - Perla Zaragoza-Vargas
- Department of Pathology, Hospital de Gineco-Obstetricia, Instituto Mexicano del Seguro Social, Mexico City 64000, Mexico;
| | - Aldo Alcaraz-Wong
- Department of Pathology, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico;
| | - Ana Karen Soto-Sañudo
- Department of Pathology, Hospital Regional “Dr. Manuel Cárdenas de la Vega”, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Culiacán Rosales 80230, Mexico;
| | | | - Moisés Salamanca-García
- Department of Pathology, Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Mexico City 03229, Mexico;
| | - Georgina Loyola-Rodríguez
- Department of Pathology, Instituto de Seguridad y Servicios Sociales de los Trabajadores al Servicio de los Poderes del Estado de Puebla, Puebla 72550, Mexico;
| | - Gabriela Sofia Gómez-Macías
- Department of Pathology, University Hospital Dr. José Eleuterio González, Monterrey 64460, Mexico; (J.P.F.-G.); (G.S.G.-M.)
| | - Mario Murguia-Perez
- Department of Pathology, Centro Médico Nacional del Bajío, Instituto Mexicano del Seguro Social, León 37320, Mexico;
| | | | - Ricardo Villalobos-Valencia
- Department of Oncology, Hospital de Oncología CMN Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Enrique Talamantes
- Department of Oncology, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City 02990, Mexico;
| | - Claudia Arce-Salinas
- Department of Oncology, Instituto Nacional de Cancerología, Mexico City 14000, Mexico;
| |
Collapse
|
26
|
Huynh M, Vinck R, Gibert B, Gasser G. Strategies for the Nuclear Delivery of Metal Complexes to Cancer Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311437. [PMID: 38174785 DOI: 10.1002/adma.202311437] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/20/2023] [Indexed: 01/05/2024]
Abstract
The nucleus is an essential organelle for the function of cells. It holds most of the genetic material and plays a crucial role in the regulation of cell growth and proliferation. Since many antitumoral therapies target nucleic acids to induce cell death, tumor-specific nuclear drug delivery could potentiate therapeutic effects and prevent potential off-target side effects on healthy tissue. Due to their great structural variety, good biocompatibility, and unique physico-chemical properties, organometallic complexes and other metal-based compounds have sparked great interest as promising anticancer agents. In this review, strategies for specific nuclear delivery of metal complexes are summarized and discussed to highlight crucial parameters to consider for the design of new metal complexes as anticancer drug candidates. Moreover, the existing opportunities and challenges of tumor-specific, nucleus-targeting metal complexes are emphasized to outline some new perspectives and help in the design of new cancer treatments.
Collapse
Affiliation(s)
- Marie Huynh
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for Inorganic Chemistry, Paris, F-75005, France
- Gastroenterology and technologies for Health, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS5286, Université Lyon 1, Lyon, 69008, France
| | - Robin Vinck
- Orano, 125 avenue de Paris, Châtillon, 92320, France
| | - Benjamin Gibert
- Gastroenterology and technologies for Health, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS5286, Université Lyon 1, Lyon, 69008, France
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for Inorganic Chemistry, Paris, F-75005, France
| |
Collapse
|
27
|
Park M, Jung E, Park JM, Park S, Ko D, Seo J, Kim S, Nam KD, Kang YK, Farrand L, Hoang VH, Nguyen CT, La MT, Nam G, Park HJ, Ann J, Lee J, Kim YJ, Kim JY, Seo JH. The HSP90 inhibitor HVH-2930 exhibits potent efficacy against trastuzumab-resistant HER2-positive breast cancer. Theranostics 2024; 14:2442-2463. [PMID: 38646654 PMCID: PMC11024854 DOI: 10.7150/thno.93236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/21/2024] [Indexed: 04/23/2024] Open
Abstract
Rationale: Resistance to targeted therapies like trastuzumab remains a critical challenge for HER2-positive breast cancer patients. Despite the progress of several N-terminal HSP90 inhibitors in clinical trials, none have achieved approval for clinical use, primarily due to issues such as induction of the heat shock response (HSR), off-target effects, and unfavorable toxicity profiles. We sought to examine the effects of HVH-2930, a novel C-terminal HSP90 inhibitor, in overcoming trastuzumab resistance. Methods: The effect of HVH-2930 on trastuzumab-sensitive and -resistant cell lines in vitro was evaluated in terms of cell viability, expression of HSP90 client proteins, and impact on cancer stem cells. An in vivo model with trastuzumab-resistant JIMT-1 cells was used to examine the efficacy and toxicity of HVH-2930. Results: HVH-2930 was rationally designed to fit into the ATP-binding pocket interface cavity of the hHSP90 homodimer in the C-terminal domain of HSP90, stabilizing its open conformation and hindering ATP binding. HVH-2930 induces apoptosis without inducing the HSR but by specifically suppressing the HER2 signaling pathway. This occurs with the downregulation of HER2/p95HER2 and disruption of HER2 family member heterodimerization. Attenuation of cancer stem cell (CSC)-like properties was associated with the downregulation of stemness factors such as ALDH1, CD44, Nanog and Oct4. Furthermore, HVH-2930 administration inhibited angiogenesis and tumor growth in trastuzumab-resistant xenograft mice. A synergistic effect was observed when combining HVH-2930 and paclitaxel in JIMT-1 xenografts. Conclusion: Our findings highlight the potent efficacy of HVH-2930 in overcoming trastuzumab resistance in HER2-positive breast cancer. Further investigation is warranted to fully establish its therapeutic potential.
Collapse
Affiliation(s)
- Minsu Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Eunsun Jung
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul 08308, Republic of Korea
| | - Jung Min Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Soeun Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Dongmi Ko
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Juyeon Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Seongjae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Kee Dal Nam
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul 08308, Republic of Korea
| | - Yong Koo Kang
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul 08308, Republic of Korea
| | - Lee Farrand
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, South Australia 5000, Australia
| | - Van-Hai Hoang
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam
| | - Cong-Truong Nguyen
- Department of Organic Chemistry, Hanoi University of Pharmacy, Hanoi 10000, Vietnam
| | - Minh Thanh La
- Laboratory of Medicinal Chemistry, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Gibeom Nam
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Hyun-Ju Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Jihyae Ann
- Laboratory of Medicinal Chemistry, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeewoo Lee
- Laboratory of Medicinal Chemistry, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoon-Jae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul 08308, Republic of Korea
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul 08308, Republic of Korea
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul 08308, Republic of Korea
| |
Collapse
|
28
|
Ibrahim NSM, Kadry HH, Zaher AF, Mohamed KO. Synthesis of novel pyrimido[4,5-b]quinoline derivatives as dual EGFR/HER2 inhibitors as anticancer agents. Arch Pharm (Weinheim) 2024; 357:e2300513. [PMID: 38148301 DOI: 10.1002/ardp.202300513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023]
Abstract
A series of novel N-aryl-5-aryl-6,7,8,9-tetrahydropyrimido[4,5-b]quinolin-4-amines 4a-4l was synthesized as potential anticancer agents through Dimroth rearrangement reaction of intermediates 3a-3c. Pyrimido[4,5-b]quinolines 4a-4l showed promising activity against the Michigan Cancer Foundation-7 (MCF-7) cell line, compared with lapatinib as the reference drug. Compounds 4d, 4h, 4i, and 4l demonstrated higher cytotoxic activity than lapatinib, with IC50 values of 2.67, 6.82, 4.31, and 1.62 µM, respectively. Compounds 4d, 4i, and 4l showed promising epidermal growth factor receptor (EGFR) inhibition with IC50 values of 0.065, 0.116, and 0.052 µM, respectively. These compounds were subjected to human epidermal growth factor receptor 2 (HER2) inhibition and showed IC50 values of 0.09, 0.164, and 0.055 µM, respectively. Compounds 4d, 4i, and 4l are good candidates as dual EGFR/HER2 inhibitors. The most active compound, 4l, was subjected to cell-cycle analysis and induced cell-cycle arrest at the S phase. Compound 4l induced apoptosis 60-fold compared with control untreated MCF-7 cells. 4l can inhibit cancer metastasis. It reduced MCF-7 cell infiltration and metastasis by 45% compared with control untreated cells.
Collapse
Affiliation(s)
- Nahla Said M Ibrahim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hanan H Kadry
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ashraf F Zaher
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Khaled O Mohamed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University-Arish Branch, Arish, Egypt
| |
Collapse
|
29
|
Barzi A, Weipert CM, Espenschied CR, Raymond VM, Wang-Gillam A, Nezami MA, Gordon EJ, Mahadevan D, Mody K. ERBB2 (HER2) amplifications and co-occurring KRAS alterations in the circulating cell-free DNA of pancreatic ductal adenocarcinoma patients and response to HER2 inhibition. Front Oncol 2024; 14:1339302. [PMID: 38406801 PMCID: PMC10885695 DOI: 10.3389/fonc.2024.1339302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/16/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose Despite accumulating data regarding the genomic landscape of pancreatic ductal adenocarcinoma (PDAC), olaparib is the only biomarker-driven FDA-approved targeted therapy with a PDAC-specific approval. Treating ERBB2(HER2)-amplified PDAC with anti-HER2 therapy has been reported with mixed results. Most pancreatic adenocarcinomas have KRAS alterations, which have been shown to be a marker of resistance to HER2-targeted therapies in other malignancies, though the impact of these alterations in pancreatic cancer is unknown. We describe two cases of ERBB2-amplified pancreatic cancer patients treated with anti-HER2 therapy and provide data on the frequency of ERBB2 amplifications and KRAS alterations identified by clinical circulating cell-free DNA testing. Methods De-identified molecular test results for all patients with pancreatic cancer who received clinical cell-free circulating DNA analysis (Guardant360) between 06/2014 and 01/2018 were analyzed. Cell-free circulating DNA analysis included next-generation sequencing of up to 73 genes, including select small insertion/deletions, copy number amplifications, and fusions. Results Of 1,791 patients with pancreatic adenocarcinoma, 36 (2.0%) had an ERBB2 amplification, 26 (72.2%) of whom had a KRAS alteration. Treatment data were available for seven patients. Two were treated with anti-HER2 therapy after their cell-free circulating DNA result, with both benefiting from therapy, including one with a durable response to trastuzumab and no KRAS alteration detected until progression. Conclusion Our case series illustrates that certain patients with ERBB2-amplified pancreatic adenocarcinoma may respond to anti-HER2 therapy and gain several months of prolonged survival. Our data suggests KRAS mutations as a possible mechanism of primary and acquired resistance to anti-HER2 therapy in pancreatic cancer. Additional studies are needed to clarify the role of KRAS in resistance to anti-HER2 therapy.
Collapse
Affiliation(s)
- Afsaneh Barzi
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, United States
| | | | | | | | - Andrea Wang-Gillam
- Division of Oncology, Siteman Cancer Center, St. Louis, MO, United States
| | | | - Eva J. Gordon
- Private Health Management, Inc., Los Angeles, CA, United States
| | - Daruka Mahadevan
- Division of Hematology and Oncology, Department of Medicine, University of Texas Health, San Antonio, San Antonio, TX, United States
| | - Kabir Mody
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
30
|
Di Cosimo S, Pizzamiglio S, Ciniselli CM, Duroni V, Cappelletti V, De Cecco L, De Marco C, Silvestri M, De Santis MC, Vingiani A, Paolini B, Orlandi R, Iorio MV, Pruneri G, Verderio P. A gene expression-based classifier for HER2-low breast cancer. Sci Rep 2024; 14:2628. [PMID: 38297001 PMCID: PMC10830477 DOI: 10.1038/s41598-024-52148-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/15/2024] [Indexed: 02/02/2024] Open
Abstract
In clinical trials evaluating antibody-conjugated drugs (ADCs), HER2-low breast cancer is defined through protein immunohistochemistry scoring (IHC) 1+ or 2+ without gene amplification. However, in daily practice, the accuracy of IHC is compromised by inter-observer variability. Herein, we aimed to identify HER2-low breast cancer primary tumors by leveraging gene expression profiling. A discovery approach was applied to gene expression profile of institutional INT1 (n = 125) and INT2 (n = 84) datasets. We identified differentially expressed genes (DEGs) in each specific HER2 IHC category 0, 1+, 2+ and 3+. Principal Component Analysis was used to generate a HER2-low signature whose performance was evaluated in the independent INT3 (n = 95), and in the publicly available TCGA and GSE81538 datasets. The association between the HER2-low signature and HER2 IHC categories was evaluated by Kruskal-Wallis test with post hoc pair-wise comparisons. The HER2-low signature discriminatory capability was assessed by estimating the area under the receiver operating characteristic curve (AUC). Gene Ontology and KEGG analyses were performed to evaluate the HER2-low signature genes functional enrichment. A HER2-low signature was computed based on HER2 IHC category-specific DEGs. The twenty genes included in the signature were significantly enriched with lipid and steroid metabolism pathways, peptidase regulation, and humoral immune response. The HER2-low signature values showed a bell-shaped distribution across IHC categories (low values in 0 and 3+; high values in 1+ and 2+), effectively distinguishing HER2-low from 0 (p < 0.001) to 3+ (p < 0.001). Notably, the signature values were higher in tumors scored with 1+ as compared to 0. The HER2-low signature association with IHC categories and its bell-shaped distribution was confirmed in the independent INT3, TCGA and GSE81538 datasets. In the combined INT1 and INT3 datasets, the HER2-low signature achieved an AUC value of 0.74 (95% confidence interval, CI 0.67-0.81) in distinguishing HER2-low vs. the other categories, outperforming the individual ERBB2 mRNA AUC value of 0.52 (95% CI 0.43-0.60). These results represent a proof-of-concept for an observer-independent gene-expression-based classifier of HER2-low status. The herein identified 20-gene signature shows promise in distinguishing between HER2 0 and HER2-low expressing tumors, including those scored as 1+ at IHC, and in developing a selection approach for ADCs candidates.
Collapse
Affiliation(s)
- Serena Di Cosimo
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Sara Pizzamiglio
- Bioinformatics and Biostatistics Unit, Department of Epidemiology and Data Science, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy.
| | - Chiara Maura Ciniselli
- Bioinformatics and Biostatistics Unit, Department of Epidemiology and Data Science, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Valeria Duroni
- Bioinformatics and Biostatistics Unit, Department of Epidemiology and Data Science, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Vera Cappelletti
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Loris De Cecco
- Molecular Mechanisms Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Cinzia De Marco
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Marco Silvestri
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Maria Carmen De Santis
- Radiation Oncology 1, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
- Breast Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Andrea Vingiani
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
- Breast Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Biagio Paolini
- Department of Pathology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Rosaria Orlandi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Marilena Valeria Iorio
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Giancarlo Pruneri
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
- Breast Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Paolo Verderio
- Bioinformatics and Biostatistics Unit, Department of Epidemiology and Data Science, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
31
|
Rubin E, Shan KS, Dalal S, Vu DUD, Milillo-Naraine AM, Guaqueta D, Ergle A. Molecular Targeting of the Human Epidermal Growth Factor Receptor-2 (HER2) Genes across Various Cancers. Int J Mol Sci 2024; 25:1064. [PMID: 38256137 PMCID: PMC10816365 DOI: 10.3390/ijms25021064] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) belongs to the ErbB family, a group of four transmembrane glycoproteins with tyrosine kinase activity, all structurally related to epidermal growth factor receptor (EGFR). These tyrosine kinases are involved in the transmission of cellular signals controlling normal cell growth and differentiation. If this transmission goes awry, it can lead to dysregulated growth of the cell. HER2 specifically can be implicated in the pathogenesis of at least eight malignancies. HER2 positivity quickly became a well-characterized indicator of aggressiveness and poor prognosis, with high rates of disease progression and mortality. After realizing the implication of HER2, it first became investigated as a target for treatment in breast cancer, and later expanded to areas of research in other cancer types. To this day, the most therapeutic advancements of anti-HER2 therapy have been in breast cancer; however, there have been strong advancements made in the incorporation of anti-HER2 therapy in other cancer types as well. This comprehensive review dissects HER2 to its core, incorporating the most up to date information. The topics touched upon are discussed in detail and up to 200 published sources from the most highly recognized journals have been integrated. The importance of knowing about HER2 is exemplified by the groundbreaking advancements that have been made, and the change in treatment plans it has brought to the oncological world in the last twenty years. Since its groundbreaking discovery there have been significant breakthroughs in knowledge regarding the actual receptor, the receptors biology, its mechanism of action, and advancements in tests to detect HER2 and significant strides on how to best incorporate targeted treatment. Due to the success of this field thus far, the review concludes by discussing the future of novel anti-HER2 therapy currently in development that everyone should be aware of.
Collapse
Affiliation(s)
- Elizabeth Rubin
- Memorial Cancer Institute, Pembroke Pines, FL 33028, USA; (K.S.S.); (S.D.); (D.U.D.V.); (A.M.M.-N.); (D.G.); (A.E.)
| | | | | | | | | | | | | |
Collapse
|
32
|
Yang T, Li W, Huang T, Zhou J. Genetic Testing Enhances the Precision Diagnosis and Treatment of Breast Cancer. Int J Mol Sci 2023; 24:16607. [PMID: 38068930 PMCID: PMC10706486 DOI: 10.3390/ijms242316607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
The contemporary comprehension of breast cancer has progressed to the molecular level. As a heterogeneous malignancy, conventional pathological diagnosis and histological classification could no longer meet the needs of precisely managing breast cancer. Genetic testing based on gene expression profiles and gene mutations has emerged and substantially contributed to the precise diagnosis and treatment of breast cancer. Multigene assays (MGAs) are explored for early-stage breast cancer patients, aiding the selection of adjuvant therapy and predicting prognosis. For metastatic breast cancer patients, testing specific genes indicates potentially effective antitumor agents. In this review, genetic testing in early-stage and metastatic breast cancer is summarized, as well as the advantages and challenges of genetic testing in breast cancer.
Collapse
Affiliation(s)
| | | | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China (W.L.)
| | - Jun Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China (W.L.)
| |
Collapse
|
33
|
Zou SP, Yang HY, Ouyang ML, Cheng Q, Shi X, Sun MH. A disproportionality analysis of adverse events associated to pertuzumab in the FDA Adverse Event Reporting System (FAERS). BMC Pharmacol Toxicol 2023; 24:62. [PMID: 37957717 PMCID: PMC10642055 DOI: 10.1186/s40360-023-00702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Pertuzumab is widely used for the treatment of HER2 + breast cancer. But its safety in the real world should be continuously monitored. So, we evaluated the safety of pertuzumab by pharmacovigilance analyze based on related adverse events (AEs) from the FDA Adverse Event Reporting System (FAERS) and find whether potential or uncertain adverse events were present. METHODS In disproportionality analysis, four algorithms were employed to detect the signals of pertuzumab from the FAERS between 2012 and 2022. In addition, we also used MYSQL 8.0, Navicat Premium 15, and Microsoft EXCEL 2019 to analyze the potential and high-ROR (reporting odds ratio) signals of pertuzumab. We also collected the onset times of pertuzumab-associated AEs. RESULTS From January 2012 to December 2022, there are 39,190,598 AEs reported from the FAERS database, of which 14,707 AEs listed pertuzumab as the 'primary suspected (PS)' drug. A total of 115 (46 potential) significant disproportionality preferred terms (PTs) conforming to the four algorithms were retained. Finally, we detected that the pertuzumab-induced AEs occurred in 12 organ systems. For pertuzumab, unexpected and significant PTs of AEs were found, including but not limited to below PTs: haematotoxicity, cardiotoxicity, cardiomyopathy, mitral valve incompetence, tachycardia, intestinal perforation, hemorrhoids, erysipelas, dehydration, pneumonitis, skin toxicity, onychomadesis, cyanosis, and circulatory collapse. We found there were 9 strong signals (5 potential safety signals) and 68 medium intensity signals (21 potential safety signals) according to IC025 (information component). The potential strong signals (IC025 > 3.0) were myelosuppression, cardiotoxicity, cardiac dysfunction, ejection fraction decreased, interstitial lung disease, and onychomadesis. Excluding unreported or unreasonable onset time reports, a total of 2016 AEs reported onset time and the median onset time was 117 days (4, 96), as median (Q1, Q3). Notably, most of the all AEs (n = 1133, 56%) and cardiac-related events (n = 405, 53%) all occurred within one month after pertuzumab therapy. CONCLUSION Analysis of FAERS data identified pertuzumab-associated AEs, and our findings supported continuous clinical monitoring, pharmacovigilance, and further studies of pertuzumab. A significant association was detected between pertuzumab and some potential adverse events which should be regarded with some care. We have to pay attention to the first month after pertuzumab therapy and prepare emergency measures, especially for the elderly and patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Shu-Peng Zou
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei Province, 430000, China
| | - Hai-Yun Yang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu Province, 730000, China
| | - Meng-Ling Ouyang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei Province, 430000, China
| | - Qian Cheng
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei Province, 430000, China
| | - Xuan Shi
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei Province, 430000, China
| | - Ming-Hui Sun
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei Province, 430000, China.
| |
Collapse
|
34
|
Song CH, Jeong M, In H, Kim JH, Lin CW, Han KH. Trends in the Development of Antibody-Drug Conjugates for Cancer Therapy. Antibodies (Basel) 2023; 12:72. [PMID: 37987250 PMCID: PMC10660735 DOI: 10.3390/antib12040072] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023] Open
Abstract
In cancer treatment, the first-generation, cytotoxic drugs, though effective against cancer cells, also harmed healthy ones. The second-generation targeted cancer cells precisely to inhibit their growth. Enter the third-generation, consisting of immuno-oncology drugs, designed to combat drug resistance and bolster the immune system's defenses. These advanced therapies operate by obstructing the uncontrolled growth and spread of cancer cells through the body, ultimately eliminating them effectively. Within the arsenal of cancer treatment, monoclonal antibodies offer several advantages, including inducing cancer cell apoptosis, precise targeting, prolonged presence in the body, and minimal side effects. A recent development in cancer therapy is Antibody-Drug Conjugates (ADCs), initially developed in the mid-20th century. The second generation of ADCs addressed this issue through innovative antibody modification techniques, such as DAR regulation, amino acid substitutions, incorporation of non-natural amino acids, and enzymatic drug attachment. Currently, a third generation of ADCs is in development. This study presents an overview of 12 available ADCs, reviews 71 recent research papers, and analyzes 128 clinical trial reports. The overarching objective is to gain insights into the prevailing trends in ADC research and development, with a particular focus on emerging frontiers like potential targets, linkers, and drug payloads within the realm of cancer treatment.
Collapse
Affiliation(s)
- Chi Hun Song
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Minchan Jeong
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Hyukmin In
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Ji Hoe Kim
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Chih-Wei Lin
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 406, Taiwan;
| | - Kyung Ho Han
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| |
Collapse
|
35
|
Seban RD. Reply to: HER2-Low Breast Cancer Can Be Visualized by HER2 PET. J Nucl Med 2023; 64:1841-1842. [PMID: 37709533 DOI: 10.2967/jnumed.123.266490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 09/16/2023] Open
|
36
|
Ibrahim NSM, Kadry HH, Zaher AF, Mohamed KO. Synthesis of novel pyrimido[4,5-b]quinolines as potential anticancer agents and HER2 inhibitors. Chem Biol Drug Des 2023; 102:996-1013. [PMID: 37527951 DOI: 10.1111/cbdd.14307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/27/2023] [Accepted: 07/14/2023] [Indexed: 08/03/2023]
Abstract
A series of N-arylpyrimido[4,5-b]quinolines 3a-e and 2-aryl-2,3-dihydropyrimido[4,5-b]quinoline-4(1H)-ones 5a-e was designed and synthesized as potential anticancer agents against breast cancer. Compounds 3e, 5a, 5b, 5d, and 5e showed promising activity against the MCF-7 cell line. Among them, compound 5b was the most active with IC50 of 1.67 μM. Compound 5b promoted apoptosis and induced cell cycle arrest at S phase. 5b increased the level of pro-apoptotic proteins p53, Bax, and caspase-7 and inhibited the anti-apoptotic protein Bcl-2. Furthermore, all the synthesized compounds were docked into the crystal structure of HER2 (PBD: 3 pp0). Compounds 3e, 5a, 5b, 5d, and 5e showed good energy scores and binding modes. Finally, Compound 5b was evaluated on the HER2 assay and revealed good inhibition with IC50 of 0.073 μM.
Collapse
Affiliation(s)
- Nahla Said M Ibrahim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hanan H Kadry
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ashraf F Zaher
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Khaled O Mohamed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
37
|
Zahedipour F, Jamialahmadi K, Zamani P, Reza Jaafari M. Improving the efficacy of peptide vaccines in cancer immunotherapy. Int Immunopharmacol 2023; 123:110721. [PMID: 37543011 DOI: 10.1016/j.intimp.2023.110721] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Peptide vaccines have shown great potential in cancer immunotherapy by targeting tumor antigens and activating the patient's immune system to mount a specific response against cancer cells. However, the efficacy of peptide vaccines in inducing a sustained immune response and achieving clinical benefit remains a major challenge. In this review, we discuss the current status of peptide vaccines in cancer immunotherapy and strategies to improve their efficacy. We summarize the recent advancements in the development of peptide vaccines in pre-clinical and clinical settings, including the use of novel adjuvants, neoantigens, nano-delivery systems, and combination therapies. We also highlight the importance of personalized cancer vaccines, which consider the unique genetic and immunological profiles of individual patients. We also discuss the strategies to enhance the immunogenicity of peptide vaccines such as multivalent peptides, conjugated peptides, fusion proteins, and self-assembled peptides. Although, peptide vaccines alone are weak immunogens, combining peptide vaccines with other immunotherapeutic approaches and developing novel approaches such as personalized vaccines can be promising methods to significantly enhance their efficacy and improve the clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
38
|
Riccardi F, Dal Bo M, Macor P, Toffoli G. A comprehensive overview on antibody-drug conjugates: from the conceptualization to cancer therapy. Front Pharmacol 2023; 14:1274088. [PMID: 37790810 PMCID: PMC10544916 DOI: 10.3389/fphar.2023.1274088] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023] Open
Abstract
Antibody-Drug Conjugates (ADCs) represent an innovative class of potent anti-cancer compounds that are widely used in the treatment of hematologic malignancies and solid tumors. Unlike conventional chemotherapeutic drug-based therapies, that are mainly associated with modest specificity and therapeutic benefit, the three key components that form an ADC (a monoclonal antibody bound to a cytotoxic drug via a chemical linker moiety) achieve remarkable improvement in terms of targeted killing of cancer cells and, while sparing healthy tissues, a reduction in systemic side effects caused by off-tumor toxicity. Based on their beneficial mechanism of action, 15 ADCs have been approved to date by the market approval by the Food and Drug Administration (FDA), the European Medicines Agency (EMA) and/or other international governmental agencies for use in clinical oncology, and hundreds are undergoing evaluation in the preclinical and clinical phases. Here, our aim is to provide a comprehensive overview of the key features revolving around ADC therapeutic strategy including their structural and targeting properties, mechanism of action, the role of the tumor microenvironment and review the approved ADCs in clinical oncology, providing discussion regarding their toxicity profile, clinical manifestations and use in novel combination therapies. Finally, we briefly review ADCs in other pathological contexts and provide key information regarding ADC manufacturing and analytical characterization.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| |
Collapse
|
39
|
Crosby EJ, Hartman ZC, Lyerly HK. Beyond Neoantigens: Antigens Derived from Tumor Drivers as Cancer Vaccine Targets. Clin Cancer Res 2023; 29:3256-3258. [PMID: 37428103 PMCID: PMC10472089 DOI: 10.1158/1078-0432.ccr-23-1244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/28/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023]
Abstract
A vaccine targeting HER2, a nonmutated but overexpressed tumor antigen, readily primed T cells for ex vivo expansion and adoptive transfer with minimal toxicity. This regimen led to intramolecular epitope spreading in a majority of patients and offers a treatment modality that may improve outcomes for patients with metastatic breast cancer expressing HER2. See related article by Disis et al., p. 3362.
Collapse
Affiliation(s)
- Erika J. Crosby
- Department of Surgery, Duke University, Durham, North Carolina
| | - Zachary C. Hartman
- Departments of Surgery, Integrative Immunobiology, and Pathology, Duke University, Durham, North Carolina
| | - H. Kim Lyerly
- Departments of Surgery, Integrative Immunobiology, and Pathology, Duke University, Durham, North Carolina
| |
Collapse
|
40
|
Konda P, Garinet S, Van Allen EM, Viswanathan SR. Genome-guided discovery of cancer therapeutic targets. Cell Rep 2023; 42:112978. [PMID: 37572322 DOI: 10.1016/j.celrep.2023.112978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/22/2023] [Accepted: 07/28/2023] [Indexed: 08/14/2023] Open
Abstract
The success of precision oncology-which aims to match the right therapies to the right patients based on molecular status-is predicated on a robust pipeline of molecular targets against which therapies can be developed. Recent advances in genomics and functional genetics have enabled the unbiased discovery of novel molecular targets at scale. We summarize the promise and challenges in integrating genomic and functional genetic landscapes of cancer to establish the next generation of cancer targets.
Collapse
Affiliation(s)
- Prathyusha Konda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Simon Garinet
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Srinivas R Viswanathan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Gogia P, Ashraf H, Bhasin S, Xu Y. Antibody-Drug Conjugates: A Review of Approved Drugs and Their Clinical Level of Evidence. Cancers (Basel) 2023; 15:3886. [PMID: 37568702 PMCID: PMC10417123 DOI: 10.3390/cancers15153886] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/17/2023] [Accepted: 07/13/2023] [Indexed: 08/13/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are an innovative family of agents assembled through linking cytotoxic drugs (payloads) covalently to monoclonal antibodies (mAbs) to be delivered to tumor tissue that express their particular antigen, with the theoretical advantage of an augmented therapeutic ratio. As of June 2023, eleven ADCs have been approved by the Food and Drug Administration (FDA) and are on the market. These drugs have been added to the therapeutic armamentarium of acute myeloblastic and lymphoblastic leukemias, various types of lymphoma, breast, gastric or gastroesophageal junction, lung, urothelial, cervical, and ovarian cancers. They have proven to deliver more potent and effective anti-tumor activities than standard practice in a wide variety of indications. In addition to targeting antigen-expressing tumor cells, bystander effects have been engineered to extend cytotoxic killing to low-antigen-expressing or negative tumor cells in the heterogenous tumor milieu. Inevitably, myelosuppression is a common side effect with most of the ADCs due to the effects of the cytotoxic payload. Also, other unique side effects are specific to the tissue antigen that is targeted for, such as the cardiac toxicity with Her-2 targeting ADCs, and the hemorrhagic side effects with the tissue factor (TF) targeting Tisotumab vedotin. Further exciting developments are centered in the strategies to improve the tolerability and efficacy of the ADCs to improve the therapeutic window; as well as the development of novel payloads including (1) peptide-drug conjugates (PDCs), with the peptide replacing the monoclonal antibody, rendering greater tumor penetration; (2) immune-stimulating antibody conjugates (ISACs), which upon conjugation of the antigen, cause an influx of pro-inflammatory cytokines to activate dendritic cells and harness an anti-tumor T-cell response; and (3) the use of radioactive isotopes as a payload to enhance cytotoxic activity.
Collapse
Affiliation(s)
- Pooja Gogia
- Department of Hematology/Oncology, Maimonides Medical Center, Brooklyn, NY 11219, USA;
| | - Hamza Ashraf
- Department of Internal Medicine, Overlook Medical Center, Summit, NJ 07901, USA;
| | - Sidharth Bhasin
- Department of Pulmonary Medicine, Saint Peter’s University Hospital, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA;
| | - Yiqing Xu
- Department of Hematology/Oncology, Maimonides Medical Center, Brooklyn, NY 11219, USA;
| |
Collapse
|
42
|
Shi D, Li Y, Liang X, Chen L. Cost-effectiveness of sacituzumab govitecan in hormone receptor-positive/human epidermal growth factor receptor 2-negative metastatic breast cancer. Front Oncol 2023; 13:1162360. [PMID: 37251935 PMCID: PMC10213649 DOI: 10.3389/fonc.2023.1162360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Background The efficiency and safety of sacituzumab govitecan (SG) for the therapy of hormone receptor-positive (HR+)/human epidermal receptor 2-negative (HER2-) metastatic breast cancer (BC) has been demonstrated. The aim of this study is to evaluate its cost-effectiveness on HR+/HER2- metastatic BC from the third-party payer perspective in the United States. Methods We performed the cost-effectiveness of SG and chemotherapy using a partitioned survival model. TROPiCS-02 provided clinical patients for this study. We evaluated the robustness of this study by one-way and probabilistic sensitivity analyses. Subgroup analyses were also conducted. The outcomes were costs, life-years, quality-adjusted life years (QALYs), incremental cost-effectiveness ratio (ICER), incremental net health benefit (INHB), and incremental net monetary benefit (INMB). Results SG treatment was related to an increase of 0.284 life years and 0.217 QALYs over chemotherapy, as well as a cost increase of $132,689, reaching an ICER of $612,772/QALY. The INHB was -0.668 QALYs, and the INMB was -$100,208. SG was not cost-effective at the willingness to pay (WTP) threshold of $150,000/QALY. The outcomes were sensitive to patient body weight and cost of SG. SG may be cost-effective at the WTP threshold of $150,000/QALY if the price is less than $3.997/mg or the weight of patients is under 19.88 kg. Based on the subgroup analysis, SG did not prove cost-effective in all subgroups at the WTP threshold of $150,000/QALY. Conclusion From a third-party payer standpoint in the United States, SG was not cost-effective, even though it had a clinically significant advantage over chemotherapy for the treatment of HR+/HER2- metastatic BC. The cost-effectiveness of SG can be improved if the price is substantially reduced.
Collapse
Affiliation(s)
- Demin Shi
- Department of Reproductive Medicine, The People’s Hospital of Hechi, Hechi, Guangxi, China
| | - Yan Li
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Xueyan Liang
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Lingyuan Chen
- Department of Pharmacy, The People’s Hospital of Hechi, Hechi, Guangxi, China
| |
Collapse
|
43
|
Grinda T, Rassy E, Pistilli B. Antibody-Drug Conjugate Revolution in Breast Cancer: The Road Ahead. Curr Treat Options Oncol 2023; 24:442-465. [PMID: 36966267 PMCID: PMC10122624 DOI: 10.1007/s11864-023-01072-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2023] [Indexed: 03/27/2023]
Abstract
OPINION STATEMENT Antibody drug-conjugates (ADCs) have revolutionized the treatment of many types of cancer, including breast cancer. Recently, two new ADCs have been approved, trastuzumab deruxtecan and sacituzumab govitecan; both have demonstrated impressive improvements in overall survival, trastuzumab deruxtecan in all three subtypes of metastatic breast cancer and sacituzumab govitecan in luminal and triple negative metastatic breast cancer. These drugs are the results of significant progress and innovation in the construction of the three components of an ADC, the monoclonal antibody, the payload, and the linker, and of the discovery of new target antigens. ADC engineering has profoundly changed the paradigm of cancer treatment, on one side being effective on tumors considered inherently resistant to the payload class of drugs and on the other side demonstrating activity in tumors with very low target expression. Yet, it is likely that we are just at the beginning of a new era as the identification of new targets and the introduction of new ADC constructs and combinations will expand the field of ADC rapidly over the coming years.
Collapse
Affiliation(s)
- Thomas Grinda
- Department of Cancer Medicine, Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| | - Barbara Pistilli
- Department of Cancer Medicine, Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France.
| |
Collapse
|