1
|
Aoyama T, Ooki A, Oba K, Nishikawa K, Kawabata R, Honda M, Maeda H, Kanda M, Sugiyama K, Makiyama A, Segami K, Takahashi M, Shindo Y, Namikawa T, Oshima T, Katayama A, Shiosakai K, Sakamoto J. A multicenter randomized open-label phase 2 study investigating optimal antiemetic therapy for patients with advanced/recurrent gastric cancer treated with trastuzumab deruxtecan: the EN-hance study. Int J Clin Oncol 2025; 30:1162-1173. [PMID: 40293658 PMCID: PMC12122603 DOI: 10.1007/s10147-025-02748-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/14/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Trastuzumab deruxtecan (T-DXd) has been approved for the treatment of human epidermal growth factor receptor-2 (HER2)-positive gastric cancer and other indications in several countries and is considered moderately or highly emetogenic. The management of nausea and vomiting associated with T-DXd treatment has not been fully evaluated and the effectiveness of conventional prophylaxis remains unknown. METHODS This open-label, randomized, multicenter, phase 2 study aimed to investigate the optimal antiemetic therapy for Japanese patients with gastric cancer undergoing T-DXd treatment. Patients were randomized to a doublet regimen group (dexamethasone and palonosetron) or triplet regimen group (aprepitant, dexamethasone, and palonosetron) at a ratio of one to one, stratified by sex, gastrectomy status, and study institution. Both antiemetic treatments were administered from day 1 before T-DXd administration, and emetic events and nausea were observed for 21 days. The primary endpoint was the antiemetic complete response (CR) rate to assess control for emetic events based on voluntary patient-reported outcomes (PROs) during cycle 1 (1-21 days). RESULTS Of the 60 enrolled patients, 58 were eligible for inclusion in this analysis (29 patients in each regimen group). The overall CR rates for the doublet and triplet regimens were 41.4% (12/29 patients) and 37.9% (11/29 patients), respectively, and neither regimen met the pre-specified threshold (> 18/29 patients). The CR rate in the acute phase (0-24 h) was 86.2% (25/29 patients) for both regimens, and the CR rates in the delayed phase (2-21 days) were 41.4% (12/29 patients) and 37.9% (11/29 patients) for the doublet and triplet regimens, respectively. CONCLUSIONS Given that the primary endpoint was not met, further research is needed to better characterize nausea and vomiting with T-DXd to tailor an anti-emetic regimen that suits the needs of the patients.
Collapse
Affiliation(s)
- Toru Aoyama
- Department of Surgery, Yokohama City University Hospital, Yokohama, Japan.
- Department of Gastric Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan.
| | - Akira Ooki
- The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto, Japan
| | - Koji Oba
- The University of Tokyo Graduate School of Medicine, Bunkyō, Japan
| | | | | | | | | | - Mitsuro Kanda
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | - Kenki Segami
- Saiseikai Yokohama-Shi Nanbu Hospital, Yokohama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Haque M, Atallah N, Patke R, Harris AE, Woodcock CL, Varun D, Thompson RL, Jackson-Oxley J, Okui CH, Dean A, Alsaleem M, Rakha E, Irshad S, Davis MB, Jeyapalan JN, Mongan NP, Rutland CS. Cardiotoxicity of breast cancer drug treatments. Transl Oncol 2025; 55:102352. [PMID: 40090070 PMCID: PMC11952852 DOI: 10.1016/j.tranon.2025.102352] [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: 01/14/2025] [Accepted: 03/04/2025] [Indexed: 03/18/2025] Open
Abstract
Breast cancer (BC) is a leading cause of cancer-related mortality among women worldwide. BC is regarded as a systemic disease. Consequently, the majority of BC patients undergo systemic therapy to reduce the risk of distant metastasis (DM) and associated mortality. Although systemic therapies improve patients' outcome, they are associated with significant side effects, particularly cardiotoxicity and cardiovascular complications. Such side effects vary significantly in severity and duration. Cardiotoxicity may remain undetected for years post-treatment until the symptoms of heart failure (HF) present in the patient, which can reduce life expectancy, quality of life, and even lead to death. Therefore, it is vital to investigate the cardiotoxic and cardiovascular risks of BC treatment regimens to identify patients who would benefit most from cardiac monitoring. As it stands, cardiotoxicity is reported from individual studies or for selected drug therapies only. This review fulfils a gap in the literature by combining clinical studies of cardiotoxicity together from clinical trial data published on clinicaltrials.gov.uk.
Collapse
Affiliation(s)
- Maria Haque
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Nehal Atallah
- Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Rodhan Patke
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Anna E Harris
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Corinne L Woodcock
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Dhruvika Varun
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Rachel L Thompson
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Jorja Jackson-Oxley
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Cyntholia H Okui
- Department of Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Alexander Dean
- Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Mansour Alsaleem
- Unit of Scientific Research, Applied College, Qassim University, Saudi Arabia
| | - Emad Rakha
- Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK; Pathology Department, Hamad Medical Corporation, Doha, Qatar
| | - Sheeba Irshad
- Cancer & Pharmaceutical Sciences, King's College London & Guys & St Thomas NHS Trust, London, UK
| | - Melissa B Davis
- Department of Surgery, Weill Cornell Medical College, New York, NY, USA; Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA; Institute of Translational Genomic Medicine, Morehouse School of Medicine, Atlanta, GA, USA
| | - Jennie N Jeyapalan
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK
| | - Nigel P Mongan
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK; Department of Pharmacology, Weill Cornell Medicine, New York, New York, USA.
| | - Catrin S Rutland
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences University of Nottingham, UK; Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, UK.
| |
Collapse
|
3
|
Sundar R, Nakayama I, Markar SR, Shitara K, van Laarhoven HWM, Janjigian YY, Smyth EC. Gastric cancer. Lancet 2025:S0140-6736(25)00052-2. [PMID: 40319897 DOI: 10.1016/s0140-6736(25)00052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/13/2024] [Accepted: 01/09/2025] [Indexed: 05/07/2025]
Abstract
Gastric cancer remains a major health challenge worldwide, with nearly 1 million new cases annually contributing to more than 650 000 deaths. Epidemiologically, gastric cancer shows substantial geographical variation in incidence, with higher rates in Asia, South America, and eastern Europe, and a rapid increase in early-onset cases among people younger than 50 years. Key risk factors for gastric cancer include Helicobacter pylori infection, diet, obesity, smoking, and genetic predisposition. Early detection through comprehensive diagnostic procedures is crucial for optimising treatment outcomes. Standard treatment approaches for locally advanced gastric cancer include surgical resection, particularly D2 lymphadenectomy, complemented by chemotherapy and radiotherapy. There is increasing implementation of minimally invasive surgical techniques for operable disease and integration of immune checkpoint inhibitors and targeted therapies for advanced stages. Emerging therapies, such as novel targeted treatments and next-generation immunotherapies, show promise in improving survival and quality of life. Future directions in the management of gastric cancer focus on precision medicine, continued advancement in immunotherapy, novel early detection methods, and a multidisciplinary approach to care. These strategies aim to enhance the overall effectiveness of treatment and prognosis worldwide.
Collapse
Affiliation(s)
- Raghav Sundar
- Department of Medicine, Section of Medical Oncology, Yale School of Medicine, New Haven, CT, USA; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Izuma Nakayama
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Sheraz R Markar
- Surgical Intervention Trials Unit, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands; Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Elizabeth C Smyth
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
4
|
Tsao LC, Wang JS, Ma X, Sodhi S, Ragusa JV, Liu B, McBane J, Wang T, Wei J, Liu CX, Yang X, Lei G, Spasojevic I, Fan P, Trotter TN, Morse M, Lyerly HK, Hartman ZC. Effective extracellular payload release and immunomodulatory interactions govern the therapeutic effect of trastuzumab deruxtecan (T-DXd). Nat Commun 2025; 16:3167. [PMID: 40175391 PMCID: PMC11965298 DOI: 10.1038/s41467-025-58266-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/15/2025] [Indexed: 04/04/2025] Open
Abstract
Trastuzumab deruxtecan (T-DXd) is an antibody-drug conjugate (ADC) targeting HER2, exhibiting significant clinical efficacy in breast cancer (BC) with varying HER2 expression, including HER2-low and HER2-ultralow. However, the precise mechanism underlying its efficacy and the contribution of immune activation in these settings remain unclear. Here, we demonstrate that T-DXd efficacy in HER2-low and HER2-negative BC is independent of HER2 engagement and ADC internalization. Instead, its activity relies on extracellular proteases, such as cathepsin L (CTSL), within the tumor microenvironment. Irrespective of their HER2 status, tumor and stromal compartments of invasive BC abundantly express CTSL, which efficiently cleaves the specialized linker of T-DXd, facilitating payload release and inducing cytotoxicity against HER2-low/negative tumors. In HER2-positive BC, the antibody backbone of T-DXd engages Fcγ-receptors and drives antibody-dependent cellular phagocytosis (ADCP). Concurrently, its cytotoxic payload (DXd) induces immunogenic cell death, further activating myeloid cells via TLR4 and STING pathways to enhance tumor antigen presentation to CD8+ T cells. Notably, T-DXd cytotoxicity also upregulates tumor CD47 expression, dampening immune activation. Combining T-DXd with CD47 checkpoint blockade significantly enhances anti-tumor immune responses in a HER2-transgenic BC mouse model, while also inducing durable CD8+ T cell memory to prevent tumor recurrence after therapy cessation.
Collapse
Affiliation(s)
- Li-Chung Tsao
- Department of Surgery, Duke University, Durham, NC, USA
| | - John S Wang
- Department of Medicine, Duke University, Durham, NC, USA
| | - Xingru Ma
- Department of Pathology, Duke University, Durham, NC, USA
| | - Sirajbir Sodhi
- Department of Medicine, Duke University, Durham, NC, USA
| | - Joey V Ragusa
- Department of Pathology, Duke University, Durham, NC, USA
| | - Bushangqing Liu
- Program in Cell and Molecular Biology, Duke University, Durham, NC, USA
| | - Jason McBane
- Department of Surgery, Duke University, Durham, NC, USA
| | - Tao Wang
- Department of Surgery, Duke University, Durham, NC, USA
| | - Junping Wei
- Department of Surgery, Duke University, Durham, NC, USA
| | - Cong-Xiao Liu
- Department of Surgery, Duke University, Durham, NC, USA
| | - Xiao Yang
- Department of Surgery, Duke University, Durham, NC, USA
| | - Gangjun Lei
- Department of Surgery, Duke University, Durham, NC, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University, Durham, NC, USA
- PK/PD Core Laboratory, Duke Cancer Institute, Durham, NC, USA
| | - Ping Fan
- PK/PD Core Laboratory, Duke Cancer Institute, Durham, NC, USA
| | | | - Michael Morse
- Department of Surgery, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA
| | - Zachary C Hartman
- Department of Surgery, Duke University, Durham, NC, USA.
- Department of Pathology, Duke University, Durham, NC, USA.
- Program in Cell and Molecular Biology, Duke University, Durham, NC, USA.
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
5
|
Liao D, Zhang J, Yan T, Chen Y, Fu Y, Xie N, Long M. A Systematic Review of Mechanisms, Incidence, and Management of Trastuzumab Deruxtecan Induced ILD/Pneumonitis in Solid Tumors. Drug Des Devel Ther 2025; 19:1655-1668. [PMID: 40083848 PMCID: PMC11904318 DOI: 10.2147/dddt.s508773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Trastuzumab deruxtecan (T-DXd) has been approved to treat various tumors. While most adverse events (AEs) associated with T-DXd are manageable, interstitial lung disease (ILD)/pneumonitis is a notable AE of special concern. This review describes the incidence, severity, and management of T-DXd-induced ILD/pneumonitis across different tumors. We conducted a systematic search of PubMed, Embase, Cochrane Library, and Web of Science for literature published up to 13 September 2024, regarding the use of T-DXd in the treatment of HER2-positive tumors. Studies included were clinical trials involving HER2-positive tumors with reported ILD/pneumonitis cases.The main data extracted from the full-text articles included the incidence and severity of T-DXd-induced ILD. 18 studies involving 3380 patients with various advanced solid malignancies were included in our review. The overall incidence of adjudicated drug-related ILD/pneumonitis was 12.40%. Although most ILD/pneumonitis cases were low-grade, the risk of ILD/pneumonitis-related death should not be overlooked. Given the prolonged exposure to the drug, careful monitoring and management of T-DXd-induced ILD/pneumonitis are critical. Management strategies include dose reduction, treatment interruption, discontinuation, corticosteroids, and supportive care. Further research is needed to clarify the risk factors and mechanisms underlying T-DXd-induced ILD/pneumonitis. This review highlights critical gaps in understanding the risk factors and mechanisms of T-DXd-induced ILD, underscoring the need for further research.
Collapse
Affiliation(s)
- Dehua Liao
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Jiwen Zhang
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
- School of Pharmacy, University of South China, Hengyang, People’s Republic of China
| | - Ting Yan
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Yun Chen
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Yilan Fu
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Ning Xie
- Medical Department of Breast Cancer, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Minghui Long
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
6
|
Malemnganba T, Pandey AK, Mishra A, Mehrotra S, Prajapati VK. Exploring immunotherapy with antibody-drug conjugates in solid tumor oncology. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 144:259-286. [PMID: 39978968 DOI: 10.1016/bs.apcsb.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Immunotherapy has emerged as a hallmark of hope in the formidable battle against solid tumors such as breast cancer, colorectal cancer, etc., with antibody-drug conjugates (ADCs) starting a new era of precision medicine. This chapter delves into the dynamic landscape of immunotherapeutic strategies, focusing on the transformative potential of ADCs. ADCs represent a combination of chemotherapy and immunotherapy, more innovative chemotherapy. We emphasize the intricate interplay between tumor biology and therapeutic intervention, uncovering the mechanisms underlying ADC efficacy and the hurdles they must overcome. Each facet of ADC development is carefully examined, from the delicate balance between payload potency and safety to the quest for enhanced tumor penetration. We also elucidate the synergistic potential of combining ADCs with existing modalities, including chemotherapy and radiation therapy, to amplify therapeutic outcomes while mitigating adverse effects. As we navigate the complexities of solid tumor oncology, a profound understanding of the immunotherapeutic potential of ADCs is gained, offering hope for a cure for patients and clinicians alike. Henceforth, we delve into this transformative journey as we advance in solid tumor treatment regimens using immunotherapy with ADCs, poised at the forefront of oncological innovation.
Collapse
Affiliation(s)
- Takhellambam Malemnganba
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Anurag Kumar Pandey
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, UP, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
7
|
Hamilton E, Galsky MD, Ochsenreither S, Del Conte G, Martín M, De Miguel MJ, Yu EY, Williams A, Gion M, Tan AR, Agrawal L, Rutten A, Machiels JP, Cresta S, Debruyne PR, Hennequin A, Moreno V, Minchom A, Valdes-Albini F, Petrylak D, Li L, Tsuchihashi Z, Suto F, Cheng FC, Kandil M, Barrios D, Hurvitz S. Trastuzumab Deruxtecan with Nivolumab in HER2-Expressing Metastatic Breast or Urothelial Cancer: Analysis of the Phase Ib DS8201-A-U105 Study. Clin Cancer Res 2024; 30:5548-5558. [PMID: 39405343 PMCID: PMC11647201 DOI: 10.1158/1078-0432.ccr-24-1513] [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: 06/06/2024] [Revised: 07/31/2024] [Accepted: 10/10/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE This multicenter phase Ib study investigated trastuzumab deruxtecan (T-DXd) plus nivolumab in patients with HER2-expressing metastatic breast cancer (mBC) and metastatic urothelial cancer (mUC). PATIENTS AND METHODS Part 1 determined the recommended dose for expansion of T-DXd plus nivolumab. Part 2 evaluated efficacy and safety; the primary endpoint was confirmed objective response rate by independent central review. RESULTS In part 1, seven patients with mBC were enrolled and received T-DXd 3.2 mg/kg (four patients) or 5.4 mg/kg (three patients) plus nivolumab. The recommended dose for expansion for T-DXd was 5.4 mg/kg plus nivolumab 360 mg intravenously every 3 weeks. In part 2, 32 patients with HER2-positive mBC (cohort 1; inclusive of three administered 5.4 mg/kg in part 1), 16 with HER2-low mBC (cohort 2), 30 with HER2-high mUC (cohort 3), and four with HER2-low mUC (cohort 4) were enrolled. At data cutoff (July 22, 2021), the confirmed objective response rates (95% confidence interval) for cohorts 1 to 4 were 65.6% (46.8%-81.4%), 50.0% (24.7%-75.3%), 36.7% (19.9%-56.1%), and not assessed due to small sample size, respectively. The median treatment duration (range) with T-DXd in cohorts 1 to 4 was 8.9 (1-23) months, 6.9 (1-21) months, 3.9 (1-21) months, and not assessed, respectively; the most common treatment-emergent adverse event was nausea (55.2%, 62.5%, 73.3%, and 75.0%, respectively). Adjudicated drug-related interstitial lung disease/pneumonitis rates (cohorts 1-3) were 20.7%, 0%, and 20.0%, respectively (one grade 5 each, cohorts 1 and 3). CONCLUSIONS T-DXd plus nivolumab demonstrated promising antitumor activity in HER2-expressing mBC or mUC and safety consistent with the known profile of T-DXd. Interstitial lung disease/pneumonitis is an important risk and requires careful monitoring and prompt intervention.
Collapse
Affiliation(s)
- Erika Hamilton
- Department of Medical Oncology, Sarah Cannon Research Institute, Nashville, Tennessee
| | - Matthew D. Galsky
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sebastian Ochsenreither
- Department of Hematology, Oncology and Cancer Immunology, Charité Comprehensive Cancer Center, Berlin, Germany
| | - Gianluca Del Conte
- Department of Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Miguel Martín
- Department of Medical Oncology, Hospital Gregorio Maranon, Madrid, Spain
| | - Maria José De Miguel
- Early Phase Clinical Trial Unit, START Madrid CIOCC HM Sanchinarro, Madrid, Spain
| | - Evan Y. Yu
- Department of Medicine, Fred Hutchinson Cancer Center and University of Washington, Seattle, Washington
| | - Anja Williams
- Clinical Research, Sarah Cannon Research Institute, London, United Kingdom
| | - Maria Gion
- Department of Medical Oncology, Hospital Ruber International, Madrid, Spain
- Ramón y Cajal University Hospital, Madrid, Spain
| | - Antoinette R. Tan
- Department of Solid Tumor Oncology and Investigational Therapeutics, Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | - Laila Agrawal
- Department of Medical Oncology, Norton Cancer Institute, Louisville, Kentucky
| | - Annemie Rutten
- Department of Medical Oncology, GZA Hospitals Campus Sint-Augustinus, Antwerp, Belgium
| | - Jean-Pascal Machiels
- Department of Medical Oncology, Institut Roi Albert II Cliniques Universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale (UC Louvain, IREC), Brussels, Belgium
| | - Sara Cresta
- Division of Medical Oncology, Department of Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Philip R. Debruyne
- Kortrijk Cancer Centre, General Hospital AZ Groeninge, Kortrijk, Belgium
- Medical Technology Research Centre (MTRC), School of Life Sciences, Anglia Ruskin University, Cambridge, United Kingdom
- School of Nursing and Midwifery, University of Plymouth, Plymouth, United Kingdom
| | - Audrey Hennequin
- Department of Medical Oncology, Centre Georges Francois Leclerc, Dijon, France
| | - Victor Moreno
- Early Phase Trials Unit, START Madrid-FJD, Fundacion Jimenez Diaz, Madrid, Spain
| | - Anna Minchom
- Drug Development Unit, Royal Marsden Hospital, Sutton, United Kingdom
| | - Frances Valdes-Albini
- Division of Medical Oncology, Department of Medicine, University of Miami Hospital & Clinics, Sylvester Comprehensive Cancer Center, Miami, Florida
- Department of Medical Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Daniel Petrylak
- Division of Genitourinary Oncology, Yale University, New Haven, Connecticut
| | - Li Li
- Quantitative Clinical Pharmacology, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | - Zenta Tsuchihashi
- Department of Translational Science, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | - Fumitaka Suto
- Department of Translational Science, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | - Fu-Chih Cheng
- Biostatistics and Data Management, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | - Maha Kandil
- CSPV, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | - Daniel Barrios
- Department of Global Oncology R&D, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | - Sara Hurvitz
- Department of Medicine, Fred Hutchinson Cancer Center and University of Washington, Seattle, Washington
- Division of Hematology and Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
8
|
Scheuher B, Ghusinga KR, McGirr K, Nowak M, Panday S, Apgar J, Subramanian K, Betts A. Towards a platform quantitative systems pharmacology (QSP) model for preclinical to clinical translation of antibody drug conjugates (ADCs). J Pharmacokinet Pharmacodyn 2024; 51:429-447. [PMID: 37787918 DOI: 10.1007/s10928-023-09884-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/16/2023] [Indexed: 10/04/2023]
Abstract
A next generation multiscale quantitative systems pharmacology (QSP) model for antibody drug conjugates (ADCs) is presented, for preclinical to clinical translation of ADC efficacy. Two HER2 ADCs (trastuzumab-DM1 and trastuzumab-DXd) were used for model development, calibration, and validation. The model integrates drug specific experimental data including in vitro cellular disposition data, pharmacokinetic (PK) and tumor growth inhibition (TGI) data for T-DM1 and T-DXd, as well as system specific data such as properties of HER2, tumor growth rates, and volumes. The model incorporates mechanistic detail at the intracellular level, to account for different mechanisms of ADC processing and payload release. It describes the disposition of the ADC, antibody, and payload inside and outside of the tumor, including binding to off-tumor, on-target sinks. The resulting multiscale PK model predicts plasma and tumor concentrations of ADC and payload. Tumor payload concentrations predicted by the model were linked to a TGI model and used to describe responses following ADC administration to xenograft mice. The model was translated to humans and virtual clinical trial simulations were performed that successfully predicted progression free survival response for T-DM1 and T-DXd for the treatment of HER2+ metastatic breast cancer, including differential efficacy based upon HER2 expression status. In conclusion, the presented model is a step toward a platform QSP model and strategy for ADCs, integrating multiple types of data and knowledge to predict ADC efficacy. The model has potential application to facilitate ADC design, lead candidate selection, and clinical dosing schedule optimization.
Collapse
Affiliation(s)
- Bruna Scheuher
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
- DMPK and Modeling, Takeda, Boston, MA, United States
| | | | - Kimiko McGirr
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
| | | | - Sheetal Panday
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
| | - Joshua Apgar
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
| | - Kalyanasundaram Subramanian
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA
- Differentia Bio, Pleasanton, California, United States
| | - Alison Betts
- Applied BioMath, 561 Virginia Road, Concord, MA, 01742, USA.
- DMPK and Modeling, Takeda, Boston, MA, United States.
| |
Collapse
|
9
|
Malla RR, Nellipudi HR, Srilatha M, Nagaraju GP. HER-2 positive gastric cancer: Current targeted treatments. Int J Biol Macromol 2024; 274:133247. [PMID: 38906351 DOI: 10.1016/j.ijbiomac.2024.133247] [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: 01/03/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
Gastric cancer (GC) is highly metastatic and characterized by HER2 amplification. Aberrant HER2 expression drives metastasis, therapy resistance, and tumor recurrence. HER2 amplification contributes to drug resistance by upregulating DNA repair enzymes and drug afflux proteins, reducing drug efficacy. HER2 modulates transcription factors critical for cancer stem cell properties, further impacting drug resistance. HER2 activity is influenced by HER-family ligands, promoting oncogenic signaling. These features point to HER2 as a targetable driver in GC. This review outlines recent advances in HER2-mediated mechanisms and their upstream and downstream signaling pathways in GC. Additionally, it discusses preclinical research investigation that comprehends trastuzumab-sensitizing phytochemicals, chemotherapeutics, and nanoparticles as adjunct therapies. These developments hold promise for improving outcomes and enhancing the management of HER2-positive GC.
Collapse
Affiliation(s)
- Rama Rao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, AP 530045, India
| | | | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati 517502, AP, India
| | | |
Collapse
|
10
|
High P, Guernsey C, Subramanian S, Jacob J, Carmon KS. The Evolving Paradigm of Antibody-Drug Conjugates Targeting the ErbB/HER Family of Receptor Tyrosine Kinases. Pharmaceutics 2024; 16:890. [PMID: 39065587 PMCID: PMC11279420 DOI: 10.3390/pharmaceutics16070890] [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: 06/14/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
Current therapies targeting the human epidermal growth factor receptor (HER) family, including monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs), are limited by drug resistance and systemic toxicities. Antibody-drug conjugates (ADCs) are one of the most rapidly expanding classes of anti-cancer therapeutics with 13 presently approved by the FDA. Importantly, ADCs represent a promising therapeutic option with the potential to overcome traditional HER-targeted therapy resistance by delivering highly potent cytotoxins specifically to HER-overexpressing cancer cells and exerting both mAb- and payload-mediated antitumor efficacy. The clinical utility of HER-targeted ADCs is exemplified by the immense success of HER2-targeted ADCs including trastuzumab emtansine and trastuzumab deruxtecan. Still, strategies to improve upon existing HER2-targeted ADCs as well as the development of ADCs against other HER family members, particularly EGFR and HER3, are of great interest. To date, no HER4-targeting ADCs have been reported. In this review, we extensively detail clinical-stage EGFR-, HER2-, and HER3-targeting monospecific ADCs as well as novel clinical and pre-clinical bispecific ADCs (bsADCs) directed against this receptor family. We close by discussing nascent trends in the development of HER-targeting ADCs, including novel ADC payloads and HER ligand-targeted ADCs.
Collapse
Affiliation(s)
- Peyton High
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Cara Guernsey
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Shraddha Subramanian
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Joan Jacob
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
| | - Kendra S. Carmon
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
| |
Collapse
|
11
|
Jiang M, Li Q, Xu B. Spotlight on ideal target antigens and resistance in antibody-drug conjugates: Strategies for competitive advancement. Drug Resist Updat 2024; 75:101086. [PMID: 38677200 DOI: 10.1016/j.drup.2024.101086] [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: 10/24/2023] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a novel and promising approach in targeted therapy, uniting the specificity of antibodies that recognize specific antigens with payloads, all connected by the stable linker. These conjugates combine the best targeted and cytotoxic therapies, offering the killing effect of precisely targeting specific antigens and the potent cell-killing power of small molecule drugs. The targeted approach minimizes the off-target toxicities associated with the payloads and broadens the therapeutic window, enhancing the efficacy and safety profile of cancer treatments. Within precision oncology, ADCs have garnered significant attention as a cutting-edge research area and have been approved to treat a range of malignant tumors. Correspondingly, the issue of resistance to ADCs has gradually come to the fore. Any dysfunction in the steps leading to the ADCs' action within tumor cells can lead to the development of resistance. A deeper understanding of resistance mechanisms may be crucial for developing novel ADCs and exploring combination therapy strategies, which could further enhance the clinical efficacy of ADCs in cancer treatment. This review outlines the brief historical development and mechanism of ADCs and discusses the impact of their key components on the activity of ADCs. Furthermore, it provides a detailed account of the application of ADCs with various target antigens in cancer therapy, the categorization of potential resistance mechanisms, and the current state of combination therapies. Looking forward, breakthroughs in overcoming technical barriers, selecting differentiated target antigens, and enhancing resistance management and combination therapy strategies will broaden the therapeutic indications for ADCs. These progresses are anticipated to advance cancer treatment and yield benefits for patients.
Collapse
Affiliation(s)
- Mingxia Jiang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiao Li
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Binghe Xu
- Department of Medical Oncology, State Key Laboratory of Mocelular Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
12
|
Kuwata T. Molecular classification and intratumoral heterogeneity of gastric adenocarcinoma. Pathol Int 2024; 74:301-316. [PMID: 38651937 PMCID: PMC11551831 DOI: 10.1111/pin.13427] [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: 01/21/2024] [Revised: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024]
Abstract
Gastric cancers frequently harbor striking histological complexity and diversity between lesions as well as within single lesions, known as inter- and intratumoral heterogeneity, respectively. The latest World Health Organization Classification of Tumors designated more than 30 histological subtypes for gastric epithelial tumors, assigning 12 subtypes for gastric adenocarcinoma (GAD). Meanwhile, recent advances in genome-wide analyses have provided molecular aspects to the histological classification of GAD, and consequently revealed different molecular traits underlying these histological subtypes. Moreover, accumulating knowledge of comprehensive molecular profiles has led to establishing molecular classifications of GAD, which are often associated with clinical biomarkers for therapeutics and prognosis. However, most of our knowledge of GAD molecular profiles is based on inter-tumoral heterogeneity, and the molecular profiles underlying intratumoral heterogeneity are yet to be determined. In this review, recently established molecular classifications of GAD are introduced in the aspect of pathological diagnosis and are discussed in the context of intratumoral heterogeneity.
Collapse
Affiliation(s)
- Takeshi Kuwata
- Department of Genetic Medicine and ServicesNational Cancer Center Hospital EastKashiwaChibaJapan
| |
Collapse
|
13
|
Wang H, Nie C, Xu W, Li J, Gou H, Lv H, Chen B, Wang J, Liu Y, He Y, Zhao J, Chen X. In era of immunotherapy: the value of trastuzumab beyond progression in patients with trastuzumab-resistant HER2-positive advanced or metastatic gastric cancer. Therap Adv Gastroenterol 2024; 17:17562848241245455. [PMID: 38617123 PMCID: PMC11010747 DOI: 10.1177/17562848241245455] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/20/2024] [Indexed: 04/16/2024] Open
Abstract
Background For patients with human epidermal growth factor receptor-2 (HER2)-positive advanced or metastatic gastric cancer who have progressed on first-line trastuzumab therapy, the clinical value of the continuous use of trastuzumab beyond progression (TBP) is controversial. Objectives The present study was conducted to evaluate the efficacy and explore new treatment strategies of TBP for patients with trastuzumab-resistant HER2-positive advanced or metastatic gastric cancer in the era of cancer immunotherapy. Design Retrospective analysis. Methods Patients with HER2-positive advanced or metastatic gastric cancer who have failed first-line treatment based on trastuzumab-targeted therapy from June 2019 to December 2020 were retrospectively analyzed. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), objective response rate (ORR), disease control rate (DCR), and safety. Survival curves of patients were estimated by the Kaplan-Meier method and compared using the log-rank test. Results In all, 30 patients received TBP with chemotherapy, immunotherapy, or anti-angiogenic therapy, and the other 26 patients received treatment of physician's choice without trastuzumab. The median PFS in the TBP and non-TBP population was 6.0 [95% confidence interval (CI) = 3.8-8.2] and 3.5 (95% CI = 2.2-4.8) months, respectively (p = 0.038), and the median OS was 12.3 (95% CI = 10.4-14.2) and 9.0 (95% CI = 6.6-11.4) months (p = 0.008). The patients who received TBP treatment had more favorable PFS and OS than the non-TBP population. In the TBP group, patients who received trastuzumab plus chemotherapy and immunotherapy had higher ORR (40.0% versus 16.7%), DCR (90.0% versus 50.0%), and showed a significant improvement in PFS (7.0 versus 1.9 m) compared to TBP with chemotherapy alone. Subgroup analysis suggested that patients with male, HER2 positive with immunohistochemistry score 3+ and PFS of first-line treatment less than 6 months had a greater benefit from TBP. The incidence of Grade 3-4 adverse events in the TBP and non-TBP groups was 43.3% and 38.5%. Conclusion The continuous use of TBP improves PFS and OS in patients with trastuzumab-resistant HER2-positive advanced or metastatic gastric cancer with well-tolerated toxicity. In the era of immunotherapy, TBP combined with chemotherapy and immunotherapy may further enhance the clinical benefit and provide a new treatment strategy. Trial registration This study is a retrospective study, which does not require clinical registration.
Collapse
Affiliation(s)
- Hui Wang
- Department of Endoscopic Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Caiyun Nie
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Weifeng Xu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Jing Li
- Department of Endoscopic Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - He Gou
- Department of Endoscopic Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Huifang Lv
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Beibei Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Jianzheng Wang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Yingjun Liu
- Department of General Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yunduan He
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Jing Zhao
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Xiaobing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127 Dongming Road, Jinshui, Zhengzhou, Henan 450008, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| |
Collapse
|
14
|
Sasso J, Tenchov R, Bird R, Iyer KA, Ralhan K, Rodriguez Y, Zhou QA. The Evolving Landscape of Antibody-Drug Conjugates: In Depth Analysis of Recent Research Progress. Bioconjug Chem 2023; 34:1951-2000. [PMID: 37821099 PMCID: PMC10655051 DOI: 10.1021/acs.bioconjchem.3c00374] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Antibody-drug conjugates (ADCs) are targeted immunoconjugate constructs that integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity. Their design allows for higher doses of the cytotoxic drug to be administered, potentially increasing efficacy. They are currently among the most promising drug classes in oncology, with efforts to expand their application for nononcological indications and in combination therapies. Here we provide a detailed overview of the recent advances in ADC research and consider future directions and challenges in promoting this promising platform to widespread therapeutic use. We examine data from the CAS Content Collection, the largest human-curated collection of published scientific information, and analyze the publication landscape of recent research to reveal the exploration trends in published documents and to provide insights into the scientific advances in the area. We also discuss the evolution of the key concepts in the field, the major technologies, and their development pipelines with company research focuses, disease targets, development stages, and publication and investment trends. A comprehensive concept map has been created based on the documents in the CAS Content Collection. We hope that this report can serve as a useful resource for understanding the current state of knowledge in the field of ADCs and the remaining challenges to fulfill their potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
15
|
Mikami Y, Iwase F, Ohshima D, Tomida T, Adachi-Akahane S. Compensatory role of neuregulin-1 in diabetic cardiomyopathy. J Pharmacol Sci 2023; 153:130-141. [PMID: 37770154 DOI: 10.1016/j.jphs.2023.08.009] [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: 07/13/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Diabetes mellitus is a prevalent risk factor for congestive heart failure. Diabetic cardiomyopathy patients present with left ventricular (LV) diastolic dysfunction at an early stage, then systolic dysfunction as the disease progresses. The mechanism underlying the development of diabetic cardiomyopathy has not yet been fully understood. This study aimed to elucidate the mechanisms by which diastolic dysfunction precedes systolic dysfunction at the early stage of diabetic cardiomyopathy. We hypothesized that the downregulation of cardioprotective factors is involved in the pathogenesis of diabetic cardiomyopathy. LV diastolic dysfunction, but not systolic dysfunction, was observed in type-1 diabetes mellitus model mice 4 weeks after STZ administration (STZ-4W), mimicking the early stage of diabetic cardiomyopathy. Counter to expectations, neuregulin-1 (NRG1) was markedly upregulated in the vascular endothelial cell in the ventricles of STZ-4W mice. To clarify the functional significance of the upregulated NRG1, we blocked its receptor ErbB2 with trastuzumab (TRZ). In STZ-4W mice, TRZ significantly reduced the systolic function without affecting diastolic function and caused a more prominent reduction in Akt phosphorylation levels. These results indicate that the compensatory upregulated NRG1 contributes to maintaining the LV systolic function, which explains why diastolic dysfunction precedes systolic dysfunction at the early stage of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yoshinori Mikami
- Department of Physiology, Faculty of Medicine, Toho University, Tokyo 143-8540, Japan
| | - Fumiki Iwase
- Department of Physiology, Faculty of Medicine, Toho University, Tokyo 143-8540, Japan
| | - Daisuke Ohshima
- Department of Physiology, Faculty of Medicine, Toho University, Tokyo 143-8540, Japan
| | - Taichiro Tomida
- Department of Physiology, Faculty of Medicine, Toho University, Tokyo 143-8540, Japan
| | - Satomi Adachi-Akahane
- Department of Physiology, Faculty of Medicine, Toho University, Tokyo 143-8540, Japan.
| |
Collapse
|
16
|
Kang C. Trastuzumab Deruxtecan: A Review in Gastric or Gastro-Oesophageal Junction Adenocarcinoma. Target Oncol 2023; 18:981-989. [PMID: 37787931 DOI: 10.1007/s11523-023-00998-y] [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: 08/31/2023] [Indexed: 10/04/2023]
Abstract
Trastuzumab deruxtecan (Enhertu®) is a human epidermal growth factor receptor type 2 (HER2)-directed antibody-drug conjugate that is approved in several countries globally for adults with advanced HER2-positive gastric or gastro-oesophageal junction (GOJ) adenocarcinoma who have received a prior trastuzumab-based regime. In the phase II DESTINY-Gastric01 trial, intravenous trastuzumab deruxtecan was significantly more effective than standard chemotherapy (physician's choice of intravenous irinotecan or paclitaxel) in achieving objective response and improving overall survival in Japanese or South Korean adults with advanced HER2-positive gastric or GOJ adenocarcinoma who had received two or more previous therapies. In the phase II DESTINY-Gastric02 trial, trastuzumab deruxtecan was able to induce durable response in adults from the USA or Europe with unresectable or metastatic HER2-positive gastric or GOJ adenocarcinoma. Trastuzumab deruxtecan was generally tolerable in these patients; the most common adverse events included nausea, neutropenia, fatigue and decreased appetite. Trastuzumab deruxtecan carries regulatory warnings (including boxed warnings in the USA) for interstitial lung disease/pneumonitis and embryo-foetal toxicity. Current evidence indicates that trastuzumab deruxtecan is an effective treatment option, and is generally tolerable, in previously treated adults with advanced HER2-positive gastric or GOJ adenocarcinoma.
Collapse
Affiliation(s)
- Connie Kang
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand.
| |
Collapse
|
17
|
Klein-Scory S, Ladigan-Badura S, Mika T, Verdoodt B, Tannapfel A, Pohl M, Schroers R, Baraniskin A. Liquid biopsy based HER2 amplification status in gastric cancer patients indicates clinical response. Heliyon 2023; 9:e21339. [PMID: 38027576 PMCID: PMC10665680 DOI: 10.1016/j.heliyon.2023.e21339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Gastric carcinomas are among the most common cancers in Germany, with approximately 18,000 new cases per year. About 10 years ago, based on results of the Trastuzumab for gastric cancer (ToGA) trial, the addition of the monoclonal antibody trastuzumab to a platinum-fluoropyrimidine chemotherapy backbone became the standard-of-care 1st-line therapy for human epidermal growth factor receptor 2 (HER2)-positive gastric cancers. Only patients with primary HER2 gene amplification benefit from this therapy. Thus, accurate HER2 gene amplification detection is predictive and critical for therapy selection. As a gold standard the HER2 status is currently determined in tumor tissue specimens using immune histochemistry and fluorescent in situ hybridisation. However, HER2 amplification is detectable in only about 20 % of gastric carcinomas. The recent approval of an antibody-drug conjugate Trastuzumab deruxtecan (T-DXd) and the establishment of a new subgroup of HER2-low tumors due to the bystander effect associated with T-DXd increases the relevance of precise HER2 diagnostics. Aim of this analysis was to determine the HER2 amplification status from circulating DNA fragments in blood using a HER2 Copy Number Variation assay to establish a minimal invasive approach. For the present study, a digital droplet PCR-based method was validated relative to established tissue-based methods. Furthermore and most importantly, the changes of HER2 status during therapy were investigated in seven patients indicating that the changes of HER2 status and number of HER2 copies detected in blood can reflect on therapy efficiency and uncover treatment resistance.
Collapse
Affiliation(s)
- Susanne Klein-Scory
- Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum GmbH, Germany
| | - Swetlana Ladigan-Badura
- Department of Hematology, Oncology and Palliative Care, Evangelical Hospital Hamm gGmbH, Germany
| | - Thomas Mika
- Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum GmbH, Germany
| | - Berlinda Verdoodt
- Department of Medicine, Institute of Pathology, Ruhr University Bochum, Germany
| | - Andrea Tannapfel
- Department of Medicine, Institute of Pathology, Ruhr University Bochum, Germany
| | - Michael Pohl
- Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum GmbH, Germany
| | - Roland Schroers
- Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum GmbH, Germany
| | - Alexander Baraniskin
- Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum GmbH, Germany
- Department of Hematology, Oncology and Palliative Care, Evangelical Hospital Hamm gGmbH, Germany
| |
Collapse
|
18
|
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
|
19
|
Hu Q, Oki E, Yamada T, Kashiwada T, Sonoda H, Kataoka M, Kawanaka H, Tsuji Y, Makiyama A, Nakashima Y, Ota M, Kimura Y, Yoshizumi T. Genomic characterization between HER2-positive and negative gastric cancer patients in a prospective trial. Cancer Med 2023; 12:16649-16660. [PMID: 37325934 PMCID: PMC10469643 DOI: 10.1002/cam4.6269] [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: 05/02/2023] [Revised: 05/27/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND We aimed to clarify the genomic characteristics of HER2-positive and negative gastric cancer cases that potentially affect tumor progression and treatment response in a prospective trial. METHODS We collected 80 formalin-fixed paraffin-embedded (FFPE) samples (49 HER2+ and 31 HER2-) from gastric cancer patients who participated in the TROX-A1 trial (UMIN000036865). We queried a 435-gene panel (CANCERPLEX-JP) to generate comprehensive genomic profiling data, including the tumor mutation burden, somatic mutations, and copy number variations. In addition, the genomic differences between HER2+ and HER2- gastric cancer patients were analyzed. RESULTS Mutational analyses showed that TP53 was the most frequently mutated gene regardless of HER2 status. ARID1A mutation was significantly enriched in HER2-negative patients. The number of total mutations in HER2-negative patients with ARID1A mutation was remarkably higher than that in HER2-positive patients. Next, copy number variation analyses showed that the number of amplified genes (such as CCNE1, PGAP3, and CDK12) in HER2-positive cases was significantly higher than that in HER2-negative cases. Moreover, PTEN deletion was more common in HER2-positive cases. Finally, we found that, compared with HER2-positive patients, HER2-negative patients tended to have a higher tumor mutation burden, particularly in patients with ARID1A mutation. Pathway analyses of the gene alterations showed an enrichment of several immune-related pathways in HER2-negative patients. CONCLUSIONS According to the genomic profiling of HER2-positive and negative gastric cancer, several gene alterations in the HER2 pathway may be the potential mechanism underlying trastuzumab resistance. Relative to HER2-positive gastric cancer, HER2-negative gastric tumors with ARID1A mutation may be sensitive to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Qingjiang Hu
- Department of Surgery and ScienceKyushu University HospitalFukuokaJapan
| | - Eiji Oki
- Department of Surgery and ScienceKyushu University HospitalFukuokaJapan
| | - Teppei Yamada
- Department of Gastroenterological SurgeryFukuoka University HospitalFukuokaJapan
| | - Tomomi Kashiwada
- Department of Medical OncologySaga Medical Center KoseikanSagaJapan
| | | | - Masato Kataoka
- Department of SurgeryNational Hospital Organization Nagoya Medical CenterNagoyaJapan
| | - Hirofumi Kawanaka
- Clinical Research Institute / Department of Gastroenterological SurgeryNational Hospital Organization Beppu Medical CenterBeppuJapan
| | - Yasushi Tsuji
- Department of Medical OncologyTonan HospitalSapporoJapan
| | | | | | - Mitsuhiko Ota
- Department of Surgery and ScienceKyushu University HospitalFukuokaJapan
| | - Yasue Kimura
- Department of Surgery and ScienceKyushu University HospitalFukuokaJapan
| | | |
Collapse
|
20
|
Sun T, Niu X, He Q, Liu M, Qiao S, Qi RQ. Development, efficacy and side effects of antibody‑drug conjugates for cancer therapy (Review). Mol Clin Oncol 2023; 18:47. [PMID: 37206431 PMCID: PMC10189422 DOI: 10.3892/mco.2023.2643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/22/2023] [Indexed: 05/21/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are anticancer drugs that combine cytotoxic small-molecule drugs (payloads) with monoclonal antibodies through a chemical linker and that transfer toxic payloads to tumor cells expressing target antigens. All ADCs are based on human IgG. In 2009, the Food and Drug Administration (FDA) approved gemtuzumab ozogamicin as the initial first-generation ADC. Since then, at least 100 ADC-related projects have been initiated, and 14 ADCs are currently being tested in clinical trials. The limited success of gemtuzumab ozogamicin has led to the development of optimization strategies for the next generation of drugs. Subsequently, experts have improved the first-generation ADCs and have developed second-generation ADCs such as ado-trastuzumab emtansine. Second-generation ADCs have higher specific antigen levels, more stable linkers and longer half-lives and show great potential to transform cancer treatment models. Since the first two generations of ADCs have served as a good foundation, the development of ADCs is accelerating, and third-generation ADCs, represented by trastuzumab deruxtecan, are ready for wide application. Third-generation ADCs are characterized by strong pharmacokinetics and high pharmaceutical activity, and their drug-to-antibody ratio mainly ranges from 2 to 4. In the past decade, the research prospects of ADCs have broadened, and an increasing number of specific antigen targets and mechanisms of cytotoxic drug release have been discovered and studied. To date, seven ADCs have been approved by the FDA for lymphoma, and three have been approved to treat breast cancer. The present review explores the function and development of ADCs and their clinical use in cancer treatment.
Collapse
Affiliation(s)
- Te Sun
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
| | - Xueli Niu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
| | - Qing He
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
| | - Min Liu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shuai Qiao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
- Correspondence to: Professor Rui-Qun Qi or Mrs. Shuai Qiao, Department of Dermatology, The First Hospital of China Medical University, 155 Nanjing Bei Street, Shenyang, Liaoning 110001, P.R. China
| | - Rui-Qun Qi
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
- Correspondence to: Professor Rui-Qun Qi or Mrs. Shuai Qiao, Department of Dermatology, The First Hospital of China Medical University, 155 Nanjing Bei Street, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
21
|
ARID1A deficiency is targetable by AKT inhibitors in HER2-negative gastric cancer. Gastric Cancer 2023; 26:379-392. [PMID: 36811690 DOI: 10.1007/s10120-023-01373-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND The PI3K/AKT signaling pathway is frequently activated in gastric cancer (GC); however, AKT inhibitors are not effective in unselected GC patients in clinical trials. Mutations in AT-rich interactive domain 1A (ARID1A), which are found in approximately 30% of GC patients, activate PI3K/AKT signaling, suggesting that targeting the ARID1A deficiency-activated PI3K/AKT pathway is a therapeutic candidate for ARID1A-deficient GC. METHODS The effect of AKT inhibitors was evaluated using cell viability and colony formation assays in ARID1A-deficient and ARID1A knockdown ARID1A-WT GC cells as well as in HER2-positive and HER2-negative GC. The Cancer Genome Atlas cBioPortal and Gene Expression Omnibus microarray databases were accessed to determine the extent of dependence of GC cell growth on the PI3K/AKT signaling pathway. RESULTS AKT inhibitors decreased the viability of ARID1A-deficient cells and the inhibitory effect was greater in ARID1A-deficient/HER2-negative GC cells. Bioinformatics data suggested that PI3K/AKT signaling plays a greater role in proliferation and survival in ARID1A-deficient/HER2-negative GC cells than in ARID1A-deficient/HER2-positive cells, supporting the higher therapeutic efficacy of AKT inhibitors. CONCLUSIONS The effect of AKT inhibitors on cell proliferation and survival is affected by HER2 status, providing a rationale for exploring targeted therapy using AKT inhibitors in ARID1A-deficient/HER2-negative GC.
Collapse
|
22
|
An anti-EGFR antibody-drug conjugate overcomes resistance to HER2-targeted drugs. Cancer Lett 2023; 554:216024. [PMID: 36455759 DOI: 10.1016/j.canlet.2022.216024] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/02/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
Trastuzumab-emtansine (T-DM1) is an antibody-drug conjugate (ADC) that was approved in 2013 to treat HER2+ breast cancer. Despite its efficacy in the clinic, some patients exhibit intrinsic or acquired resistance to such ADC. To characterize mechanisms of resistance to T-DM1, we isolated several HER2+ resistant clones derived from the HCC1954 HER2+ cell line. The isolated clones were different as per their transcriptomic profiles. However, all the T-DM1-resistant clones showed decreased HER2 levels. Yet, the clones were still oncogenically dependent on HER2, as indicated by knock down experiments. The decrease in HER2 expression caused acquired resistance to T-DM1 and to other anti-HER2 therapies. Antibody array analyses showed that the epidermal growth factor receptor (EGFR) was expressed in these T-DM1-resistant HCC1954 clones. Indeed, therapies targeting EGFR, particularly cetuximab-DM1, demonstrated a strong anti-proliferative action on cells with acquired resistance to T-DM1 and HER2 loss. The expression of EGFR in cells resistant to T-DM1 offers the possibility of using therapies directed to this receptor to combat resistance to anti-HER2 drugs and loss of HER2 overexpression.
Collapse
|
23
|
Advances in antibody-based therapy in oncology. NATURE CANCER 2023; 4:165-180. [PMID: 36806801 DOI: 10.1038/s43018-023-00516-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 01/10/2023] [Indexed: 02/22/2023]
Abstract
Monoclonal antibodies are a growing class of targeted cancer therapeutics, characterized by exquisite specificity, long serum half-life, high affinity and immune effector functions. In this review, we outline key advances in the field with a particular focus on recent and emerging classes of engineered antibody therapeutic candidates, discuss molecular structure and mechanisms of action and provide updates on clinical development and practice.
Collapse
|
24
|
Fu Z, Liu J, Li S, Shi C, Zhang Y. Treatment-related adverse events associated with HER2-Targeted antibody-drug conjugates in clinical trials: a systematic review and meta-analysis. EClinicalMedicine 2023; 55:101795. [PMID: 36712893 PMCID: PMC9874347 DOI: 10.1016/j.eclinm.2022.101795] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Given the increasing use of HER2-targeted antibody-drug conjugates (ADCs) worldwide, the summary of toxicity incidence and profiles of these drugs is crucial to provide reference for clinical application. This meta-analysis aimed to estimate the mean incidences of treatment-related adverse events of HER2-targeted ADCs and to investigate the differences between different drugs and cancer types. METHODS We performed a systematic search of literature in PubMed, Embase, Web of Science, and Scopus databases from inception to February 1, 2022 and the last search was updated to August 1, 2022. Published prospective clinical trials on single-agent of the US Food and Drug Administration approved HER2-targeted ADCs with available count data regarding treatment-related adverse events were included. The primary outcomes were pooled incidences of treatment-related adverse events and differences between different drugs and cancer types. The data synthesis was performed using a Bayesian hierarchical modelling method and the protocol was registered in PROSPERO (CRD42022331627). FINDINGS A total of 39 studies (37 trials) involving 7688 patients across five cancer types were included in the final analysis. On pooling the data using Bayesian hierarchical modelling, the overall mean incidence of all-grade adverse events, high-grade adverse events, serious adverse events, and adverse events that resulted in drug discontinuation were 98.29% (95% CrI, 97.33%-99.07%, τ = 1.49), 47.88% (95% CrI, 42.74%-53.17%, τ = 0.37), 19.45% (95% CrI, 15.70%-23.67%, τ = 0.55), and 10.52% (95% CrI, 8.03%-13.21%, τ = 0.56), respectively. The most common all-grade adverse events were nausea (41.57%; 95% CrI, 40.46%-42.64%, τ = 0.81), fatigue (35.86%; 95% CrI, 34.85%-36.96%, τ = 0.65), and decreased appetite (28.84%; 95% CrI, 22.93%-36.87%, τ = 0.76). The most common high-grade adverse events were thrombocytopenia (8.37%; 95% CrI, 7.75%-9.07%, τ = 0.71), anaemia (6.49%; 95% CrI, 5.86%-7.11%, τ = 1.06), and neutropenia (6.42%; 95% CrI, 5.76%-7.04%, τ = 1.21). We found no difference in the mean incidences of adverse events among different cancer types, as well as different dosing regimens. However, trastuzumab deruxtecan (T-DXd) appeared to have higher mean incidences of adverse events compared with trastuzumab emtansine (T-DM1), especially for the higher dose of T-DXd (6.4 mg/kg Q3W). INTERPRETATION The incidences of adverse events between two HER2-targeted ADCs were similar in different cancer types, but different HER2-targeted ADCs appeared to have different mean incidences of adverse events. The comprehensive summary of the adverse events of HER2-targeted ADCs is critical for clinicians caring for patients with cancer receiving HER2-targeted ADCs therapy. FUNDING The National Natural Science Foundation of China (Grant No. 82073402) and Key R&D Plan of Hubei Province, China (No.2020BCA060) funded this study.
Collapse
Affiliation(s)
- Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Province Clinical Research Centre for Precision Medicine for Critical Illness, China
| | - Jinmei Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Province Clinical Research Centre for Precision Medicine for Critical Illness, China
| | - Shijun Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Province Clinical Research Centre for Precision Medicine for Critical Illness, China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Province Clinical Research Centre for Precision Medicine for Critical Illness, China
- Corresponding author. Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Jianghan District, Wuhan, Hubei Province, 430000, China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Province Clinical Research Centre for Precision Medicine for Critical Illness, China
- Corresponding author. Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Jianghan District, Wuhan, Hubei Province, 430000, China.
| |
Collapse
|
25
|
Meric-Bernstam F, Beeram M, Hamilton E, Oh DY, Hanna DL, Kang YK, Elimova E, Chaves J, Goodwin R, Lee J, Nabell L, Rha SY, Mayordomo J, El-Khoueiry A, Pant S, Raghav K, Kim JW, Patnaik A, Gray T, Davies R, Ozog MA, Woolery J, Lee KW. Zanidatamab, a novel bispecific antibody, for the treatment of locally advanced or metastatic HER2-expressing or HER2-amplified cancers: a phase 1, dose-escalation and expansion study. Lancet Oncol 2022; 23:1558-1570. [DOI: 10.1016/s1470-2045(22)00621-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022]
|
26
|
Guo Z, Ding Y, Wang M, Liu J, Zhai Q, Du Q. Safety of trastuzumab deruxtecan: A meta-analysis and pharmacovigilance study. J Clin Pharm Ther 2022; 47:1837-1844. [PMID: 36200429 PMCID: PMC9827941 DOI: 10.1111/jcpt.13777] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/04/2022] [Indexed: 01/12/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE This study aimed to explore the safety profile of trastuzumab deruxtecan (T-DXd, formerly DS-8201a) using multi-source medical data. METHODS We explored trastuzumab deruxtecan related adverse events (AEs) in clinical trials available in ClinicalTrials.gov and electronic databases (MEDLINE, EMBASE and PubMed) up to July 16, 2022. Meta-analysis was performed by using incidence rate with 95%CIs. In the pharmacovigilance study of FDA Adverse Event Reporting System (FAERS), the reporting odds ratio (ROR) and the medicines and healthcare products regulatory agency (MHRA) methods were used to analyse the real-world AEs (up to June 28, 2022). RESULTS AND DISCUSSION A 8 clinical trials enrolled 1457 patients were included. The most common AEs of any grade were gastrointestinal disorders and blood and lymphatic system disorders. The most common AE of grade 3 or higher was neutropenia (21.4%, 95%CI: 14.7%-28.1%, I2 = 91%). The incidence of interstitial lung disease (ILD) and decreased left ventricular ejection fraction were 10.9% (95%CI: 7.2%-14.5%, I2 = 82%) and 1.2% (95%CI: 0.7%-2.2%, I2 = 98%), respectively. A total of 1244 AE reports were identified in the pharmacovigilance study. Gastrointestinal toxicity (ROR = 21.65), myelosuppression (ROR = 36.88), interstitial lung disease (ROR = 50.30), pneumonitis (ROR = 36.59), decreased ejection fraction (ROR = 16.08), and taste disorder (ROR = 14.06) mentioned in the instructions showed strong signals. Also, ascites (ROR = 14.90), lung opacity (ROR = 78.80), pulmonary fibrosis (ROR = 5.59), and increased KL-6 (ROR = 1761.97), which were not mentioned in the instructions, showed strong signals. WHAT IS NEW AND CONCLUSION Trastuzumab deruxtecan was well tolerated, and more attention should be paid on ILD as well as decreased ejection fraction.
Collapse
Affiliation(s)
- Zihan Guo
- Department of PharmacyFudan University Shanghai Cancer CenterShanghaiChina,Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Yunlan Ding
- Department of PharmacyFudan University Shanghai Cancer CenterShanghaiChina,Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Mengmeng Wang
- Department of PharmacyFudan University Shanghai Cancer CenterShanghaiChina,Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Jiyong Liu
- Department of PharmacyFudan University Shanghai Cancer CenterShanghaiChina,Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Qing Zhai
- Department of PharmacyFudan University Shanghai Cancer CenterShanghaiChina,Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Qiong Du
- Department of PharmacyFudan University Shanghai Cancer CenterShanghaiChina,Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| |
Collapse
|
27
|
Ma Z, Zhang Y, Zhu M, Feng L, Zhang Y, An Z. Interstitial lung disease associated with anti-HER2 anti-body drug conjugates: results from clinical trials and the WHO's pharmacovigilance database. Expert Rev Clin Pharmacol 2022; 15:1351-1361. [DOI: 10.1080/17512433.2022.2121705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Zhuo Ma
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yi Zhang
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Min Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Lin Feng
- Department of Clinical Epidemiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Yuhui Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Zhuoling An
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Maiuolo J, Musolino V, Gliozzi M, Carresi C, Oppedisano F, Nucera S, Scarano F, Scicchitano M, Guarnieri L, Bosco F, Macrì R, Ruga S, Cardamone A, Coppoletta AR, Ilari S, Mollace A, Muscoli C, Cognetti F, Mollace V. The Employment of Genera Vaccinium, Citrus, Olea, and Cynara Polyphenols for the Reduction of Selected Anti-Cancer Drug Side Effects. Nutrients 2022; 14:1574. [PMID: 35458136 PMCID: PMC9025632 DOI: 10.3390/nu14081574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/02/2022] [Accepted: 04/08/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is one of the most widespread diseases globally and one of the leading causes of death. Known cancer treatments are chemotherapy, surgery, radiation therapy, targeted hormonal therapy, or a combination of these methods. Antitumor drugs, with different mechanisms, interfere with cancer growth by destroying cancer cells. However, anticancer drugs are dangerous, as they significantly affect both cancer cells and healthy cells. In addition, there may be the onset of systemic side effects perceived and mutagenicity, teratogenicity, and further carcinogenicity. Many polyphenolic extracts, taken on top of common anti-tumor drugs, can participate in the anti-proliferative effect of drugs and significantly reduce the side effects developed. This review aims to discuss the current scientific knowledge of the protective effects of polyphenols of the genera Vaccinium, Citrus, Olea, and Cynara on the side effects induced by four known chemotherapy, Cisplatin, Doxorubicin, Tamoxifen, and Paclitaxel. In particular, the summarized data will help to understand whether polyphenols can be used as adjuvants in cancer therapy, although further clinical trials will provide crucial information.
Collapse
Affiliation(s)
- Jessica Maiuolo
- Laboratoy of Pharmaceutical Biology, IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Canzaro, Italy;
| | - Vincenzo Musolino
- Laboratoy of Pharmaceutical Biology, IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Canzaro, Italy;
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
| | - Francesca Oppedisano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
| | - Lorenza Guarnieri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
| | - Roberta Macrì
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Antonio Cardamone
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Anna Rita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Sara Ilari
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
| | - Annachiara Mollace
- Medical Oncology 1, Regina Elena National Cancer Institute, IRCCS, 00144 Rome, Italy; (A.M.); (F.C.)
| | - Carolina Muscoli
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
| | - Francesco Cognetti
- Medical Oncology 1, Regina Elena National Cancer Institute, IRCCS, 00144 Rome, Italy; (A.M.); (F.C.)
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (C.C.); (F.O.); (S.N.); (F.S.); (M.S.); (L.G.); (F.B.); (R.M.); (S.R.); (A.C.); (A.R.C.); (S.I.); (V.M.)
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
| |
Collapse
|
29
|
Chia CSB. A Patent Review on FDA-Approved Antibody-Drug Conjugates, Their Linkers and Drug Payloads. ChemMedChem 2022; 17:e202200032. [PMID: 35384350 DOI: 10.1002/cmdc.202200032] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/22/2022] [Indexed: 12/30/2022]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a promising class of biologics since the first approval of Gemtuzumab ozogamicin in 2000. Compared to small molecule drugs, ADCs are structurally much more complex as they comprise of an antibody conjugated to cytotoxic payloads by specially-designed linkers. Correspondingly, the ADC patent landscape is also much more complex. This review collates and discusses the patents protecting ADCs approved by the FDA up to 31 December 2021, with particular emphasis on their linker and cytotoxin payload technologies.
Collapse
Affiliation(s)
- C S Brian Chia
- Experimental Drug Development Centre, 10 Biopolis Road, Chromos #08-01, 138670, Singapore, Singapore
| |
Collapse
|
30
|
Fu Z, Li S, Han S, Shi C, Zhang Y. Antibody drug conjugate: the "biological missile" for targeted cancer therapy. Signal Transduct Target Ther 2022; 7:93. [PMID: 35318309 PMCID: PMC8941077 DOI: 10.1038/s41392-022-00947-7] [Citation(s) in RCA: 722] [Impact Index Per Article: 240.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 02/08/2023] Open
Abstract
Antibody-drug conjugate (ADC) is typically composed of a monoclonal antibody (mAbs) covalently attached to a cytotoxic drug via a chemical linker. It combines both the advantages of highly specific targeting ability and highly potent killing effect to achieve accurate and efficient elimination of cancer cells, which has become one of the hotspots for the research and development of anticancer drugs. Since the first ADC, Mylotarg® (gemtuzumab ozogamicin), was approved in 2000 by the US Food and Drug Administration (FDA), there have been 14 ADCs received market approval so far worldwide. Moreover, over 100 ADC candidates have been investigated in clinical stages at present. This kind of new anti-cancer drugs, known as "biological missiles", is leading a new era of targeted cancer therapy. Herein, we conducted a review of the history and general mechanism of action of ADCs, and then briefly discussed the molecular aspects of key components of ADCs and the mechanisms by which these key factors influence the activities of ADCs. Moreover, we also reviewed the approved ADCs and other promising candidates in phase-3 clinical trials and discuss the current challenges and future perspectives for the development of next generations, which provide insights for the research and development of novel cancer therapeutics using ADCs.
Collapse
Affiliation(s)
- Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China
| | - Shijun Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, (Parkville Campus) 381 Royal Parade,, Parkville, VIC, 3052, Australia
- Faculty of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, People's Republic of China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
31
|
Zhang L, Hamdani O, Gjoerup O, Cho-Phan C, Snider J, Castellanos E, Nimeiri H, Frampton G, Venstrom JM, Oxnard G, Klempner SJ, Schrock AB. ERBB2 Copy Number as a Quantitative Biomarker for Real-World Outcomes to Anti-Human Epidermal Growth Factor Receptor 2 Therapy in Advanced Gastroesophageal Adenocarcinoma. JCO Precis Oncol 2022; 6:e2100330. [PMID: 35050711 PMCID: PMC8789214 DOI: 10.1200/po.21.00330] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Human epidermal growth factor receptor 2 (HER2) overexpression or amplification (ERBB2amp) are biomarkers for approved anti-HER2 therapies. ERBB2amp may better predict response compared with immunohistochemistry or in situ hybridization, and quantitative copy number (CN) may further stratify patients. We characterized ERBB2amp in advanced gastroesophageal adenocarcinomas (GEA) and hypothesized that increased CN was associated with better outcome to trastuzumab. METHODS Comprehensive genomic profiling, including assessment of ERBB2amp, was performed for 12,905 GEA tissue cases. Clinical outcomes were assessed using a clinicogenomic database linking deidentified electronic health record–derived clinical data to genomic data. Multivariable Cox proportional hazard models were used for real-world progression-free survival (rwPFS) comparisons. RESULTS ERBB2amp (CN ≥ 5) was detected in 15% (1,934 of 12,905) of GEA; median CN 22 (interquartile range 9-73). Median ERBB2 amplicon size was 0.27 megabase (interquartile range 0.13-0.95), and smaller amplicons were associated with higher CN (P < .001). In the clinicogenomic database, of 101 evaluable first-line trastuzumab-treated patients, ERBB2 CN was a significant predictor of rwPFS as a continuous variable (adjusted hazard ratio = 0.73; 95% CI, 0.60 to 0.89; P = .002), whereas ERBB2 CN was not predictive of rwPFS on chemotherapy (adjusted hazard ratio = 0.93; 95% CI, 0.73 to 1.20; P = .59). Among trastuzumab-treated patients, no significant associations with ERBB2 CN were observed for disease site, age, stage at advanced diagnosis, or most selected coalterations. CONCLUSION ERBB2amp was detected in 15% of GEA tissue samples, with significant diversity in ERBB2 CN and amplicon focality. ERBB2 CN was predictive of rwPFS as a continuous variable for patients treated with trastuzumab. Further studies exploring the clinical utility of quantitative ERBB2 CN, particularly in the setting of the evolving anti-HER2 landscape and combination therapies, are warranted. ERBB2 copy number is a quantitative biomarker for outcomes to anti-HER2 therapy in advanced gastroesophageal cancer.![]()
Collapse
|
32
|
Kahraman S, Yalcin S. Recent Advances in Systemic Treatments for HER-2 Positive Advanced Gastric Cancer. Onco Targets Ther 2021; 14:4149-4162. [PMID: 34285507 PMCID: PMC8286155 DOI: 10.2147/ott.s315252] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is the fifth most common cancer worldwide. Despite recent improvements in treatment quality and options, advanced gastric cancer remains one of the hardest to cure cancers, with a median overall survival (OS) of 10–12 months and a 5-year OS of approximately 5–20%. There is an unmet need for further efforts to palliate disease-related symptoms, improve quality of life, increase tumor response rate, and prolong progression free and overall survival while balancing the toxicities of therapy. The most common type of GC is adenocarcinoma, which demonstrates morphological, biological, and clinical heterogeneity. A plethora of genomic alterations and the activation of numerous molecular pathways including human epidermal growth receptor 2 (HER2), epidermal growth factor receptor (EGFR), fibroblast growth factor receptor-2 (FGFR2), mesenchymal epidermal transforming factor receptor (MET), and the phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) are responsible for the complex heterogeneity of GC. Efforts to validate the therapeutic effects of inhibiting some of these aberrantly expressed pathways have failed to lead to a clinically meaningful outcome apart from the overexpression/amplification of the HER2 gene, inhibition of which has had a significant impact on clinical practice. The only available biomarkers to guide the effective treatment of patients with advanced GC are HER2 overexpression, MSI/PD-L1 status, and FGFR alterations. Various anti-HER2 agents have been evaluated after the success of the ToGA trial, but none led to a significant enough clinical improvement to be considered a viable alternative for HER2-targeted therapy in advanced GC until the global Keynote-811 trial, which added pembrolizumab to trastuzumab in combination with chemotherapy. This combination demonstrated a survival advantage for the first time in the 11 years since ToGA. Trastuzumab deruxtecan (T-DXd) was also found to be effective in patients who had already received >2 previous lines of treatment. Despite these promising avenues, the optimal management of HER-2 positive GC still requires further development.
Collapse
Affiliation(s)
- Seda Kahraman
- Yıldırım Beyazıt University Faculty of Medicine, Department of Medical Oncology, Ankara, Turkey
| | - Suayib Yalcin
- Hacettepe University Institute of Cancer, Department of Medical Oncology, Ankara, Turkey
| |
Collapse
|