1
|
Michaels L, Noor M, Aslam T. Clinical and imaging strategies for the assessment of the ocular side effects of systemic targeted anti-cancer therapies. Eur J Cancer 2025; 222:115452. [PMID: 40306116 DOI: 10.1016/j.ejca.2025.115452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
Systemic targeted anti-cancer therapies selectively target cancerous cells whilst limiting systemic side effects. The eye however, is a particularly sensitive organ and the expanding use of the newer targeted chemotherapy agents has been associated with multiple ocular side effects. In this review we provide an update of the ocular side effects of the newer targeted chemotherapy agents along with suggested minimum, pragmatic, evidence-based strategies for effective screening or monitoring for potential ocular side effects. This framework is designed to guide oncologists, trial managers, protocol developers and regulatory authorities so that appropriate ophthalmic clinical examinations and non-invasive modern imaging can be requested and commissioned according to a patient's specific treatment.
Collapse
Affiliation(s)
- Luke Michaels
- St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, United Kingdom
| | - Maha Noor
- Manchester Royal Eye Hospital, Oxford Road, Manchester M13 9WL, United Kingdom
| | - Tariq Aslam
- Manchester Royal Eye Hospital, Oxford Road, Manchester M13 9WL, United Kingdom; School of Health Sciences, University of Manchester, Oxford Road, Manchester M139PL, United Kingdom.
| |
Collapse
|
2
|
Nicolò E, Gianni C, Tarantino P. Redefining Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer in the Era of Novel Antibody-Drug Conjugates. JCO Oncol Pract 2025:OP2500129. [PMID: 40359470 DOI: 10.1200/op-25-00129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 05/15/2025] Open
Abstract
The advent of next-generation antibody-drug conjugates (ADCs), particularly trastuzumab deruxtecan (T-DXd), has transformed our understanding of human epidermal growth factor receptor 2 (HER2) targetability for breast cancer (BC) treatment. Historically categorized as HER2-positive or HER2-negative on the basis of trastuzumab eligibility, this classification has evolved significantly over the past 5 years. The DESTINY-Breast04 trial marked the entry of anti-HER2 therapies for patients with HER2-low BC, while DESTINY-Breast06 demonstrated the potential for earlier and broader use of T-DXd. The latter trial revealed that even minimal HER2 expression in tumors previously classified as HER2-0 might be clinically relevant and targetable with T-DXd. This has led to further refinement of HER2 classification, introducing the concepts of HER2-ultralow (HER2-0 with staining) and HER2-null BC (HER2-0 without staining). With these findings, most patients with metastatic BC are currently considered eligible for T-DXd. Accurately identifying candidates for these therapies has highlighted the limitations of current HER2 diagnostic practices, on the basis of immunohistochemistry (IHC)/in situ hybridization assessment. IHC assay, optimized to detect high levels of HER2 protein, faces limitations in discriminating finer variations at the lower end of the HER2 expression spectrum. This is further complicated by the heterogeneity of HER2 expression. To overcome these barriers, new approaches may be required. Quantitative methods for HER2 membrane assessment, genomic and transcriptomic evaluations of HER2, and the integration of artificial intelligence into tissue analysis hold promise and are currently under investigation. Additionally, noninvasive strategies, such as analysis of circulating tumor DNA or circulating tumor cells, may enable real-time HER2 status assessment and better patient selection for ADC. However, these techniques require rigorous validation to ensure their clinical utility. This evolving landscape underscores the need for improvement of diagnostic approaches to support the expanding role of ADCs in BC treatment.
Collapse
Affiliation(s)
- Eleonora Nicolò
- Division of Hematology-Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Caterina Gianni
- Breast & GYN Unit, Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paolo Tarantino
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
3
|
Yeh R, Pareja F, Shobeiri P, Ross D, Jayaprakasam VS, Razmaria AA, Drago JZ, Mauguen A, Lyashchenko SK, Zeglis BM, Lewis JS, Ulaner GA. Detection of HER2-Low Lesions Using HER2-Targeted PET Imaging in Patients with Metastatic Breast Cancer: A Paired HER2 PET and Tumor Biopsy Analysis. J Nucl Med 2025:jnumed.124.269227. [PMID: 40341092 DOI: 10.2967/jnumed.124.269227] [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: 11/25/2024] [Accepted: 03/31/2025] [Indexed: 05/10/2025] Open
Abstract
Trastuzumab deruxtecan (T-DXd), a human epidermal growth factor receptor 2 (HER2)-targeted antibody-drug conjugate, demonstrated remarkable efficacy in previously treated patients with HER2-low metastatic breast cancer (mBC), marking a new therapeutic option for this patient population. Prior studies with HER2 PET using 89Zr-radiolabeled antibodies were limited by high rates of imaging false positives for HER2-positive malignancy. In this retrospective study, we investigate whether these false positives (HER2-negative on pathology) could be explained by HER2-low lesions. Methods: A retrospective study was conducted of mBC patients who previously enrolled in 2 prospective HER2 PET imaging trials: NCT02286843 using 89Zr-trastuzumab and 89Zr-pertuzumab and NCT04692831 using 89Zr-ss-pertuzumab. Patients were included if paired HER2 PET scan and biopsy were performed within a 2-mo period. Of 56 total patients, 23 patients met the inclusion criteria. Pathology results for biopsied lesions were collected, without repeat interpretation, and lesions were classified as HER2-positive, HER2-low, or HER2-0. SUVmax of biopsied lesions were compared between pathologic classifications to determine whether lesion uptake intensity could differentiate between HER2-positive and HER2-low lesions. Results: All prior false-positive lesions on HER2 PET scans from NCT02286843 were reclassified as HER2-low (instead of HER2-negative). In the 89Zr-trastuzumab cohort, 3 lesions were HER2-positive (33%) and 6 were HER2-low (67%); in the 89Zr-pertuzumab cohort, 2 were HER2-positive (29%) and 5 were HER2-low (71%). In the 89Zr-ss-pertuzumab cohort (NCT04692831), 7 patients underwent recent biopsies of 8 total lesions demonstrating 1 HER2-positive (12%), 5 HER2-low (62%), and 2 HER2-0 lesions (25%). HER2 PET SUVmax of biopsied lesions were compared between HER2-positive and HER2-low lesions for the combination of all 3 radiotracer cohorts. HER2-low lesions had a significantly higher SUVmax (median, 12.7; interquartile range, 8.05) than did HER2-positive lesions (median, 6.4; interquartile range, 1.98; P = 0.01). Conclusion: HER2 PET imaging with 89Zr-radiolabeled antibodies detects HER2-low lesions in addition to HER2-positive lesions in patients with mBC, suggesting its ability to visualize the entire spectrum of HER2 expression. All prior false positives on 89Zr-trastuzumab and 89Zr-pertuzumab PET scans were reclassified as HER2-low. Lesion SUVmax is not reliable in differentiating HER2-positive from HER2-low lesions; however, it may be useful in distinguishing lesions expressing HER2 from HER2-0 lesions.
Collapse
Affiliation(s)
- Randy Yeh
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Fresia Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Parnian Shobeiri
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dara Ross
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Vetri S Jayaprakasam
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Ali Aria Razmaria
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Joshua Z Drago
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Audrey Mauguen
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Brian M Zeglis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
- Department of Chemistry, Hunter College, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
- Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gary A Ulaner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, California; and
- Departments of Radiology and Translational Genomics, University of Southern California, Los Angeles, California
| |
Collapse
|
4
|
Kakouri A, Debnam MJ, Ferrarotto R, Lu T, Esmaeli B. Successful Neoadjuvant Treatment Using Trastuzumab Emtansine (Kadcyla) for Locally Advanced and Metastatic Lacrimal Gland Adenocarcinoma. Ophthalmic Plast Reconstr Surg 2025:00002341-990000000-00611. [PMID: 40334147 DOI: 10.1097/iop.0000000000002967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
A 25-year-old man presented with a locally advanced lacrimal gland adenocarcinoma with parotid metastasis. The lesion was strongly HER-2 positive (3+) on immunohistochemistry. The patient was treated with neoadjuvant trastuzumab emtansine, an antibody-drug conjugate targeting HER-2. The patient had an excellent response to neoadjuvant therapy trastuzumab emtansine, with a significant decrease in the size of the lacrimal gland carcinoma, making eye-sparing surgery a viable option for the patient. Furthermore, there was nearly complete resolution of the parotid metastasis; thus, radical neck dissection and parotidectomy were avoided.
Collapse
Affiliation(s)
- Agni Kakouri
- Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center
- Department of Ophthalmology and Visual Science, University of Texas at Houston, McGovern Medical School
| | | | - Renata Ferrarotto
- Department of Head and Neck and Thoracic Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, U.S.A
| | - Tracy Lu
- Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center
| | - Bita Esmaeli
- Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center
| |
Collapse
|
5
|
Li N, Yang L, Zhao Z, Du T, Liang G, Li N, Tang J. Antibody-drug conjugates in breast cancer: current evidence and future directions. Exp Hematol Oncol 2025; 14:41. [PMID: 40114224 PMCID: PMC11924693 DOI: 10.1186/s40164-025-00632-9] [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: 10/08/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are a rapidly evolving class of antitumor drugs and have already revolutionized the treatment strategy of many hematologic and solid cancers. So far, trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), sacituzumab govitecan (SG) and datopotamab deruxtecan (Dato-DXd) are the four ADCs that have been approved by US food and drug administration (FDA) in treatment of breast cancer, and SKB264 has been approved by Chinese national medical products administration (NMPA). Many ADCs for treatment of breast cancer are currently being tested in late-phase clinical trials, with several encouraging results achieved recently. However, major issues arise during the use of ADCs, including emergence of acquired resistance, occurrence of treated-related toxicities, and identification of biomarkers of response and resistance. ADCs are being increasingly tested in combination with other agents, and novel next-generation ADC development is progressing rapidly. A better understanding of the design and development of ADCs will promote ADC development for cancer treatment. In this review, we aim to provide a broad overview of the design and the recent advances of ADCs in breast cancer. We also propose several notable future directions of ADCs in treatment of breast cancer.
Collapse
Affiliation(s)
- Ning Li
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lu Yang
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
- Shantou University Medical College, Shantou University, Shantou, 515000, China
| | - Zixuan Zhao
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Tian Du
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Gehao Liang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Na Li
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Jun Tang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
6
|
Qin Q. Advances in research and current challenges in the treatment of advanced HER2-low breast cancer. Front Cell Dev Biol 2025; 13:1451471. [PMID: 40177129 PMCID: PMC11962219 DOI: 10.3389/fcell.2025.1451471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Human epidermal growth factor receptor 2 (HER2)-low breast cancer is defined as breast cancer with an immunohistochemistry (IHC) score of 1+ or 2+ and in situ hybridisation (ISH)-negative. The traditional HER2 classification (negative or positive) has limitations, with only 15%-20% of the breast cancer population being positive and suitable for HER2-targeted therapy. The new clinical study, DESTINY-Breast04, shows that trastuzumab deruxtecan (T-DXd) has a significant effect on advanced HER2-low breast cancers, a classification that accounts for approximately half of the advanced breast cancer population. However, the detection methods and evaluation criteria for HER2-low breast cancer have not yet been standardised, and the toxicity and resistance mechanisms associated with T-DXd therapy are still unclear. This article focuses on these issues and describes the progress and challenges of T-DXd-related therapy in the treatment of advanced breast cancer patients with low HER2 expression.
Collapse
Affiliation(s)
- Qiang Qin
- Breast and Thyroid Surgery Department, Nanning Maternal and Child Health Hospital, Nanning, China
| |
Collapse
|
7
|
Markides DM, Hita AG, Merlin J, Reyes-Gibby C, Yeung SCJ. Antibody-Drug Conjugates: The Toxicities and Adverse Effects That Emergency Physicians Must Know. Ann Emerg Med 2025; 85:214-229. [PMID: 39641680 DOI: 10.1016/j.annemergmed.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024]
Abstract
Antibody-drug conjugates are novel antineoplastic agents whose use is expanding, both in terms of the number of drugs and the number of patients being treated. This article reviews the known toxicities and complications of antibody-drug conjugates that are currently approved for the treatment of cancer in the United States, with a focus on their emergency presentation and management. Similar to many other cancer therapies, most antibody-drug conjugates can cause diarrhea, nausea/vomiting, rash, peripheral neuropathy, and cytopenia, which are generally treated following standard-of-care. Interstitial lung disease, which may mimic pneumonia and cause respiratory failure and death, has been seen with trastuzumab deruxtecan and mirvetuximab soravtansine; emergency treatment of this condition includes oxygenation, ventilatory support, and corticosteroids. Inotuzumab ozogamicin and gemtuzumab ozogamicin are both associated with sinusoidal obstruction syndrome, a potentially fatal liver dysfunction that presents with weight gain, fluid overload, and jaundice. Abnormal liver function tests in patients who have been recently treated with these agents should be cautiously evaluated. Cardiac adverse events with antibody-drug conjugates are rare, but trastuzumab emtansine and trastuzumab deruxtecan may cause a decrease in cardiac contractility, and heart rate corrected QT interval prolongation is a rare effect of trastuzumab deruxtecan. Ocular adverse events, especially blurred vision, and keratopathy, are common with mirvetuximab soravtansine and tisotumab vedotin. Progressive multifocal leukoencephalopathy has been reported with brentuximab vedotin and polatuzumab vedotin. Tumor lysis syndrome may occur after treatment with gemtuzumab ozogamicin, polatuzumab vedotin, and brentuximab vedotin. Patients receiving enfortumab vedotin or brentuximab vedotin may develop hyperglycemia, sometimes presenting as diabetic ketoacidosis. Tisotumab vedotin and trastuzumab emtansine are associated with bleeding; although it is minor in most cases, severe bleeding and intracranial hemorrhage have occurred. Several antibody-drug conjugates can cause an anaphylactoid infusion-related reaction, which occurs most commonly during or soon after infusion but may be delayed up to 24 hours. Further research is needed to establish the real-world incidence of rare complications and how often patients with these complications present to the emergency department.
Collapse
Affiliation(s)
| | - Angel Guido Hita
- Department of Emergency Medicine, MD Anderson Cancer Center, Houston, TX
| | - Jeffrey Merlin
- Department of Emergency Medicine, MD Anderson Cancer Center, Houston, TX
| | - Cielto Reyes-Gibby
- Department of Emergency Medicine, MD Anderson Cancer Center, Houston, TX
| | - Sai-Ching J Yeung
- Department of Emergency Medicine, MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
8
|
Zhou L, Zhang Y, Zhang J, Wang H, Zhao B, Cai Y, Qu Y, Li X, Zhang D. Clinical characteristics and therapeutic direction of HER2 low-expression breast cancer. Front Oncol 2025; 15:1484103. [PMID: 40083869 PMCID: PMC11903420 DOI: 10.3389/fonc.2025.1484103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/31/2025] [Indexed: 03/16/2025] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is one of the oncogenic drivers of breast cancer and is often used as a definitive therapeutic marker for breast cancer. This has led to significant improvements in both targeted therapy and prognosis for HER2-targeted breast cancer. Due to the differences in HER2 gene and protein expression levels, they are clinically classified into HER2 zero-expression breast cancer, low-expression breast cancer and high-expression breast cancer. Among them, HER2 low-expression is considered a special expression state, which is insensitive to conventional anti-HER2 therapy and has a poorer prognosis and thus has received attention from researchers. Some studies demonstrate that patients with HER2 low-expression can benefit from antibody-drug conjugates (ADC). Several studies are currently exploring the efficacy of various ADC drugs in breast cancer with HER2 low-expression, opening up new treatment avenues for patients with HER2 low-expression breast cancer. This review aims to summarize the clinical features of HER2 low-expression breast cancer and the recent advances in its therapeutic agents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dongwei Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
9
|
Nervig CS, Rice M, Marelli M, Christie RJ, Owen SC. Modular Synthesis of Anti-HER2 Dual-Drug Antibody-Drug Conjugates Demonstrating Improved Toxicity. Bioconjug Chem 2025; 36:190-202. [PMID: 39841105 DOI: 10.1021/acs.bioconjchem.4c00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Antibodies have gained clinical success in the last two decades for the targeted delivery of highly toxic small molecule chemotherapeutics. Yet antibody-drug conjugates (ADCs) often fail in the clinic due to the development of resistance. The delivery of two mechanistically distinct small molecule drugs on one antibody is of increasing interest to overcome these challenges with single-drug ADCs. We have developed a modular synthetic strategy for the construction of a library of 19 dual-drug ADCs where drugs are conjugated through unnatural cyclopentadiene-containing amino acids and native cysteine residues on an anti-HER2 trastuzumab scaffold. Importantly, this strategy utilizes the same functional group on the linker-drug construct; this allows for the facile addition of drugs at either conjugation site and enables the evaluation of different drug-to-antibody ratios and combinations of drug pairs. We tested the library on high- and mid-HER2 expressing cell lines and observed increased toxicity in several dual-drug ADCs compared with single-drug constructs. The strategy developed herein provides a method for the facile synthesis, characterization, and evaluation of dual-payload ADCs. Simultaneous delivery of combinations of drugs with distinct mechanisms of action is critical for the next generation of targeted drug delivery.
Collapse
Affiliation(s)
- Christine S Nervig
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112 United States
| | - Megan Rice
- Biologics Engineering, AstraZeneca Oncology R&D, One MedImmune Way, Gaithersburg, Maryland 20878 United States
| | - Marcello Marelli
- Biologics Engineering, AstraZeneca Oncology R&D, One MedImmune Way, Gaithersburg, Maryland 20878 United States
| | - R James Christie
- Biologics Engineering, AstraZeneca Oncology R&D, One MedImmune Way, Gaithersburg, Maryland 20878 United States
| | - Shawn C Owen
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112 United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Biomedical Engineering, University of Utah Salt Lake City, Utah 84112, United States
| |
Collapse
|
10
|
Caltavituro A, Buonaiuto R, Salomone F, Pecoraro G, Martorana F, Lauro VD, Barchiesi G, Puglisi F, Del Mastro L, Montemurro F, Giuliano M, Arpino G, De Laurentiis M. Warming-up the immune cell engagers (ICEs) era in breast cancer: state of the art and future directions. Crit Rev Oncol Hematol 2025; 206:104577. [PMID: 39613237 DOI: 10.1016/j.critrevonc.2024.104577] [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: 07/22/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) has deeply reshaped the therapeutic algorithm of triple-negative breast cancer (TNBC). However, there is considerable scope for better engagement of the immune system in other BC subtypes. ICIs have paved the way for investigations into emerging immunotherapeutic strategies, such as immune cell engagers (ICEs) that work by promoting efficient tumor cell killing through the redirection of immune system against cancer cells. Most ICEs are bispecific antibodies that simultaneously recognize and bind to both cancer and immune cells generating an artificial synapse. Major side effects are cytokine release syndrome, hepatotoxicity, and neurotoxicity related to inappropriate immune system activation. Here, we provide a comprehensive overview of this compounds, the available preclinical and clinical evidence supporting their investigation and development in BC also highlighting the challenges that have prevented their widespread use in oncology. Finally, major strategies are explored to broaden their use in BC.
Collapse
Affiliation(s)
- Aldo Caltavituro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy; Clinical and Translational Oncology, Scuola Superiore Meridionale, Naples, Italy
| | - Roberto Buonaiuto
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy; Clinical and Translational Oncology, Scuola Superiore Meridionale, Naples, Italy
| | - Fabio Salomone
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy
| | - Giovanna Pecoraro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy
| | - Federica Martorana
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - Vincenzo Di Lauro
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy.
| | - Giacomo Barchiesi
- Azienda Ospedaliera Universitaria Policlinico Umberto I, UOC Oncologia, Roma, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Via Palladio 8, Udine 33100, Italy; Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Via Franco Gallini 2, Aviano, Pordenone 33081, Italy
| | - Lucia Del Mastro
- Department of Medical Oncology, UO Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, Genova 16132, Italy
| | - Filippo Montemurro
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 -KM 3.95, Candiolo, Torino 10060, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy
| | - Michelino De Laurentiis
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| |
Collapse
|
11
|
Li S, Zhao X, Fu K, Zhu S, Pan C, Yang C, Wang F, To KK, Fu L. Resistance to antibody-drug conjugates: A review. Acta Pharm Sin B 2025; 15:737-756. [PMID: 40177568 PMCID: PMC11959940 DOI: 10.1016/j.apsb.2024.12.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 04/05/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are antitumor drugs composed of monoclonal antibodies and cytotoxic payload covalently coupled by a linker. Currently, 15 ADCs have been clinically approved worldwide. More than 100 clinical trials at different phases are underway to investigate the newly developed ADCs. ADCs represent one of the fastest growing classes of targeted antitumor drugs in oncology drug development. It takes advantage of the specific targeting of tumor-specific antigen by antibodies to deliver cytotoxic chemotherapeutic drugs precisely to tumor cells, thereby producing promising antitumor efficacy and favorable adverse effect profiles. However, emergence of drug resistance has severely hindered the clinical efficacy of ADCs. In this review, we introduce the structure and mechanism of ADCs, describe the development of ADCs, summarized the latest research about the mechanisms of ADC resistance, discussed the strategies to overcome ADCs resistance, and predicted biomarkers for treatment response to ADC, aiming to contribute to the development of ADCs in the future.
Collapse
Affiliation(s)
- Sijia Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xinyu Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shuangli Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kenneth K.W. To
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong 999077, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
12
|
Liao S, Li X, Lu Y, Luo K. Nanomedicine in Immunotherapy for Non-Small Cell Lung Cancer: Applications and Perspectives. SMALL METHODS 2025:e2401783. [PMID: 39871783 DOI: 10.1002/smtd.202401783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/17/2025] [Indexed: 01/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) has a strikingly high incidence rate globally. Although immunotherapy brings a great breakthrough in its clinical treatment of NSCLC, significant challenges still need to be overcome. The development of novel multi-functional nanomedicines in the realm of tumor immunotherapy offers promising opportunities for NSCLC patients, as nanomedicines exhibit significant advantages, including specific targeting of tumor cells, improved drug bioavailability, reduced systemic toxicity, and overcoming of immune resistance. In this review, the core features and current clinical status of strategies for NSCLC immunotherapy including immune checkpoint blockade, antibody-drug conjugates, cell engagers, adoptive cells, and cancer vaccines, are surveyed. Particular emphasis is placed on the recent development of nanomedicines that boost these strategies. Nanomedicine can provide novel perspectives for NSCLC immunotherapy.
Collapse
Affiliation(s)
- Shuangsi Liao
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoling Li
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
13
|
Pareeth F, Babu G, Mathew BS. Trastuzumab emtansine induced hyponatremia in breast cancer - A case report. J Cancer Res Ther 2025; 21:240-242. [PMID: 39207114 DOI: 10.4103/jcrt.jcrt_1602_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/04/2023] [Indexed: 09/04/2024]
Abstract
ABSTRACT Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate of trastuzumab and the cytotoxic agent emtansine (DM1), approved for use as an adjuvant treatment for patients with residual disease after neoadjuvant chemotherapy and antihuman epidermal growth factor receptor 2 (HER-2) therapy and in metastatic HER-2-positive breast cancer. Previous studies have shown that T-DM1 has a favorable safety profile, with few high-grade toxicities reported so far. We describe a patient who developed profound hyponatremia-which has not been reported previously-following treatment with adjuvant T-DM1 for HER-2+ breast cancer.
Collapse
Affiliation(s)
- Firoz Pareeth
- Department of Radiation Oncology, Regional Cancer Centre, Thiruvananthapuram, Kerala, India
| | | | | |
Collapse
|
14
|
Önder T, Karaçin C. Trastuzumab-induced optic neuritis: "blindness" side effect. J Oncol Pharm Pract 2025; 31:164-167. [PMID: 39140810 DOI: 10.1177/10781552241275538] [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] [Indexed: 08/15/2024]
Abstract
INTRODUCTION Trastuzumab improved the prognosis of patients with human epidermal growth factor receptor 2 (HER2)+ breast cancer (BC). Here, we present a patient who developed acute vision loss due to optic atrophy in both eyes after trastuzumab. CASE REPORT A 60-year-old female patient was diagnosed with locally advanced HER2+ BC in January 2021. After four cycles of neoadjuvant anthracycline-based chemotherapy followed by four cycles of docetaxel, trastuzumab, and pertuzumab combined treatment, the patient underwent a right modified radical mastectomy. Three days after the end of the second cycle of adjuvant trastuzumab, she presented with acute vision loss. The patient's visual acuity was 90% in the right eye and 60% in the left eye. The left eye had optic nerve edema and spindle hemorrhages. First, on suspicion of optic neuritis, the patient was given a 1 gram/day pulse steroid for three days. However, optic neuritis was not considered during the follow-up. Metastasis was considered at the exit of the left optic nerve. Trastuzumab was started by making a mutual decision with the patient. Six days after the sixth dose of adjuvant trastuzumab, she presented with almost complete vision loss. MANAGEMENT AND OUTCOME The patient was diagnosed with optic neuritis, and a pulse steroid was administered. Trastuzumab was permanently discontinued. However, the patient's visual acuity in both eyes remained at 5-10%. DISCUSSION Vision loss due to optic neuritis is a devastating side effect. Understanding that trastuzumab-induced optic neuritis may develop will help clinicians detect side effects early and manage them more effectively.
Collapse
Affiliation(s)
- Tuğba Önder
- Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Cengiz Karaçin
- Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Health Sciences University, Ankara, Turkey
| |
Collapse
|
15
|
Tang SC, Wynn C, Le T, McCandless M, Zhang Y, Patel R, Maihle N, Hillegass W. Influence of antibody-drug conjugate cleavability, drug-to-antibody ratio, and free payload concentration on systemic toxicities: A systematic review and meta-analysis. Cancer Metastasis Rev 2024; 44:18. [PMID: 39704752 PMCID: PMC11662062 DOI: 10.1007/s10555-024-10231-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024]
Abstract
While in theory antibody drug conjugates (ADCs) deliver high-dose chemotherapy directly to target cells, numerous side effects are observed in clinical practice. We sought to determine the effect of linker design (cleavable versus non-cleavable), drug-to-antibody ratio (DAR), and free payload concentration on systemic toxicity. Two systematic reviews were performed via PubMed search of clinical trials published between January 1998-July 2022. Eligible studies: (1) clinical trial for cancer therapy in adults, (2) ≥ 1 study arm included a single-agent ADC, (3) ADC used was commercially available/FDA-approved. Data was extracted and pooled using generalized linear mixed effects logistic models. 40 clinical trials involving 7,879 patients from 11 ADCs, including 9 ADCs with cleavable linkers (N = 2,985) and 2 with non-cleavable linkers (N = 4,894), were included. Significantly more composite adverse events (AEs) ≥ grade 3 occurred in patients in the cleavable linkers arm (47%) compared with the non-cleavable arm (34%). When adjusted for DAR, for grade ≥ 3 toxicities, non-cleavable linkers remained independently associated with lower toxicity for any AE (p = 0.002). Higher DAR was significantly associated with higher probability of grade ≥ 3 toxicity for any AE. There was also a significant interaction between cleavability status and DAR for any AE (p = 0.002). Finally, higher measured systemic free payload concentrations were significantly associated with higher DARs (p = 0.043). Our results support the hypothesis that ADCs with cleavable linkers result in premature payload release, leading to increased systemic free payload concentrations and associated toxicities. This may help to inform future ADC design and rational clinical application.
Collapse
Affiliation(s)
- Shou-Ching Tang
- LSU-LCMC Cancer Center, Louisiana State University (LSU) Health Sciences Center, New Orleans, LA, U.S.A..
| | - Carrie Wynn
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, U.S.A
| | - Tran Le
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, U.S.A
| | - Martin McCandless
- Department of Neurological Surgery, Medical Center, University of Kansas, Kansas City, KS, U.S.A
| | - Yunxi Zhang
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, U.S.A
| | - Ritesh Patel
- Trinity Health Ann Arbor Hospital, Ann Arbor, MI, U.S.A
| | - Nita Maihle
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, U.S.A
| | - William Hillegass
- Departments of Data Science and Medicine, University of Mississippi Medical Center, Jackson, MS, U.S.A
| |
Collapse
|
16
|
Chen H, He G, Huang J, Hu L, Ma J. Ocular adverse events associated with antibody-drug conjugates: a comprehensive pharmacovigilance analysis. Front Immunol 2024; 15:1495137. [PMID: 39742259 PMCID: PMC11685049 DOI: 10.3389/fimmu.2024.1495137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/03/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Antibody-drug conjugates (ADCs) are increasingly utilized in patients with solid tumors and hematologic malignancies. However, the adverse ocular toxicity induced by ADCs has not been comprehensively evaluated in real-world clinical settings. Methods Data from April 2019 to March 2024 based on the FDA Adverse Event Reporting System (FAERS) were extracted and analyzed. Disproportionality analysis was used to evaluate the association between ADCs and ocular adverse events (AEs). The median time to onset (TTO) of various ADCs was compared. Results A comprehensive analysis identified 2,686 ocular AEs associated with ADCs. Among these, Tisotumab vedotin had the most positive signals at the preferred terms (PTs) level, followed by trastuzumab emtansine and enfortumab vedotin. In contrast, gemtuzumab ozogamicin demonstrated minimal ocular toxicity signals. Cluster analysis revealed that ADC-related ocular toxicities predominantly manifested as corneal disorders or ocular neuromuscular disorders. The median onset of ocular toxicity varied considerably, with enfortumab vedotin showing the earliest median onset at 12.5 days. Conclusions Our study demonstrates the association between ADCs and ocular AEs based on real-world data, providing valuable guidance for clinicians when prescribing ADCs. And we found some important safety signals that have not been mentioned in the label or previous studies.
Collapse
Affiliation(s)
- Heng Chen
- Department of Pharmacy, The First Hospital of Changsha, Changsha, Hunan, China
| | - Gefei He
- Department of Pharmacy, The First Hospital of Changsha, Changsha, Hunan, China
| | - Juanjuan Huang
- Department of Pharmacy, The First Hospital of Changsha, Changsha, Hunan, China
| | - Lin Hu
- Department of Pharmacy, The First Hospital of Changsha, Changsha, Hunan, China
| | - Junlong Ma
- Center of Clinical Pharmacology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
17
|
Willis C, Tan CJ, Chhibber A, Watanabe AH, Lam C, Mehta S, Kwong J, Park L, Pavilack-Kirker M, Xu X, Kelley K, Stenehjem D. Patient characteristics and treatment patterns of patients with locally advanced or metastatic HER2-low breast cancer, a single site descriptive study. Breast Cancer Res Treat 2024; 208:619-630. [PMID: 39172305 PMCID: PMC11522147 DOI: 10.1007/s10549-024-07458-9] [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: 05/09/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
PURPOSE To evaluate the prevalence and characteristics of different HER2 categories among patients with advanced breast cancer (aBC) and describe treatment patterns and outcomes of those with HER2-low disease. METHODS A retrospective cohort study was conducted via chart review at the Huntsman Cancer Institute, including patients diagnosed with aBC (stages IIIB, IIIC and IV) between 2010 and 2019. All patients with IHC1+ were considered HER2-low unless FISH was positive. Patients with IHC2+ were only classified as HER2-low if a negative FISH was documented. The prevalence and characteristics of each HER2 category were reported. Treatment patterns and survival outcomes of HER2-low patients who received first line treatment in 2017 or later were presented. RESULTS A total of 240 of 414 patients (58%) with aBC were HER2-low, with the majority of patients (83%) classified as hormone receptor (HR)-positive. In first line, most HR-positive patients received endocrine therapy with chemotherapy for stage IIIB/IIIC (47%) and with CDK4/6 inhibitors for stage IV breast cancer (50%) Most HR-negative patients received chemotherapy alone (92% for stage IIIB/IIIC, 60% for stage IV). In second line, chemotherapy alone was the most common modality (21.4% for HR-positive; 45.5% for HR-negative). Median overall survival was 37.7 months while median progression-free survival from first line was 18.0 months, decreasing to 8.0 months in second line. CONCLUSION A substantial proportion of patients previously classified as HER2-negative have low but detectable HER2 expression and may benefit from novel HER2-directed agents, which have demonstrated clinical benefit in this population post-chemotherapy.
Collapse
Affiliation(s)
- Connor Willis
- Pharmacotherapy Outcomes Research Center, Department of Pharmacotherapy, College of Pharmacy, Salt Lake City, UT, USA
| | - Chia Jie Tan
- Pharmacotherapy Outcomes Research Center, Department of Pharmacotherapy, College of Pharmacy, Salt Lake City, UT, USA
| | - Anindit Chhibber
- Pharmacotherapy Outcomes Research Center, Department of Pharmacotherapy, College of Pharmacy, Salt Lake City, UT, USA
| | - Alexandre H Watanabe
- Pharmacotherapy Outcomes Research Center, Department of Pharmacotherapy, College of Pharmacy, Salt Lake City, UT, USA
| | | | | | | | | | | | | | | | - David Stenehjem
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, MN, USA.
| |
Collapse
|
18
|
Hu S, Zhao Y, Xie Y, You S, Hu X, Zhang J, Wang L, Cao J, Gong C, Wang B. Clinicopathological characteristics, evolution, and treatment outcomes of hormone receptor-negative/HER2-low metastatic breast cancer: a pooled analysis of individual patient data from three prospective clinical trials. Front Endocrinol (Lausanne) 2024; 15:1449278. [PMID: 39640887 PMCID: PMC11617163 DOI: 10.3389/fendo.2024.1449278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Objective With the approval of trastuzumab deruxtecan for the treatment of unresectable/metastatic HER2-low breast cancer, human epidermal growth factor receptor 2 (HER2)-low has emerged as a clinically actionable biomarker. There is an urgent need for a deeper understanding of HER2-low breast cancer patients. Therefore, this study was conducted to explore the clinicopathological characteristics, the evolution of HER2-low status, and its impact on the prognosis of hormone receptor (HoR)-negative/HER2-low metastatic breast cancer (MBC) patients. Methods This pooled analysis included 350 metastatic triple-negative breast cancer (mTNBC) patients who received first-line platinum-based chemotherapy at Fudan University Shanghai Cancer Center from November 2007 to July 2022. Patients were categorized into HER2-0 and HER2-low groups based on their HER2 status. Baseline clinicopathological characteristics, evolution of HER2 status between primary and metastatic lesions, and treatment efficacy were compared between the two groups. Results Among the 350 mTNBC patients, 34.9% (122/350) were HER2-low and 65.1% (228/350) were HER2-0. Significant differences were observed between HER2-low and HER2-0 patients in terms of age and menopausal status. HER2-low patients were older (54 vs. 49 years, p=0.002) and had a lower proportion of premenopausal patients (32.8% vs. 52.6%, p<0.001) compared to HER2-0 patients. No significant differences were observed in progression-free survival (PFS) and overall survival (OS) between HER2-low and HER2-0 patients receiving first-line platinum-based chemotherapy (mPFS: 7.43 vs. 8.30 months, p=0.389, HR=1.11, 95% CI 0.88-1.40; mOS: 25.37 vs. 26.63 months, p=0.907, HR=1.02, 95% CI 0.76-1.37). Additionally, 32.3% (41/127) of patients exhibited discordant HER2 status between primary and metastatic lesions, primarily evolving from HER2-0 to HER2-low. Notably, patients with discordant HER2 status had significantly longer PFS compared to those with concordant status (mPFS: 11.07 vs. 7.53 months, p=0.020). The Cox multivariate analysis showed that HER2 status consistency (p=0.026) was an independent predictor of PFS. Conclusion In mTNBC patients, those with HER2-low status had similar responses to platinum-based chemotherapy as HER2-0 patients. There was significant discordance in HER2 status between primary and metastatic lesions. Patients with discordant HER2 status had better responses to platinum-based chemotherapy. Therefore, for patients with HER2-0 primary lesions, re-evaluation of HER2 status in metastatic lesions through biopsy may offer new treatment opportunities.
Collapse
Affiliation(s)
- Shihui Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yannan Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizhao Xie
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuhui You
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Leiping Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chengcheng Gong
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Biyun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Michelon I, Dacoregio MI, Vilbert M, Priantti J, do Rego Castro CE, Vian L, Tarantino P, de Azambuja E, Cavalcante L. Antibody-drug conjugates in patients with advanced/metastatic HER2-low-expressing breast cancer: a systematic review and meta-analysis. Ther Adv Med Oncol 2024; 16:17588359241297079. [PMID: 39574495 PMCID: PMC11580099 DOI: 10.1177/17588359241297079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024] Open
Abstract
Background Until recently, targeted therapies have failed to benefit patients with human epidermal growth factor receptor 2 (HER2)-low-expressing breast cancer (BC). Nevertheless, antibody-drug conjugates (ADCs) have reshaped their prognosis. Objectives We performed a systematic review and meta-analysis to assess the effectiveness of ADCs in patients with HER2-low advanced/metastatic (a/m) BC. Design This study is a systematic review and meta-analysis. Data sources We searched PubMed, Embase, and Cochrane databases as well as the American Society of Clinical Oncology, European Society for Medical Oncology, and San Antonio Breast Cancer Symposium conference proceedings. Methods Studies evaluating ADCs (trastuzumab deruxtecan (T-DXd), sacituzumab govitecan (SG), MRG002, and RC48-ADC) in patients with HER2-low a/mBC were included. We used R software (v.4.2.2) and random effects models for all analyses. Heterogeneity was assessed using the I 2 test. Results Overall, 14 studies were included (five real-world studies and nine clinical trials (CTs)), with 2883 HER2-low a/mBC patients: 808 received treatment of physician's choice (TPC), and 2075 ADCs. Most were treated with T-DXd (n = 1691), followed by SG (n = 310), MRG002 (n = 56), and RC48-ADC (n = 18). Patients treated with T-DXd achieved a significantly higher objective response rate (ORR), disease control rate (DCR), and clinical benefit rate (CBR) than those receiving other ADCs. In the pooled analysis of four randomized CTs, ADCs statistically prolonged progression-free survival (n = 1828, hazard ratio (HR) 0.50, 95% confidence interval (CI) 0.36-0.68, I 2 = 82%, p < 0.001) and overall survival (n = 1546, HR 0.70, 95% CI 0.57-0.86, I 2 = 43%, p < 0.001) compared with TPC. Patients on ADCs also achieved a greater antitumor response than TPC, including better ORR (odds ratio (OR), 3.7, 95% CI 2.5-5.6, I 2 = 59%, p < 0.001), DCR (OR, 2.7, 95% CI 2.1-3.5, I 2 = 0%, p < 0.001), and CBR (OR, 3.6, 95% CI 2.6-5.2, I 2 = 56%, p < 0.01). Conclusion Our systematic review and meta-analysis confirms the efficacy of ADCs in HER2-low a/m BC patients over TPC. Future studies should focus on bringing ADCs into earlier lines of therapy in this population. Trial registration This study was registered in PROSPERO (CRD42024452962).
Collapse
Affiliation(s)
- Isabella Michelon
- Department of Medicine, Catholic University of Pelotas, 373 Gonçalves Chaves, Pelotas 96010-000, Brazil
| | | | - Maysa Vilbert
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan Priantti
- School of Medicine, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | | | - Lucas Vian
- Rede AMO de Campo Grande, Divisão de Oncologia, Departamento do Hospital CASSEMS de Campo Grande, Campo Grande, Mato Grosso do Sul, Brazil
| | - Paolo Tarantino
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Evandro de Azambuja
- Institut Jules Bordet, Hopital Universitaire de Bruxelles and l’Université Libre de Bruxelles, Brussels, Belgium
| | - Ludimila Cavalcante
- Department of Medical Oncology and Hematology, University of Virginia Comprehensive Cancer Center, Charlottesville, VA, USA
| |
Collapse
|
20
|
Colombo R, Tarantino P, Rich JR, LoRusso PM, de Vries EGE. The Journey of Antibody-Drug Conjugates: Lessons Learned from 40 Years of Development. Cancer Discov 2024; 14:2089-2108. [PMID: 39439290 DOI: 10.1158/2159-8290.cd-24-0708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/16/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024]
Abstract
Antibody-drug conjugates (ADC) represent one of the most rapidly expanding treatment modalities in oncology, with 11 ADCs approved by the FDA and more than 210 currently being tested in clinical trials. Spanning over 40 years, ADC clinical development has enhanced our understanding of the multifaceted mechanisms of action for this class of therapeutics. In this article, we discuss key insights into the toxicity, efficacy, stability, distribution, and fate of ADCs. Furthermore, we highlight ongoing challenges related to their clinical optimization, the development of rational sequencing strategies, and the identification of predictive biomarkers. Significance: The development and utilization of ADCs have allowed for relevant improvements in the prognosis of multiple cancer types. Concomitantly, the rise of ADCs in oncology has produced several challenges, including the prediction of their activity, their utilization in sequence, and minimization of their side effects, that still too often resemble those of the cytotoxic molecule that they carry. In this review, we retrace 40 years of development in the field of ADCs and delve deep into the mechanisms of action of these complex therapeutics and reasons behind the many achievements and failures observed in the field to date.
Collapse
Affiliation(s)
| | - Paolo Tarantino
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Jamie R Rich
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, Canada
| | - Patricia M LoRusso
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
21
|
Pous A, Bernat-Peguera A, López-Paradís A, Cirauqui B, Quiroga V, Teruel I, Felip E, Ferrando-Díez A, Bergamino M, Boronat L, Romeo M, Soler G, Mariño C, Rodríguez-Martínez P, Pons L, Ballana E, Martinez-Cardús A, Margelí M. Deciphering HER2-low breast cancer (BC): insights from real-world data in early stage breast cancer. Ther Adv Med Oncol 2024; 16:17588359241290720. [PMID: 39449733 PMCID: PMC11500235 DOI: 10.1177/17588359241290720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Background Human epidermal growth factor receptor 2 (HER2)-low has emerged as a potential new entity in breast cancer (BC). Data on this subset are limited, and prognostic results are controversial, evidencing the need of further data in a BC real-world cohort. Methods Patients with HER2-negative stage I-III BC diagnosed between 2006 and 2016 were retrospectively reviewed in a single cohort from the Catalan Institute of Oncology Badalona. Demographics and clinicopathological characteristics were examined via medical charts/electronic health records. We aim to describe and compare HER2-0/HER2-low populations through Chi-square or Fisher test, and explore its prognostic impact using Kaplan-Meier curves and Cox regression models. Results From a cohort of 1755 BC patients, 1401 invasive HER2-negative, stage I-III cases were evaluated. 87% were hormone receptor (HR)-positive versus 13% triple negative (TNBC). Overall, 43% were HER2-0 and 57% HER2-low (61% immunohistochemistry (IHC) 1+ and 39% IHC 2+). Comparing HER2-low versus HER2-0, HER2-low showed higher proportion of estrogen receptor (ER)-positive (91.6% vs 79.9%, p ⩽ 0.001) and progesterone receptor (PR)-positive (79.8% vs 68.9%, p ⩽ 0.001) cases. HER2-0 exhibited higher proportion of TNBC (20.1% vs 8.4%, p = 0.001), grade III tumors (28.8% vs 23.5%, p = 0.039), and higher Ki67 median value (26.47% vs 23.88%, p = 0.041). HER2-low was associated with longer time to distant recurrence (TTDR) compared to HER2-0 (67.8 vs 54.1 months; p = 0.015) and better BC-related survival (19.2 vs 16.3 years; p = 0.033). In the multivariable analysis, HER2-low was not an independent prognostic factor for TTDR and BC-related survival. ER expression showed a strong association with longer TTDR (Hazard Ratio: 0.425, p ⩽ 0.001) and improved BC-related survival (Hazard Ratio: 0.380, p ⩽ 0.001). PR expression was also associated with longer TTDR (Hazard Ratio: 0.496, p ⩽ 0.001), and improved BC-related survival (Hazard Ratio: 0.488, p ⩽ 0.001). Histological grade III was significantly associated with shorter TTDR (Hazard Ratio: 1.737, p = 0.002). Positive nodal status was the strongest factor correlated with worse BC-related survival (Hazard Ratio: 2.747, p ⩽ 0.001). Conclusion HER2-low was significantly associated with HR-positive disease, whereas HER2-0 group had higher incidence of TNBC, histological grade III and higher Ki67%. Although HER2-low group was associated with longer TTDR and improved BC-related survival, these findings could be explained by the greater proportion of favorable prognostic features in this subgroup compared to HER2-0.
Collapse
Affiliation(s)
- Anna Pous
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Departament de Medicina, Campus Can Ruti, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Adrià Bernat-Peguera
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program, Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Assumpció López-Paradís
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Beatriz Cirauqui
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Vanesa Quiroga
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Iris Teruel
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Eudald Felip
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program, Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- IrsiCaixa, Barcelona, Spain
| | - Angelica Ferrando-Díez
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Milana Bergamino
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Laia Boronat
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Margarita Romeo
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Gemma Soler
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Christian Mariño
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
| | - Paula Rodríguez-Martínez
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Department of Pathology; Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
| | - Laura Pons
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Department of Pathology; Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
| | - Ester Ballana
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- IrsiCaixa, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, CIBERINFEC
| | - Anna Martinez-Cardús
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Mireia Margelí
- Catalan Institute of Oncology (ICO)-Badalona, Germans Trias i Pujol Universitary Hospital, Barcelona, Spain
- Badalona-Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
- Translational Program in Cancer Research (CARE) Program; Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| |
Collapse
|
22
|
Mukherjee A, Bandyopadhyay D. Targeted Therapy in Breast Cancer: Advantages and Advancements of Antibody-Drug Conjugates, a Type of Chemo-Biologic Hybrid Drugs. Cancers (Basel) 2024; 16:3517. [PMID: 39456611 PMCID: PMC11505910 DOI: 10.3390/cancers16203517] [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: 08/25/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Cancer is a significant health challenge globally, with millions of people affected every year, resulting in high morbidity and mortality. Although other treatment options are available with limitations, chemotherapy, either standalone or combined with other therapeutic procedures, is the most commonly used practice of treating cancer. In chemotherapy, cancer cells/malignant tumors are targeted; however, due to less target specificity, along with malignant cells, normal cells are also affected, which leads to various off-target effects (side effects) that impact the patient quality of life. Out of all the different types of cancers, breast cancer is the most common type of cancer in humans worldwide. Current anticancer drug discovery research aims to develop therapeutics with higher potency and lower toxicity, which is only possible through target-specific therapy. Antibody-drug conjugates (ADCs) are explicitly designed to target malignant tumors and minimize off-target effects by reducing systemic cytotoxicity. Several ADCs have been approved for clinical use and have shown moderate to good efficacy so far. Considering various aspects, chemotherapy and ADCs are useful in treating cancer. However, ADCs provide a more focused and less toxic approach, which is especially helpful in cases where resistance to chemotherapy (drug resistance) occurs and in the type of malignancies in which specific antigens are overexpressed. Ongoing ADC research aims to develop more target-specific cancer treatments. In short, this study presents a concise overview of ADCs specific to breast cancer treatment. This study provides insight into the classifications, mechanisms of action, structural aspects, and clinical trial phases (current status) of these chemo-biologic drugs (ADCs).
Collapse
Affiliation(s)
- Attrayo Mukherjee
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Patia, Bhubaneswar 751024, Odisha, India;
| | - Debasish Bandyopadhyay
- School of Integrative Biological and Chemical Sciences (SIBCS), University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- School of Earth, Environmental, and Marine Sciences (SEEMS), University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
23
|
Nader-Marta G, Singer C, Hlauschek D, DeMichele A, Tarantino P, de Azambuja E, Pfeiler G, Martin M, Balko JM, Nowecki Z, Balic M, Brufsky AM, Chan A, Morris PG, Haddad T, Loibl S, Liu Y, Soelkner L, Fesl C, Mayer EL, Gnant M. Clinical characterization, prognostic, and predictive values of HER2-low in patients with early breast cancer in the PALLAS trial (ABCSG-42/AFT-05/BIG-14-13/PrE0109). Breast Cancer Res 2024; 26:140. [PMID: 39375745 PMCID: PMC11459983 DOI: 10.1186/s13058-024-01899-2] [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: 05/15/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Bidirectional crosstalk between HER2 and estrogen receptor (ER) pathways may influence outcomes and the efficacy of endocrine therapy (ET). Low HER2 expression levels (HER2-low) have emerged as a predictive biomarker in patients with breast cancer (BC). METHODS PALLAS is an open, international, phase 3 study evaluating the addition of palbociclib for 2 years to adjuvant ET in patients with stage II-III ER-positive/HER2-negative BC. To assess the impact of HER2 expression on patient outcomes in the phase III PALLAS trial, we analyzed (1) the association between rate of HER2-low with demographic and clinicopathological parameters, (2) the prognostic value of HER2-low status on invasive disease-free survival (iDFS), distant relapse-free survival (DRFS), and overall survival (OS) and (3) HER2 expression's value as a predictive biomarker of response to palbociclib. HER2-low was defined as HER2 immunohistochemistry (IHC) 1 + or IHC 2 + with negative in situ hybridization (ISH). All pathologic evaluation was performed locally. Prognostic and predictive power of HER2 were assessed with Cox models. RESULTS From the original PALLAS intention-to-treat population (N = 5753), 5304 patients (92.2%) were included in this analysis. Among these, 2254 patients (42.5%) were classified as having HER2 IHC 0 (HER2-0), and 3050 (57.5%) as having HER2-low disease (1838 with IHC 1 + and 1212 with IHC 2 +). Median follow-up was 59.8 months. HER2-low prevalence varied significantly across 21 participating countries (range 16.7% to 75.6%; p < 0.001) and was more frequent in patients enrolled in North America (63.1%) than in Europe (53.4%) or other regions (53.4%) (p < 0.001). HER2 status was not significantly associated with iDFS in a multivariable Cox model (hazard ratio 0.93, 95% confidence interval 0.81 - 1.06). No significant interaction was observed between treatment arm and HER2 status for iDFS (p = 0.43). Similar results were obtained for DRFS and OS. CONCLUSIONS In this large, prospective, global patient cohort, no differences were observed in clinical parameters, prognosis, or differential benefit from palbociclib between HER2-0 and HER2-low tumors. Significant geographic variability was observed in the prevalence of HER2-low status, suggesting a high degree of variation in pathologic assessment of HER2 expression without impact on outcomes.
Collapse
Affiliation(s)
- Guilherme Nader-Marta
- Institut Jules Bordet, Academic Trials Promoting Team (ATPT), Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Christian Singer
- Department of Obstetrics and Gynaecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Dominik Hlauschek
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Angela DeMichele
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Evandro de Azambuja
- Institut Jules Bordet, Academic Trials Promoting Team (ATPT), Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Georg Pfeiler
- Department of Obstetrics and Gynaecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Miguel Martin
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Justin M Balko
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zbigniew Nowecki
- The Maria Sklodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Marija Balic
- Division of Oncology, Department of Internal Medicine, Medical University Graz, Graz, Austria
- University of Pittsburgh Hillman Cancer Center, Magee-Women's Hospital, Pittsburgh, PA, USA
| | - Adam M Brufsky
- University of Pittsburgh Hillman Cancer Center, Magee-Women's Hospital, Pittsburgh, PA, USA
| | - Arlene Chan
- Breast Cancer Research Centre-WA & Curtin University, Perth, Australia
| | - Patrick G Morris
- Cancer Trials Ireland, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Tufia Haddad
- Mayo Clinic Comprehensive Cancer Center, Rochester, MN, USA
| | - Sibylle Loibl
- German Breast Group, Prof. (Apl), Goethe University Frankfurt, Frankfurt am Main, Germany
- Clinical Consultant Centre for Haematology and Oncology/Bethanien, Frankfurt, Germany
| | - Yuan Liu
- Translational Oncology Global Product Development Pfizer Inc, San Diego, CA, USA
| | - Lidija Soelkner
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Christian Fesl
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Erica L Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael Gnant
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
24
|
Zalaquett Z, Rita Hachem MC, Assi A, Mohanna R, Farhat M, Noujaim C, Kourie HR. Cardiac toxicity of HER-2 targeting antibody-drug conjugates: overview and clinical implications. Future Oncol 2024; 20:3151-3167. [PMID: 39373602 DOI: 10.1080/14796694.2024.2407756] [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: 07/22/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have recently emerged as a promising therapeutic option that combine the specificity of monoclonal antibodies and the cytotoxic effect of chemotherapy. With numerous ADCs approved and on the market, a particular concern of ADCs that target HER-2 has been their cardiac side effects, in view of the crucial role of HER-2 in cardiac development and physiology. While rarely toxic and generally safe, numerous publications have outlined the consistent association of trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd) with the development of cardiac toxicity. Despite not being clinically relevant in most cases, cardiac baseline evaluation, monitoring and early detection of cardiac adverse events remain pivotal with HER-2 targeting ADCs. This review aims to summarize and better characterize the complete cardiac toxicity profile of HER-2 ADCs, with the goal of improving clinical understanding of this adverse event, leading to better recognition, monitoring and management.
Collapse
Affiliation(s)
- Ziad Zalaquett
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | | | - Ahmad Assi
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Rami Mohanna
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Mohamad Farhat
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | | | - Hampig-Raphael Kourie
- Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| |
Collapse
|
25
|
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
|
26
|
Gobbi H, Carvalho FM, Brot MD, Logullo AF, Silva CAM, Soares FA, Landeiro L, Rahal R, Barrios CH. Challenges in the evaluation of HER2 and HER2-low in breast cancer in Brazil and recommendations of a multidisciplinary working group. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20240313. [PMID: 39356956 PMCID: PMC11444227 DOI: 10.1590/1806-9282.20240313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/01/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Helenice Gobbi
- Universidade Federal do Triângulo Mineiro, Discipline of Special Pathology – Uberaba (MG), Brazil
| | | | - Marina De Brot
- A.C.Camargo Cancer Center, Department of Pathological Anatomy – São Paulo (SP), Brazil
| | | | | | - Fernando Augusto Soares
- Universidade de São Paulo, Institute of Pathological Anatomy, School of Dentistry, D´Or São Luiz Network – São Paulo (SP), Brazil
| | | | - Rosemar Rahal
- Universidade Federal de Goiás, Department of Gynecology – Goiânia (GO), Brazil
| | - Carlos Henrique Barrios
- Latin American Cooperative Oncology Group and Oncoclínicas Group – Porto Alegre (RS), Brazil
| |
Collapse
|
27
|
Dong W, Wang W, Cao C. The Evolution of Antibody-Drug Conjugates: Toward Accurate DAR and Multi-specificity. ChemMedChem 2024; 19:e202400109. [PMID: 38758596 DOI: 10.1002/cmdc.202400109] [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: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
Antibody-drug conjugates (ADCs) consist of antibodies, linkers and payloads. They offer targeted delivery of potent cytotoxic drugs to tumor cells, minimizing off-target effects. However, the therapeutic efficacy of ADCs is compromised by heterogeneity in the drug-to-antibody ratio (DAR), which impacts both cytotoxicity and pharmacokinetics (PK). Additionally, the emergence of drug resistance poses significant challenges to the clinical advancement of ADCs. To overcome these limitations, a variety of strategies have been developed, including the design of multi-specific drugs with accurate DAR. This review critically summarizes the current challenges faced by ADCs, categorizing key issues and evaluating various innovative solutions. We provide an in-depth analysis of the latest methodologies for achieving homogeneous DAR and explore design strategies for multi-specific drugs aimed at combating drug resistance. Our discussion offers a current perspective on the advancements made in refining ADC technologies, with an emphasis on enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Wenge Dong
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Wanqi Wang
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chan Cao
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
28
|
He J, Zeng X, Wang C, Wang E, Li Y. Antibody-drug conjugates in cancer therapy: mechanisms and clinical studies. MedComm (Beijing) 2024; 5:e671. [PMID: 39070179 PMCID: PMC11283588 DOI: 10.1002/mco2.671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Antibody-drug conjugates (ADCs) consist of monoclonal antibodies that target tumor cells and cytotoxic drugs linked through linkers. By leveraging antibodies' targeting properties, ADCs deliver cytotoxic drugs into tumor cells via endocytosis after identifying the tumor antigen. This precise method aims to kill tumor cells selectively while minimizing harm to normal cells, offering safe and effective therapeutic benefits. Recent years have seen significant progress in antitumor treatment with ADC development, providing patients with new and potent treatment options. With over 300 ADCs explored for various tumor indications and some already approved for clinical use, challenges such as resistance due to factors like antigen expression, ADC processing, and payload have emerged. This review aims to outline the history of ADC development, their structure, mechanism of action, recent composition advancements, target selection, completed and ongoing clinical trials, resistance mechanisms, and intervention strategies. Additionally, it will delve into the potential of ADCs with novel markers, linkers, payloads, and innovative action mechanisms to enhance cancer treatment options. The evolution of ADCs has also led to the emergence of combination therapy as a new therapeutic approach to improve drug efficacy.
Collapse
Affiliation(s)
- Jun He
- Department of General Surgery Jiande Branch of the Second Affiliated Hospital, School of Medicine, Zhejiang University Jiande Zhejiang China
| | - Xianghua Zeng
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| | - Chunmei Wang
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| | - Enwen Wang
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| | - Yongsheng Li
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| |
Collapse
|
29
|
Önder T, Ateş Ö, Öner I, Karaçin C. Relationship between HER2-low status and efficacy of CDK4/6 inhibitors in advanced breast cancer: a real-world study. Int J Clin Oncol 2024; 29:972-984. [PMID: 38687407 DOI: 10.1007/s10147-024-02528-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/04/2024] [Indexed: 05/02/2024]
Abstract
AIMS AND OBJECTIVES Human epidermal growth factor receptor 2 (HER2)-low breast cancer (BC) is a new entity considered a biologically distinct subtype from HER2-zero BC. However, the importance of HER2 low expression on the activity of cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) remains unclear. METHODS/MATERIALS We conducted a single-center retrospective study including hormone receptor-positive (HR +) /HER2- metastatic BC (mBC) patients treated with CDK4/6i plus endocrine treatment (ET) as first-line therapy. Clinical outcomes were analyzed according to HER2 expression. RESULTS 258 women were analyzed with a median follow-up of 25.4 months; 39.9% had HER2 low, and 60.1% had HER2 zero BC. Median progression-free survival (mPFS) in the HER2-low group was 27.6 months compared with 44.3 months in the HER2-zero group (p = 0.341). In patients receiving ribociclib, the mPFS in the HER2-low group was 24.2 months compared with 53.1 months in the HER2-zero group (multivariate-adjusted HR: 1.981, 95 Cl 1.094-3.586; p = 0.024). The survival probabilities at 24, 36 and 48 months for the HER2 low and HER2 zero groups were 82%, 69%, 69% and 83%, 75% and 69%, respectively (p = 0.336). Objective response rate (p = 0.179) and disease control rate (p = 0.338) did not significantly differ between HER-2-low and HER-2-zero groups. CONCLUSIONS The mPFS in the Her2-zero group was almost twice that of the Her2-low group, but the difference was not statistically significant. mPFS was significantly longer in the HER2-zero group compared to the HER2-low group in patients receiving ribociclib. More prospective studies are needed to understand the actual consequences of this biomarker.
Collapse
Affiliation(s)
- T Önder
- Dr Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, Turkey.
| | - Ö Ateş
- Dr Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, Turkey
| | - I Öner
- Dr Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, Turkey
| | - C Karaçin
- Dr Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
30
|
Kang S, Kim SB. HER2-Low Breast Cancer: Now and in the Future. Cancer Res Treat 2024; 56:700-720. [PMID: 38291745 PMCID: PMC11261208 DOI: 10.4143/crt.2023.1138] [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: 10/17/2023] [Accepted: 01/28/2024] [Indexed: 02/01/2024] Open
Abstract
Breast cancer is a heterogeneous disease, and its subtypes are characterized by hormone receptor and human epidermal growth factor receptor 2 (HER2) expression status. "HER2-low" tumors, which exhibit a low level of HER2 expression (immunohistochemistry 1+ or 2+ without gene amplification), were conventionally considered not amenable to anti-HER2 targeting agents based on the results of a phase III trial of trastuzumab. However, this perspective is being challenged by the emergence of novel anti-HER2 antibody-drug conjugates, such as trastuzumab-deruxtecan. These innovative therapies have demonstrated remarkable efficacy against HER2-low breast cancer, shedding new light on a previously overlooked category of breast cancer. Such promising results highlight the need for in-depth investigations of the biology and prognostic implications of HER2-low tumors. In this review, we comprehensively summarize the current evidence surrounding this topic and highlight areas that warrant further exploration and research in the future.
Collapse
Affiliation(s)
- Sora Kang
- Division of Hemato-oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
31
|
Lan Y, Zhao J, Zhao F, Li J, Li X. Partial response to trastuzumab deruxtecan (DS8201) following progression in HER2-amplified breast cancer with pulmonary metastases managed with disitamab vedotin (RC48): a comprehensive case report and literature review. Front Oncol 2024; 14:1338661. [PMID: 38952555 PMCID: PMC11215061 DOI: 10.3389/fonc.2024.1338661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/27/2024] [Indexed: 07/03/2024] Open
Abstract
Breast cancer remains one of the predominant malignancies worldwide. In the context of inoperable advanced or metastatic human epidermal growth factor receptor 2 (HER2)-positive breast cancer, systemic management primarily relies on HER2-targeting monoclonal antibodies. With the successful development of anti-HER2 antibody-drug conjugates (ADCs), these agents have been increasingly integrated into therapeutic regimens for metastatic breast cancer. Here, we present the case of a 42-year-old female patient with HER2-positive pulmonary metastatic breast cancer who underwent an extensive treatment protocol. This protocol included chemotherapy, radiation therapy, hormonal therapy, surgical intervention on the breast, and anti-HER2 therapies. The anti-HER2 therapies involved both singular and dual targeting strategies using trastuzumab and the ADC disitamab vedotin (RC48) over an 8-year period. After experiencing disease progression following HER2-targeted therapy with RC48, the patient achieved noticeable partial remission through a therapeutic regimen that combined trastuzumab deruxtecan (DS8201) and tislelizumab. The data suggest a promising role for DS8201 in managing advanced stages of HER2-amplified metastatic breast cancer, especially in cases that demonstrate progression after initial HER2-directed therapies using ADCs. Furthermore, its combination with anti-PD-1 agents enhances therapeutic efficacy by augmenting the anti-tumoral immune response.
Collapse
Affiliation(s)
- Yanfang Lan
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jiahui Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fangrui Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiangpan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
32
|
Vasalou C, Proia TA, Kazlauskas L, Przybyla A, Sung M, Mamidi S, Maratea K, Griffin M, Sargeant R, Urosevic J, Rosenbaum AI, Yuan J, Aluri KC, Ramsden D, Hariparsad N, Jones RD, Mettetal JT. Quantitative evaluation of trastuzumab deruxtecan pharmacokinetics and pharmacodynamics in mouse models of varying degrees of HER2 expression. CPT Pharmacometrics Syst Pharmacol 2024; 13:994-1005. [PMID: 38532525 PMCID: PMC11179703 DOI: 10.1002/psp4.13133] [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: 10/12/2023] [Revised: 02/02/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
Trastuzumab deruxtecan (T-DXd; DS-8201; ENHERTU®) is a human epithelial growth factor receptor 2 (HER2)-directed antibody drug conjugate (ADC) with demonstrated antitumor activity against a range of tumor types. Aiming to understand the relationship between antigen expression and downstream efficacy outcomes, T-DXd was administered in tumor-bearing mice carrying NCI-N87, Capan-1, JIMT-1, and MDA-MB-468 xenografts, characterized by varying HER2 levels. Plasma pharmacokinetics (PK) of total antibody, T-DXd, and released DXd and tumor concentrations of released DXd were evaluated, in addition to monitoring γΗ2AX and pRAD50 pharmacodynamic (PD) response. A positive relationship was observed between released DXd concentrations in tumor and HER2 expression, with NCI-N87 xenografts characterized by the highest exposures compared to the remaining cell lines. γΗ2AX and pRAD50 demonstrated a sustained increase over several days occurring with a time delay relative to tumoral-released DXd concentrations. In vitro investigations of cell-based DXd disposition facilitated the characterization of DXd kinetics across tumor cells. These outputs were incorporated into a mechanistic mathematical model, utilized to describe PK/PD trends. The model captured plasma PK across dosing arms as well as tumor PK in NCI-N87, Capan-1, and MDA-MB-468 models; tumor concentrations in JIMT-1 xenografts required additional parameter adjustments reflective of complex receptor dynamics. γΗ2AX longitudinal trends were well characterized via a unified PD model implemented across xenografts demonstrating the robustness of measured PD trends. This work supports the application of a mechanistic model as a quantitative tool, reliably projecting tumor payload concentrations upon T-DXd administration, as the first step towards preclinical-to-clinical translation.
Collapse
Affiliation(s)
| | | | | | - Anna Przybyla
- AstraZeneca Research & DevelopmentWalthamMassachusettsUSA
| | - Matthew Sung
- AstraZeneca Research & DevelopmentWalthamMassachusettsUSA
| | | | - Kim Maratea
- Clinical Pharmacology & Safety SciencesWalthamMassachusettsUSA
| | - Matthew Griffin
- Clinical Pharmacology & Safety SciencesWalthamMassachusettsUSA
| | | | | | - Anton I. Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology & Safety SciencesSouth San FranciscoCaliforniaUSA
| | - Jiaqi Yuan
- Integrated Bioanalysis, Clinical Pharmacology & Safety SciencesSouth San FranciscoCaliforniaUSA
| | | | - Diane Ramsden
- AstraZeneca Research & DevelopmentWalthamMassachusettsUSA
| | | | | | | |
Collapse
|
33
|
Ascione L, Guidi L, Prakash A, Trapani D, LoRusso P, Lou E, Curigliano G. Unlocking the Potential: Biomarkers of Response to Antibody-Drug Conjugates. Am Soc Clin Oncol Educ Book 2024; 44:e431766. [PMID: 38828973 DOI: 10.1200/edbk_431766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Antibody-drug conjugates (ADCs) have reshaped the cancer treatment landscape across a variety of different tumor types. ADCs' peculiar pharmacologic design combines the cytotoxic properties of chemotherapeutic agents with the selectivity of targeted therapies. At present, the approval of many ADCs used in clinical practice has not always been biomarker-driven. Indeed, predicting ADCs' activity and toxicity through the demonstration of specific biomarkers is still a great unmet need, and the identification of patients who can derive significant benefit from treatment with ADCs may often be uncertain. With the lack of robust predictive biomarkers to anticipate primary, intrinsic resistance to ADCs and no consolidated biomarkers to aid in the early identification of treatment resistance (ie, acquired resistance), the determination of precise biologic mechanisms of ADC activity and safety becomes priority in the quest for better patient-centric outcomes. Of great relevance, whether the target antigen expression is a determinant of ADCs' primary activity is still to be clarified, and available data remain quite controversial. Antigen expression assessment is typically performed on tissue biopsy, hence only providing information on a specific tumor site, therefore unable to capture heterogeneous patterns of tumor antigen expression. Quantifying the expression of the target antigen across all tumor sites would help better understand tumor heterogeneity, whereas molecularly characterizing tumor-intrinsic features over time might provide information on resistance mechanisms. In addition, toxicity can represent a critical concern, since most ADCs have a safety profile that resembles that of chemotherapies, with often unique adverse events requiring special management, possibly because of the differential in pharmacokinetics between the small-molecule agent versus payload of a similar class (eg, deruxtecan conjugate-related interstitial lung disease). As such, the identification of robust predictive biomarkers of safety and activity of ADCs has the potential to improve patient selection and enrich the population of patients most likely to derive a substantial clinical benefit, especially in those disease settings where different ADCs happen to be approved in competing clinical indications, with undefined biomarkers to make precise decision making and unclear data on how to sequence ADCs. At this point, the identification of clinically actionable biomarkers in the space of ADCs remains a top research priority.
Collapse
Affiliation(s)
- Liliana Ascione
- Division of Early Drug Development, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology (DIPO), University of Milan, Milan, Italy
| | - Lorenzo Guidi
- Division of Early Drug Development, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology (DIPO), University of Milan, Milan, Italy
| | - Ajay Prakash
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Dario Trapani
- Division of Early Drug Development, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology (DIPO), University of Milan, Milan, Italy
| | - Patricia LoRusso
- Yale University School of Medicine, Yale Cancer Center, New Haven, CT
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Giuseppe Curigliano
- Division of Early Drug Development, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
34
|
Zhao S, Wang Y, Zhou A, Liu X, Zhang Y, Zhang J. Neoadjuvant chemotherapy efficacy and prognosis in HER2-low and HER2-zero breast cancer patients by HR status: a retrospective study in China. PeerJ 2024; 12:e17492. [PMID: 38827304 PMCID: PMC11143972 DOI: 10.7717/peerj.17492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
Background The promising efficacy of novel anti-HER2 antibody-drug conjugates (ADC) in HER2-low breast cancer has made HER2-low a research hotspot. However, controversy remains regarding the neoadjuvant chemotherapy (NAC) efficacy, prognosis, and the relationship with hormone receptor (HR) status of HER2-low. Methods A retrospective analysis was conducted on 975 patients with HER2-negative breast cancer undergoing NAC at Tianjin Medical University Cancer Institute and Hospital, evaluating pathological complete response (pCR) rate and prognosis between HER2-low and HER2-zero in the overall cohort and subgroups. Results Overall, 579 (59.4%) and 396 (40.6%) patients were HER2-low and HER2-zero disease, respectively. Compared with HER2-zero, the HER2-low cohort consists of more postmenopausal patients, with lower histological grade and higher HR positivity. In the HR-positive subgroup, HER2-low cases remain to exhibit lower histological grade, while in the HR-negative subgroup, they show higher grade. The HER2-low group had lower pCR rates than the HER2-zero group (16.4% vs. 24.0%). In the HR-positive subgroup, HER2-low consistently showed lower pCR rate (8.1% vs. 15.5%), and served as an independent suppressive factor for the pCR rate. However, no significant difference was observed in the pCR rates between HER2-low and HER2-zero in the HR-negative breast cancer. In the entire cohort and in stratified subgroups based on HR and pCR statuses, no difference in disease-free survival were observed between HER2-low and HER2-zero. Conclusions In the Chinese population, HER2-low breast cancer exhibits distinct characteristics and efficacy of NAC in different HR subgroups. Its reduced pCR rate in HR-positive subgroup is particularly important for clinical decisions. However, HER2-low is not a reliable factor for assessing long-term survival outcomes.
Collapse
Affiliation(s)
- Shaorong Zhao
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuyun Wang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Angxiao Zhou
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xu Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yi Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jin Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
35
|
Chen H, Yang G, Ma J. Ocular toxicity associated with anti-HER2 agents in breast cancer: A pharmacovigilance analysis using the FAERS database. Int J Cancer 2024; 154:1616-1625. [PMID: 38216995 DOI: 10.1002/ijc.34848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/04/2023] [Accepted: 12/27/2023] [Indexed: 01/14/2024]
Abstract
Anti-human epidermal growth factor receptor 2 (HER2) agents have exhibited pronounced tumor-inhibitory activity, yet the accompanying ocular toxicity has frequently been underestimated. We aim to conduct a comprehensive comparative analysis of ocular toxicity risk related to various anti-HER2 agents. We executed a retrospective pharmacovigilance investigation based on the FDA Adverse Event Reporting System (FAERS) database, covering the period from Q2 2018 to Q1 2023. The disproportionality analysis was performed to assess ocular toxicity risk. Multivariate logistic regression was implemented to mitigate potential biases. Moreover, the time to onset of ocular toxicity was also evaluated. A total of 3467 ocular adverse event (AE) reports concerning anti-HER2 agents were collected. At the preferred term (PT) level, there were 69 positive signals, among which excessive eye blinking, abnormal sensation in the eye, and asthenopia presented a significant risk. In comparison to tyrosine kinase inhibitors (TKIs), antibody drugs were associated with a broader range of ocular disorders at Standardized MedDRA Queries (SMQ)levels, including conjunctival disorders, corneal disorders, ocular infections, ocular motility disorders, optic nerve disorders, and retinal disorders. In terms of onset time, pertuzumab displayed an earlier onset at 21.5 days, while trastuzumab deruxtecan had the latest at 91.5 days. In summary, our study reveals varying degrees of ocular toxicity related to anti-HER2 agents, with a significantly higher risk observed in antibody drugs. Additionally, novel ocular toxicity signals, not documented in product labels, have been detected. In the future, further research will be necessary to validate our findings.
Collapse
Affiliation(s)
- Heng Chen
- Department of Pharmacy, The First Hospital of Changsha, Changsha, Hunan, China
| | - Guoping Yang
- Center of Clinical Pharmacology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junlong Ma
- Center of Clinical Pharmacology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
36
|
He M, Zhao W, Wang P, Li W, Chen H, Yuan Z, Pan G, Gao H, Sun L, Chu J, Li L, Hu Y. Efficacy and safety of Trastuzumab Emtansine in treating human epidermal growth factor receptor 2-positive metastatic breast cancer in Chinese population: a real-world multicenter study. Front Med (Lausanne) 2024; 11:1383279. [PMID: 38741766 PMCID: PMC11089149 DOI: 10.3389/fmed.2024.1383279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/04/2024] [Indexed: 05/16/2024] Open
Abstract
Background Trastuzumab emtansine (T-DM1) has been approved worldwide for treating metastatic breast cancer (mBC) in patients who have received first-line therapy, shown disease progression, and are human epidermal growth factor receptor 2 (HER2)-positive. T-DM1 received approval in China to treat early-stage breast cancer (BC) in 2020 and for mBC in 2021. In March 2023, T-DM1 was included in medical insurance coverage, significantly expanding the eligible population. Materials and methods This post-marketing observational study aimed to assess the safety and effectiveness of T-DM1 in real-world clinical practice in China. This study enrolled 31 individuals with HER2-positive early-stage BC and 70 individuals with HER2-positive advanced BC from 8 study centers in Shandong Province, China. The T-DM1 dosage was 3.6 mg/kg injected intravenously every 3 weeks until the disease advanced or the drug toxicity became uncontrollable, whichever occurred earlier. Additionally, efficacy and safety information on T-DM1 were collected. Results During the 7-month follow-up period, no recurrence or metastases were observed in patients who had early-stage BC. The disease control rate was 31.43% (22/70) in patients with advanced BC. The most common adverse effect of T-DM1 was thrombocytopenia, with an incidence of 69.31% (70/101), and the probability of Grade ≥ 3 thrombocytopenia was 11.88% (12/101). Conclusion This real-world study demonstrated that T-DM1 had good efficacy and was well tolerated by both HER2-positive early-stage BC and mBC patients.
Collapse
Affiliation(s)
- Miao He
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
- Department of Medical Oncology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Wen Zhao
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Peng Wang
- Department of Medical Oncology, Qingdao Shibei Changqing Hospital, Qingdao, Shandong Province, China
| | - Wenhuan Li
- Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Hanhan Chen
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zonghuai Yuan
- Department of General Surgery, People’s Hospital of Rizhao, Rizhao, Shandong Province, China
| | - Guangye Pan
- Department of General Surgery, People’s Hospital of Rizhao, Rizhao, Shandong Province, China
| | - Hong Gao
- Department of Breast and Thyroid Surgery, Rizhao Traditional Chinese Medical Hospital, Rizhao, Shandong Province, China
| | - Lijun Sun
- Department of Breast and Thyroid Surgery, People’s Hospital of Juxian, Rizhao, Shandong Province, China
| | - Jiahui Chu
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Li Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Yu Hu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
37
|
Qu F, Lu R, Liu Q, Wu X, Huang X, Yin Y, Li W. Antibody-drug conjugates transform the outcome of individuals with low-HER2-expression advanced breast cancer. Cancer 2024; 130:1392-1402. [PMID: 38271367 DOI: 10.1002/cncr.35205] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024]
Abstract
Antibody-drug conjugates (ADCs)-a groundbreaking class of agents for targeted oncological therapies-consist of monoclonal antibodies with strong antigenic specificity coupled with highly active cytotoxic agents (also referred to as "payloads"). Over the past 2 decades, breast cancer research has evolved into a focal point for the research and development of ADCs, leading to several recent landmark publications. These advancements are ushering in a transformative era in breast cancer treatment and redefining conventional classifications by introducing a prospective subtype termed "HER2-low." The latest iterations of ADCs have demonstrated enhanced efficacy in disease management through the optimization of various factors, notably the incorporation of the bystander effect. These conjugates are no longer limited to the oncogenic driver human epidermal growth factor receptor 2 (HER2). Other antigens, including human epidermal growth factor receptor 3 (HER3), trophoblast cell surface antigen 2 (Trop-2), zinc transporter ZIP6 (LIV-1), and folate receptor α (FRα), have recently emerged as intriguing tumor cell surface nondriver gene targets for ADCs, each with one or more specific ADCs that showed encouraging results in the breast cancer field. This article reviews recent advances in the application of ADCs in the treatment of HER2-low breast cancer. Additionally, this review explores the underlying factors contributing to the impact of target selection on ADC efficacy to provide new insights for optimizing the clinical application of ADCs in individuals with low HER2 expression in advanced breast cancer.
Collapse
Affiliation(s)
- Fei Qu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Rongrong Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Qian Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Xuefang Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Xiang Huang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
38
|
Zhang H, Shen G, Yang P, Li J, Li Z, Liu Z, Wang M, Zhao F, Ren D, Liu Z, Zhao J, Zhao Y. Incidence of antibody-drug conjugate-related fatigue in patients with breast cancer: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2024; 196:104292. [PMID: 38403093 DOI: 10.1016/j.critrevonc.2024.104292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/27/2024] [Accepted: 02/07/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Numerous studies have reported the efficacy of antibody-drug conjugates (ADCs) for treating breast cancer. However, during cytotoxic drug treatment, long-term disabling fatigue is common. Moreover, studies in the relevant literature have indicated that fatigue can significantly increase the incidence of depression and sleep disorders. Therefore, this meta-analysis aims to evaluate the incidence of fatigue in breast cancer survivors treated with ADCs. METHODS PubMed, EMBASE, Web of Science, and Cochrane Library databases were systematically searched for articles and conference abstracts published before March 16, 2023. Further, two authors independently extracted data from the included studies. The primary outcome of this study was the incidence of all-grade fatigue caused by the use of ADCs in patients with breast cancer. Finally, a random-effects model was used to calculate the incidence and 95% confidence intervals (CIs) of the outcome. RESULTS Overall, 7963 patients from 31 studies were included in this meta-analysis to assess the incidence of fatigue caused by the use of approved and marketed ADCs in patients with breast cancer. Notably, the incidence of all-grade fatigue during ADC monotherapy was 39.84% (95% CI, 35.09%-44.69%). In subgroup analyses, among ADCs, the incidence of trastuzumab deruxtecan-induced fatigue was the highest, with an all-grade fatigue incidence of 47.05% (95% CI, 42.38%-51.75%). Meanwhile, the incidence of trastuzumab emtansine (T-DM1)-induced all-grade fatigue was 35.17% (95% CI, 28.87%-41.74%), which was the lowest among ADCs. Further, the incidence of all-grade fatigue due to sacituzumab govitecan was 42.82% (95% CI, 34.54%-51.32%), which was higher than that due to T-DM1. Moreover, the incidence of fatigue was higher with T-DM1 combination therapy than with monotherapy. CONCLUSIONS Clinicians have highlighted the high incidence of ADC-related fatigue and its negative impact on patients' physical and mental health, making fatigue an important research variable. The results of this study will further contribute to a comprehensive understanding of ADCs, which have some clinical importance and are of great benefit to patients with breast cancer.
Collapse
Affiliation(s)
- Hengheng Zhang
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - GuoShuang Shen
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Ping Yang
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Jinming Li
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Zitao Li
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Zhen Liu
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Miaozhou Wang
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Fuxing Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Dengfeng Ren
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Zhilin Liu
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Jiuda Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Yi Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| |
Collapse
|
39
|
Curigliano G, Dent R, Earle H, Modi S, Tarantino P, Viale G, Tolaney SM. Open questions, current challenges, and future perspectives in targeting human epidermal growth factor receptor 2-low breast cancer. ESMO Open 2024; 9:102989. [PMID: 38613914 PMCID: PMC11024577 DOI: 10.1016/j.esmoop.2024.102989] [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: 12/07/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 04/15/2024] Open
Abstract
Approximately 60% of traditionally defined human epidermal growth factor receptor 2 (HER2)-negative breast cancers express low levels of HER2 [HER2-low; defined as immunohistochemistry (IHC) 1+ or IHC 2+/in situ hybridization (ISH)-]. HER2-low breast cancers encompass a large percentage of both hormone receptor-positive (up to 85%) and triple-negative (up to 63%) breast cancers. The DESTINY-Breast04 trial established that HER2-low tumors are targetable, leading to the approval of trastuzumab deruxtecan (T-DXd) as the first HER2-directed therapy for the treatment of HER2-low breast cancer in the United States and Europe. This change in the clinical landscape results in a number of questions and challenges-including those related to HER2 assessment and patient identification-and highlights the need for careful assessment of HER2 expression to identify patients eligible for T-DXd. This review provides context for understanding how to identify patients with HER2-low breast cancer with respect to sample types, scoring and reporting HER2 status, and testing methods and assays. It also discusses management of important T-DXd-related adverse events. Available evidence supports the efficacy of T-DXd in patients with any history of IHC 1+ or IHC 2+/ISH- scores; however, future research may further refine the population who could benefit from T-DXd or other HER2-directed therapies and identify novel methods for patient identification. Because HER2 expression can change with disease progression or treatment, and variability exists in scoring and interpretation of HER2 status, careful re-evaluation in certain scenarios may help to identify more patients who may benefit from T-DXd.
Collapse
Affiliation(s)
- G Curigliano
- European Institute of Oncology, IRCCS, Milan; Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy.
| | - R Dent
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - H Earle
- Blogger at hannahincancerland.com, New Hampshire, USA; Patient at Dana-Farber Cancer Institute, Harvard Medical School, Boston
| | - S Modi
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - P Tarantino
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - G Viale
- European Institute of Oncology, IRCCS, Milan
| | - S M Tolaney
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Liao Q, Zhang R, Ou Z, Ye Y, Zeng Q, Wang Y, Wang A, Chen T, Chai C, Guo B. TROP2 is highly expressed in triple-negative breast cancer CTCs and is a potential marker for epithelial mesenchymal CTCs. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200762. [PMID: 38596285 PMCID: PMC10869581 DOI: 10.1016/j.omton.2024.200762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/05/2023] [Accepted: 01/05/2024] [Indexed: 04/11/2024]
Abstract
Circulating tumor cells (CTCs) are the seeds of distant metastases of malignant tumors and are associated with malignancy and risk of metastasis. However, tumor cells undergo epithelial-mesenchymal transition (EMT) during metastasis, leading to the emergence of different types of CTCs. Real-time dynamic molecular and functional typing of CTCs is necessary to precisely guide personalized treatment. Most CTC detection systems are based on epithelial markers that may fail to detect EMT CTCs. Therefore, it is clinically important to identify new markers of different CTC types. In this study, bioinformatics analysis and experimental assays showed that trophoblast cell surface antigen 2 (TROP2), a target molecule for advanced palliative treatment of triple-negative breast cancer (TNBC), was highly expressed in TNBC tissues and tumor cells. Furthermore, TROP2 can promote the migration and invasion of TNBC cells by upregulating EMT markers. The specificity and potential of TROP2 as an EMT-associated marker of TNBC CTCs were evaluated by flow cytometry, immunofluorescence, spiking experiments, and a well-established CTC assay. The results indicated that TROP2 is a potential novel CTC marker associated with EMT, providing a basis for more efficacious markers that encompass CTC heterogeneity in patients with TNBC.
Collapse
Affiliation(s)
- Qingyu Liao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ruiming Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zuli Ou
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yan Ye
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qian Zeng
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yange Wang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Anqi Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
| | - Tingmei Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Chengsen Chai
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Bianqin Guo
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
- Department of Clinical Laboratory, Chongqing University Cancer Hospital, Chongqing 40030, China
| |
Collapse
|
41
|
Marra A, Chandarlapaty S, Modi S. Management of patients with advanced-stage HER2-positive breast cancer: current evidence and future perspectives. Nat Rev Clin Oncol 2024; 21:185-202. [PMID: 38191924 DOI: 10.1038/s41571-023-00849-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 01/10/2024]
Abstract
Amplification and/or overexpression of ERBB2, the gene encoding HER2, can be found in 15-20% of invasive breast cancers and is associated with an aggressive phenotype and poor clinical outcomes. Relentless research efforts in molecular biology and drug development have led to the implementation of several HER2-targeted therapies, including monoclonal antibodies, tyrosine-kinase inhibitors and antibody-drug conjugates, constituting one of the best examples of bench-to-bedside translation in oncology. Each individual drug class has improved patient outcomes and, importantly, the combinatorial and sequential use of different HER2-targeted therapies has increased cure rates in the early stage disease setting and substantially prolonged survival for patients with advanced-stage disease. In this Review, we describe key steps in the development of the modern paradigm for the treatment of HER2-positive advanced-stage breast cancer, including selecting and sequencing new-generation HER2-targeted therapies, and summarize efficacy and safety outcomes from pivotal studies. We then outline the factors that are currently known to be related to resistance to HER2-targeted therapies, such as HER2 intratumoural heterogeneity, activation of alternative signalling pathways and immune escape mechanisms, as well as potential strategies that might be used in the future to overcome this resistance and further improve patient outcomes.
Collapse
Affiliation(s)
- Antonio Marra
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Shanu Modi
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
42
|
Valenza C, Guidi L, Battaiotto E, Trapani D, Sartore Bianchi A, Siena S, Curigliano G. Targeting HER2 heterogeneity in breast and gastrointestinal cancers. Trends Cancer 2024; 10:113-123. [PMID: 38008666 DOI: 10.1016/j.trecan.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/28/2023]
Abstract
About 20% of breast and gastric cancers and 3% of colorectal carcinomas overexpress the human epidermal growth factor receptor 2 (HER2) and are sensitive to HER2-directed agents. The expression of HER2 may differ within the same tumoral lesion (spatial intralesional heterogeneity), from different tumor locations (spatial interlesional heterogeneity), and throughout treatments (temporal heterogeneity). Spatial and temporal heterogeneity may impact on response and resistance to HER2-targeting agents and its prevalence and predictive role changes across HER2-overexpressing solid tumors. Therefore, the definition and the characterization of HER2 heterogeneity pose many challenges and its implementation as a reproducible predictive biomarker would help in guiding treatment modulation.
Collapse
Affiliation(s)
- Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Lorenzo Guidi
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Elena Battaiotto
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Andrea Sartore Bianchi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
43
|
Liang Y, Zhang P, Li F, Lai H, Qi T, Wang Y. Advances in the study of marketed antibody-drug Conjugates (ADCs) for the treatment of breast cancer. Front Pharmacol 2024; 14:1332539. [PMID: 38352694 PMCID: PMC10862125 DOI: 10.3389/fphar.2023.1332539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Breast cancer continues to have a high incidence rate among female malignancies. Despite significant advancements in treatment modalities, the heterogeneous nature of breast cancer and its resistance to various therapeutic approaches pose considerable challenges. Antibody-drug conjugates (ADCs) effectively merge the specificity of antibodies with the cytotoxicity of chemotherapeutic agents, offering a novel strategy for precision treatment of breast cancer. Notably, trastuzumab emtansine (T-DM1) has provided a new therapeutic option for HER2-positive breast cancer patients globally, especially those resistant to conventional treatments. The development of trastuzumab deruxtecan (T-DXd) and sacituzumab govitecan (SG) has further broadened the applicability of ADCs in breast cancer therapy, presenting new hopes for patients with low HER2 expression and triple-negative breast cancer. However, the application of ADCs presents certain challenges. For instance, their treatment may lead to adverse reactions such as interstitial lung disease, thrombocytopenia, and diarrhea. Moreover, prolonged treatment could result in ADCs resistance, complicating the therapeutic process. Economically, the high costs of ADCs might hinder their accessibility in low-income regions. This article reviews the structure, mechanism of action, and clinical trials of commercially available ADCs for breast cancer treatment, with a focus on the clinical trials of the three drugs, aiming to provide insights for clinical applications and future research.
Collapse
Affiliation(s)
- Yan Liang
- Sichuan Cancer Hospital, Cancer Hospital Affiliate University of Electronic Science and Technology, Chengdu, China
- School of Medicine, University of Electronic Science and Technology, Chengdu, China
| | - Purong Zhang
- Sichuan Cancer Hospital, Cancer Hospital Affiliate University of Electronic Science and Technology, Chengdu, China
| | - Feng Li
- Sichuan Cancer Hospital, Cancer Hospital Affiliate University of Electronic Science and Technology, Chengdu, China
- School of Medicine, University of Electronic Science and Technology, Chengdu, China
| | - Houyun Lai
- Sichuan Cancer Hospital, Cancer Hospital Affiliate University of Electronic Science and Technology, Chengdu, China
- School of Medicine, University of Electronic Science and Technology, Chengdu, China
| | - Tingting Qi
- Sichuan Cancer Hospital, Cancer Hospital Affiliate University of Electronic Science and Technology, Chengdu, China
| | - Yixin Wang
- Sichuan Cancer Hospital, Cancer Hospital Affiliate University of Electronic Science and Technology, Chengdu, China
| |
Collapse
|
44
|
Cherifi F, Da Silva A, Martins-Branco D, Awada A, Nader-Marta G. Pharmacokinetics and pharmacodynamics of antibody-drug conjugates for the treatment of patients with breast cancer. Expert Opin Drug Metab Toxicol 2024; 20:45-59. [PMID: 38214896 DOI: 10.1080/17425255.2024.2302460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
INTRODUCTION Currently three antibody-drug-conjugates (ADC) are approved by the European Medicines Agency (EMA) for treatment of breast cancer (BC) patient: trastuzumab-emtansine, trastuzumab-deruxtecan and sacituzumab-govitecan. ADC are composed of a monoclonal antibody (mAb) targeting a specific antigen, a cytotoxic payload and a linker. Pharmacokinetics (PK) and pharmacodynamics (PD) distinguish ADC from conventional chemotherapy and must be understood by clinicians. AREAS COVERED Our review delineates the PK/PD profiles of ADC approved for the treatment of BC with insight for future development. This is an expert opinion literature review based on the EMA's Assessment Reports, enriched by a comprehensive literature search performed on Medline in August 2023. EXPERT OPINION All three ADC distributions are described by a two-compartment structure: tissue and serum. Payload concentration peak is immediate but remains at low concentration. The distribution varied for all ADC only with body weight. mAb will be metabolised firstly by the saturable complex formation of ADC/Tumour-Receptor and secondly by binding of FcgRs in immune cells. They are all excreted in the bile and faeces with minimal urine elimination. Dose adjustments, apart from weight, are not recommended. Novel ADC are composed of cleavable linkers with various targets/payloads with the same PK/PD properties, but novel structures of ADC are in development.
Collapse
Affiliation(s)
- François Cherifi
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Breast Cancer Unit, CLCC François Baclesse, Institut Normand du Sein, Caen, France
| | - Angélique Da Silva
- Departments of Pharmacology and Medical Oncology, Caen-Normandy University Hospital, PICARO Cardio-Oncology Program, Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE, Caen, France
| | - Diogo Martins-Branco
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Ahmad Awada
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Guilherme Nader-Marta
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| |
Collapse
|
45
|
Neubauer Z, Hasan S, Press RH, Chhabra AM, Fox J, Bakst R, Simone CB, Choi JI. Prognostic implications of HER2NEU-low in metastatic breast cancer. Cancer Med 2024; 13:e6979. [PMID: 38379326 PMCID: PMC10839127 DOI: 10.1002/cam4.6979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/17/2023] [Accepted: 01/18/2024] [Indexed: 02/22/2024] Open
Abstract
INTRODUCTION We explored characteristics and clinical outcomes of HER2-negative and HER2-low metastatic breast cancers using real-world data. METHODS We queried the National Cancer Database to identify MBC patients that were HER2-low or HER2-negative per immunohistochemical staining. A binomial regression analysis identified demographic and clinical correlates of each subtype. A Cox multivariable regression analysis (MVA) and propensity-match analysis were performed to identify correlates of survival. RESULTS Excluding missing data, 24,636 MBC patients diagnosed between 2008 and 2015 were identified; 27.9% were HER2-negative and 72.1% were HER2-low. There were no relevant demographic differences between the groups. HER2-low tumors were half as likely to have concomitant hormone receptor-positive status (p < 0.01). The 3-year survival rate among hormone receptor-negative patients was 33.8% for HER2-low and 32.2% for HER2-negative (p < 0.05), and 60.9% and 55.6% in HER2-low and HER2-negative cases among hormone receptor-positive patients (p < 0.05), respectively. HER2-low cases were associated with better survival on MVA (HR =0.95, 95% CI 0.91-0.99) and remained superior with propensity-matching (HR = 0.92, 95% CI 0.89-0.96). In a subset analysis isolated to hormone receptor-positive cases, HER2-low remained correlated with improved survival (HR = 0.93, 95% CI 0.89-0.98) with propensity-matched MVA. Correlates of worse survival include older age as a continuous variable (HR = 1.02, 95% CI 1.02-1.02) and Black race (HR = 1.26, 95% CI 1.20-1.32) [all p < 0.01]. CONCLUSIONS In the largest such analysis performed to date, our study demonstrates a small but statistically significant association with improved survival for HER2-low tumors compared to HER2-negative tumors in MBC.
Collapse
Affiliation(s)
| | | | | | | | - Jana Fox
- Montefiore Medical CenterDepartment of Radiation OncologyNew YorkNew YorkUSA
| | - Richard Bakst
- Mount Sinai Medical Center. Department of Radiation OncologyNew YorkNew YorkUSA
| | | | | |
Collapse
|
46
|
Trapé J, Fernández-Galán E, Auge JM, Carbonell-Prat M, Filella X, Miró-Cañís S, González-Fernández C. Factors influencing blood tumor marker concentrations in the absence of neoplasia. Tumour Biol 2024; 46:S35-S63. [PMID: 38517826 DOI: 10.3233/tub-220023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
BACKGROUND Tumor markers (TMs) are a heterogeneous group of molecules used in the diagnosis, prognosis and follow-up of cancer patients. During neoplastic differentiation, cells can either directly synthesize or induce the synthesis of TMs, and the release of these molecules into the bloodstream allows their quantification in biological fluids. Although very small concentrations of TMs are usually present in the serum or plasma of healthy subjects, increased concentrations may also be found in the presence of benign diseases or due to technical interference, producing false positive results. MATERIAL AND METHODS AND RESULTS Our review analyses the causes of false positives described between January 1970 to February 2023 for the TMs most frequently used in clinical practice: α-fetoprotein (AFP), β2-microglobulin (β2-M), cancer antigen 15-3 (CA 15-3), cancer antigen CA 19-9 (CA 19-9), cancer antigen CA 72-4 (CA 72-4), cancer antigen 125 (CA 125), carcinoembryonic antigen (CEA), chromogranin A (CgA), choriogonadotropin (hCG), cytokeratin 19 fragment (CYFRA 21-1), neuron-specific enolase (NSE), human epididymis protein 4 (HE4), serum HER2 (sHER2), squamous cell carcinoma antigen (SCCA), protein induced by vitamin K absence-II (PIVKA-II), Pro-gastrin-releasing peptide (Pro-GRP), prostate-specific antigen (PSA), Protein S-100 (S-100) and thyroglobulin (Tg). A total of 247 references were included. CONCLUSIONS A better understanding of pathophysiological processes and other conditions that affect the concentration of TMs might improve the interpretation of results and their clinical application.
Collapse
Affiliation(s)
- Jaume Trapé
- Department of Laboratory Medicine, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Catalonia, Spain
- Tissue Repair and Regeneration Laboratory, Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central, Barcelona, Spain
- Faculty of Medicine, University of Vic - Central University of Catalonia, Vic, Spain
| | - Esther Fernández-Galán
- Department of Biochemistry and Molecular Genetics - Hospital Clinic de Barcelona, Barcelona, Spain
| | - Josep Maria Auge
- Department of Biochemistry and Molecular Genetics - Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Xavier Filella
- Department of Biochemistry and Molecular Genetics - Hospital Clinic de Barcelona, Barcelona, Spain
| | - Sílvia Miró-Cañís
- Laboratori d'Anàlisis Clíniques, CLILAB Diagnòstics, Vilafranca del Penedès, Spain
| | - Carolina González-Fernández
- Department of Laboratory Medicine, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Catalonia, Spain
- Gastrointestinal Oncology, Endoscopy and Surgery Research Group, Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central, Barcelona, Spain
| |
Collapse
|
47
|
Ali A, Graff SL. Exploring DESTINY: the Past, Present, and Future of Trastuzumab Deruxtecan. Curr Oncol Rep 2024; 26:1-9. [PMID: 38091201 DOI: 10.1007/s11912-023-01478-2] [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] [Accepted: 10/29/2023] [Indexed: 02/12/2024]
Abstract
PURPOSE OF REVIEW HER2-positive breast cancer accounts for 10-15% of all breast cancers and fam-trastuzumab deruxtecan (T-DXd) has played a major role in moving the treatment of HER2-expressing disease forward. RECENT FINDINGS T-DXd is a novel antibody-drug conjugate (ADC) composed of a humanized IgG1 monoclonal antibody against HER2 receptor bound to a potent topoisomerase I cytotoxin payload by a cleavable peptide linker. It has been shown to have robust preclinical activity in pretreated cancer cell lines, as well as meaningful clinical activity in advanced HER2-expressing breast cancer. Recent studies have demonstrated T-DXd as an active agent for metastatic HER2-positive patients, and as a viable additional line for heavily pretreated patients with HER2-low disease. The toxicity of T-DXd remains manageable and burden of side effects seems to be lower when offered as an earlier line of therapy over the course of treatment. In this review, we discuss the pharmacology of T-DXd, review pertinent preclinical and clinical data, and address potential challenges and future directions related to the use of T-DXd in clinical practice.
Collapse
Affiliation(s)
- Azka Ali
- Department of Breast Medical Oncology, Cleveland Clinic Foundation, Taussig Cancer Institute, 9500 Euclid Ave / CA-060, Cleveland, OH, 44195, USA.
| | - Stephanie L Graff
- Division of Hematology & Medical Oncology, Lifespan Cancer Institute, Legorreta Cancer Center at Brown University, 593 Eddy St., Providence, RI, 02903, USA
| |
Collapse
|
48
|
Angelis V, Okines AFC. Systemic Therapies for HER2-Positive Advanced Breast Cancer. Cancers (Basel) 2023; 16:23. [PMID: 38201451 PMCID: PMC10777942 DOI: 10.3390/cancers16010023] [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: 10/03/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024] Open
Abstract
Despite recent advances, HER2-positive advanced breast cancer (ABC) remains a largely incurable disease, with resistance to conventional anti-HER2 drugs ultimately unavoidable for all but a small minority of patients who achieve an enduring remission and possibly cure. Over the past two decades, significant advances in our understanding of the underlying molecular mechanisms of HER2-driven oncogenesis have translated into pharmaceutical advances, with the developing of increasingly sophisticated therapies directed against HER2. These include novel, more potent selective HER2 tyrosine kinase inhibitors (TKIs); new anti-HER2 antibody-drug conjugates; and dual epitope targeting antibodies, with more advanced pharmacological properties and higher affinity. With the introduction of adjuvant T-DM1 for incomplete responders to neoadjuvant therapy, fewer patients are relapsing, but for those who do relapse, disease that may be resistant to standard first- and second-line therapies requires new approaches. Furthermore, the risk of CNS relapse has not been abrogated by current (neo)adjuvant strategies; therefore, current research efforts are being directed towards this challenging site of metastatic disease. In this article, we review the currently available clinical data informing the effective management of HER2-positive breast cancer beyond standard first-line therapy with pertuzumab, trastuzumab, and taxanes, and the management of relapse in patients who have already been exposed to both these agents and T-DM1 for early breast cancer (EBC). We additionally discuss novel anti-HER2 targeted agents and combinations in clinical trials, which may be integrated into standard treatment paradigms in the future.
Collapse
Affiliation(s)
| | - Alicia F. C. Okines
- Department of Medicine, Royal Marsden NHS Foundation Trust, Fulham Road, Chelsea, London SW3 6JJ, UK;
| |
Collapse
|
49
|
Filis P, Zerdes I, Soumala T, Matikas A, Foukakis T. The ever-expanding landscape of antibody-drug conjugates (ADCs) in solid tumors: A systematic review. Crit Rev Oncol Hematol 2023; 192:104189. [PMID: 37866413 DOI: 10.1016/j.critrevonc.2023.104189] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/29/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND The advent of targeted therapies signaled novel avenues for more optimal oncological outcomes. Antibody-drug conjugates (ADCs) have risen as a cornerstone of the ever-expanding targeted therapy era. The purpose of this systematic review is to delineate the rapidly evolving clinical landscape of ADCs for solid tumors. METHODS A literature search was performed in Medline, Embase and Cochrane databases for phase II and III clinical trials. Outcomes of interest were the objective response rate, overall survival, progression-free survival and adverse events. RESULTS A total of 92 clinical trials (76 phase II and 16 phase III) evaluated the efficacy and safety of ADCs for a plethora of solid tumors. Out of the 30 investigated ADCs, 8 have received approval by regulatory organizations for solid tumors. Currently, 52 phase III clinical trials for ADCs are ongoing. CONCLUSION ADCs have shown promising results for several solid tumors and various cancer settings.
Collapse
Affiliation(s)
- Panagiotis Filis
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece.
| | - Ioannis Zerdes
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Theodora Soumala
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Alexios Matikas
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
50
|
Marchiò C, Criscitiello C, Scatena C, Santinelli A, Graziano P, Malapelle U, Cursano G, Venetis K, Fanelli GN, Pepe F, Berrino E, De Angelis C, Perrone G, Curigliano G, Fusco N. Think "HER2" different: integrative diagnostic approaches for HER2-low breast cancer. Pathologica 2023; 115:292-301. [PMID: 38180137 PMCID: PMC10767801 DOI: 10.32074/1591-951x-942] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 01/06/2024] Open
Abstract
This work explores the complex field of HER2 testing in the HER2-low breast cancer era, with a focus on methodological aspects. We aim to propose clear positions to scientific societies, institutions, pathologists, and oncologists to guide and shape the appropriate diagnostic strategies for HER2-low breast cancer. The fundamental question at hand is whether the necessary tools to effectively translate our knowledge about HER2 into practical diagnostic schemes for the lower spectrum of expression are available. Our investigation is centered on the significance of distinguishing between an immunohistochemistry (IHC) score 0 and score 1+ in light of the clinical implications now apparent, as patients with HER2-low breast cancer become eligible for trastuzumab-deruxtecan treatment. Furthermore, we discuss the definition of HER2-low beyond its conventional boundaries and assess the reliability of established diagnostic procedures designed at a time when therapeutic perspectives were non-existent for these cases. In this regard, we examine potential complementary technologies, such as gene expression analysis and liquid biopsy. Ultimately, we consider the potential role of artificial intelligence (AI) in the field of digital pathology and its integration into HER2 testing, with a particular emphasis on its application in the context of HER2-low breast cancer.
Collapse
Affiliation(s)
- Caterina Marchiò
- Division of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Carmen Criscitiello
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Cristian Scatena
- Department of Laboratory Medicine, Pisa University Hospital, Anatomic Pathology 1 Universitaria, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Alfredo Santinelli
- Anatomic Pathology, Azienda Sanitaria Territoriale di Pesaro-Urbino, Pesaro, Italy
| | - Paolo Graziano
- Pathology Unit, Fondazione IRCCS Ospedale ‘Casa Sollievo della Sofferenza’, San Giovanni Rotondo (FG), Italy
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Giuseppe Nicolò Fanelli
- Department of Laboratory Medicine, Pisa University Hospital, Anatomic Pathology 1 Universitaria, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | - Enrico Berrino
- Division of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Giuseppe Perrone
- Department of Medicine and Surgery, Research Unit of Anatomical Pathology, Università Campus Bio-Medico di Roma, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Pathology, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| |
Collapse
|