1
|
Serpico L, Zhu Y, Maia RF, Sumedha S, Shahbazi MA, Santos HA. Lipid nanoparticles-based RNA therapies for breast cancer treatment. Drug Deliv Transl Res 2024; 14:2823-2844. [PMID: 38831199 PMCID: PMC11384647 DOI: 10.1007/s13346-024-01638-2] [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] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Breast cancer (BC) prevails as a major burden on global healthcare, being the most prevalent form of cancer among women. BC is a complex and heterogeneous disease, and current therapies, such as chemotherapy and radiotherapy, frequently fall short in providing effective solutions. These treatments fail to mitigate the risk of cancer recurrence and cause severe side effects that, in turn, compromise therapeutic responses in patients. Over the last decade, several strategies have been proposed to overcome these limitations. Among them, RNA-based technologies have demonstrated their potential across various clinical applications, notably in cancer therapy. However, RNA therapies are still limited by a series of critical issues like off-target effect and poor stability in circulation. Thus, novel approaches have been investigated to improve the targeting and bioavailability of RNA-based formulations to achieve an appropriate therapeutic outcome. Lipid nanoparticles (LNPs) have been largely proven to be an advantageous carrier for nucleic acids and RNA. This perspective explores the most recent advances on RNA-based technology with an emphasis on LNPs' utilization as effective nanocarriers in BC therapy and most recent progresses in their clinical applications.
Collapse
Affiliation(s)
- Luigia Serpico
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
| | - Yuewen Zhu
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Renata Faria Maia
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Sumedha Sumedha
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
2
|
Singh PK, Kim S, Smith AW. HER4 is a high-affinity dimerization partner for all EGFR/HER/ErbB family proteins. Protein Sci 2024; 33:e5171. [PMID: 39276020 PMCID: PMC11401057 DOI: 10.1002/pro.5171] [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: 04/23/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024]
Abstract
Human epidermal growth factor receptors (HER)-also known as EGFR or ErbB receptors-are a subfamily of receptor tyrosine kinases (RTKs) that play crucial roles in cell growth, division, and differentiation. HER4 (ErbB4) is the least studied member of this family, partly because its expression is lower in later stages of development. Recent work has suggested that HER4 can play a role in metastasis by regulating cell migration and invasiveness; however, unlike EGFR and HER2, the precise role that HER4 plays in tumorigenesis is still unresolved. Early work on HER family proteins suggested that there are direct interactions between the four members, but to date, there has been no single study of all four receptors in the same cell line with the same biophysical method. Here, we quantitatively measure the degree of association between HER4 and the other HER family proteins in live cells with a time-resolved fluorescence technique called pulsed interleaved excitation fluorescence cross-correlation spectroscopy (PIE-FCCS). PIE-FCCS is sensitive to the oligomerization state of membrane proteins in live cells, while simultaneously measuring single-cell protein expression levels and diffusion coefficients. Our PIE-FCCS results demonstrate that HER4 interacts directly with all HER family members in the cell plasma membrane. The interaction between HER4 and other HER family members intensified in the presence of a HER4-specific ligand. Our work suggests that HER4 is a preferred dimerization partner for all HER family proteins, even in the absence of ligands.
Collapse
Affiliation(s)
- Pradeep Kumar Singh
- Department of Chemistry and BiochemistryTexas Tech UniversityLubbockTexasUSA
| | - Soyeon Kim
- Division of Cancer Biology, Department of MedicineMetroHealth Medical CenterClevelandOhioUSA
- Department of MedicineCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Adam W. Smith
- Department of Chemistry and BiochemistryTexas Tech UniversityLubbockTexasUSA
| |
Collapse
|
3
|
Becchetti AG, Martini A, Scroccaro G, Joppi R. History of trastuzumab: a case study in health technology reassessment and natural disinvestment in Veneto Region. Front Pharmacol 2024; 15:1406351. [PMID: 39166105 PMCID: PMC11333330 DOI: 10.3389/fphar.2024.1406351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 07/09/2024] [Indexed: 08/22/2024] Open
Affiliation(s)
| | - Anna Martini
- Department Diagnostic and Public Health, University of Verona, Verona, Italy
- Direction of Pharmaceutical–Prosthetics and Medical Devices, Venice, Italy
| | - Giovanna Scroccaro
- Direction of Pharmaceutical–Prosthetics and Medical Devices, Venice, Italy
| | - Roberta Joppi
- Territorial Pharmaceutical Assistance Unit, Azienda ULSS 9 Scaligera, Verona, Italy
- Direction of Pharmaceutical–Prosthetics and Medical Devices, Venice, Italy
| |
Collapse
|
4
|
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
|
5
|
Garg P, Malhotra J, Kulkarni P, Horne D, Salgia R, Singhal SS. Emerging Therapeutic Strategies to Overcome Drug Resistance in Cancer Cells. Cancers (Basel) 2024; 16:2478. [PMID: 39001539 PMCID: PMC11240358 DOI: 10.3390/cancers16132478] [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: 06/11/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
The rise of drug resistance in cancer cells presents a formidable challenge in modern oncology, necessitating the exploration of innovative therapeutic strategies. This review investigates the latest advancements in overcoming drug resistance mechanisms employed by cancer cells, focusing on emerging therapeutic modalities. The intricate molecular insights into drug resistance, including genetic mutations, efflux pumps, altered signaling pathways, and microenvironmental influences, are discussed. Furthermore, the promising avenues offered by targeted therapies, combination treatments, immunotherapies, and precision medicine approaches are highlighted. Specifically, the synergistic effects of combining traditional cytotoxic agents with molecularly targeted inhibitors to circumvent resistance pathways are examined. Additionally, the evolving landscape of immunotherapeutic interventions, including immune checkpoint inhibitors and adoptive cell therapies, is explored in terms of bolstering anti-tumor immune responses and overcoming immune evasion mechanisms. Moreover, the significance of biomarker-driven strategies for predicting and monitoring treatment responses is underscored, thereby optimizing therapeutic outcomes. For insights into the future direction of cancer treatment paradigms, the current review focused on prevailing drug resistance challenges and improving patient outcomes, through an integrative analysis of these emerging therapeutic strategies.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, India
| | - Jyoti Malhotra
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
6
|
Kwon WA, Lee SY, Jeong TY, Kim HH, Lee MK. Antibody-Drug Conjugates in Urothelial Cancer: From Scientific Rationale to Clinical Development. Cancers (Basel) 2024; 16:2420. [PMID: 39001482 PMCID: PMC11240765 DOI: 10.3390/cancers16132420] [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: 05/14/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have been a significant advancement in cancer therapy, particularly for urothelial cancer (UC). These innovative treatments, originally developed for hematological malignancies, use target-specific monoclonal antibodies linked to potent cytotoxic agents. This rational drug design efficiently delivers cancer cell-killing agents to cells expressing specific surface proteins, which are abundant in UC owing to their high antigen expression. UC is an ideal candidate for ADC therapy, as it enhances on-target efficacy while mitigating systemic toxicity. In recent years, considerable progress has been made in understanding the biology and mechanisms of tumor progression in UC. However, despite the introduction of immune checkpoint inhibitors, advanced UC is characterized by rapid progression and poor survival rates. Targeted therapies that have been developed include the anti-nectin 4 ADC enfortumab vedotin and the fibroblast growth factor receptor inhibitor erdafitinib. Enfortumab vedotin has shown efficacy in prospective studies in patients with advanced UC, alone and in combination with pembrolizumab. The anti-Trop-2 ADC sacituzumab govitecan has also demonstrated effectiveness in single-armed studies. This review highlights the mechanism of action of ADCs, their application in mono- and combination therapies, primary mechanisms of resistance, and future perspectives for their clinical use in UC treatment. ADCs have proven to be an increasingly vital component of the therapeutic landscape for urothelial carcinoma, filling a gap in the treatment of this progressive disease.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| | - Seo-Yeon Lee
- Department of Urology, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| | - Tae Yoong Jeong
- Department of Urology, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| | - Hyeon Hoe Kim
- Department of Urology, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| | - Min-Kyung Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| |
Collapse
|
7
|
Parit S, Manchare A, Gholap AD, Mundhe P, Hatvate N, Rojekar S, Patravale V. Antibody-Drug Conjugates: A promising breakthrough in cancer therapy. Int J Pharm 2024; 659:124211. [PMID: 38750981 DOI: 10.1016/j.ijpharm.2024.124211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024]
Abstract
Antibody-drug conjugates (ADCs) provide effective cancer treatment through the selective delivery of cytotoxic payloads to the cancer cells. They offer unparalleled precision and specificity in directing drugs to cancer cells while minimizing off-target effects. Despite several advantages, there is a requirement for innovations in the molecular design of ADC owing to drug resistance, cancer heterogeneity along the adverse effects of treatment. The review critically analyses ADC function mechanisms, unraveling the intricate interplay between antibodies, linkers, and payloads in facilitating targeted drug delivery to cancer cells. The article also highlights notable advancements in antibody engineering, which aid in creating highly selective and potent ADCs. Additionally, the review details significant progress in clinical ADC development with an in-depth examination of pivotal trials and approved formulations. Antibody Drug Conjugates (ADCs) are a ground-breaking approach to targeted drug delivery, especially in cancer treatment. They offer unparalleled precision and specificity in directing drugs to cancer cells while minimizing off-target effects. This review provides a comprehensive examination of the current state of ADC development, covering their design, mechanisms of action, and clinical applications. The article emphasizes the need for greater precision in drug delivery and explains why ADCs are necessary.
Collapse
Affiliation(s)
- Swapnali Parit
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Ajit Manchare
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Prashant Mundhe
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Navnath Hatvate
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Satish Rojekar
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
8
|
Phuna ZX, Kumar PA, Haroun E, Dutta D, Lim SH. Antibody-drug conjugates: Principles and opportunities. Life Sci 2024; 347:122676. [PMID: 38688384 DOI: 10.1016/j.lfs.2024.122676] [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: 02/14/2024] [Revised: 04/15/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Antibody-drug conjugates (ADCs) are immunoconjugates that combine the specificity of monoclonal antibodies with a cytotoxic agent. The most appealing aspects of ADCs include their potential additive or synergistic effects of the innate backbone antibody and cytotoxic effects of the payload on tumors without the severe toxic side effects often associated with traditional chemotherapy. Recent advances in identifying new targets with tumor-specific expression, along with improved bioactive payloads and novel linkers, have significantly expanded the scope and optimism for ADCs in cancer therapeutics. In this paper, we will first provide a brief overview of antibody specificity and the structure of ADCs. Next, we will discuss the mechanisms of action and the development of resistance to ADCs. Finally, we will explore opportunities for enhancing ADC efficacy, overcoming drug resistance, and offer future perspectives on leveraging ADCs to improve the outcome of ADC therapy for cancer treatment.
Collapse
Affiliation(s)
- Zhi Xin Phuna
- Research and Development, Medicovestor, Inc, New York City, NY, United States of America
| | - Prashanth Ashok Kumar
- Division of Hematology and Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States of America
| | - Elio Haroun
- Division of Hematology and Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States of America
| | - Dibyendu Dutta
- Division of Hematology and Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States of America
| | - Seah H Lim
- Research and Development, Medicovestor, Inc, New York City, NY, United States of America; Division of Hematology and Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States of America.
| |
Collapse
|
9
|
Saleh K, Khoury R, Khalife N, Chahine C, Ibrahim R, Tikriti Z, Le Cesne A. Mechanisms of action and resistance to anti-HER2 antibody-drug conjugates in breast cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:22. [PMID: 39050884 PMCID: PMC11267152 DOI: 10.20517/cdr.2024.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 07/27/2024]
Abstract
Human epidermal growth factor 2 (HER2)-positive breast cancer (BC) represents nearly 20% of all breast tumors. Historically, these patients had a high rate of relapse and dismal prognosis. The advent of HER2-targeting monoclonal antibodies such as trastuzumab followed by pertuzumab had improved the prognosis of HER2-positive metastatic BC. More recently, antibody-drug conjugates (ADCs) are now reshaping the treatment paradigm of solid tumors, especially breast cancer. Tratsuzumab emtansine (T-DM1) was one of the first ADC developed in oncology and was approved for the management of HER2-positive metastatic BC. In a head-to-head comparison, trastuzumab deruxtecan (T-DXd) defeated T-DM1 as a second-line treatment. The efficacy of ADCs is counterbalanced by the appearance of acquired resistance to these agents. In this paper, we summarize the mechanisms of action and resistance of T-DM1 and T-DXd, as well as their clinical efficacy. Additionally, we also discuss potential strategies for addressing resistance to ADC.
Collapse
Affiliation(s)
- Khalil Saleh
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Rita Khoury
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Nadine Khalife
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Rebecca Ibrahim
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Zamzam Tikriti
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Axel Le Cesne
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| |
Collapse
|
10
|
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
|
11
|
Rubahamya B, Dong S, Thurber GM. Clinical translation of antibody drug conjugate dosing in solid tumors from preclinical mouse data. SCIENCE ADVANCES 2024; 10:eadk1894. [PMID: 38820153 PMCID: PMC11141632 DOI: 10.1126/sciadv.adk1894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
Antibody drug conjugates (ADCs) have made impressive strides in the clinic in recent years with 11 Food and Drug Administration approvals, including 6 for the treatment of patients with solid tumors. Despite this success, the development of new agents remains challenging with a high failure rate in the clinic. Here, we show that current approved ADCs for the treatment of patients with solid tumors can all show substantial efficacy in some mouse models when administered at a similar weight-based [milligrams per kilogram (mg/kg)] dosing in mice that is tolerated in the clinic. Mechanistically, equivalent mg/kg dosing results in a similar drug concentration in the tumor and a similar tissue penetration into the tumor due to the unique delivery features of ADCs. Combined with computational approaches, which can account for the complex distribution within the tumor microenvironment, these scaling concepts may aid in the evaluation of new agents and help design therapeutics with maximum clinical efficacy.
Collapse
Affiliation(s)
- Baron Rubahamya
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shujun Dong
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Greg M. Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
12
|
Franco-Mateos E, Souza-Egipsy V, García-Estévez L, Pérez-García J, Gion M, Garrigós L, Cortez P, Saavedra C, Gómez P, Ortiz C, Cruz VL, Ramos J, Cortés J, Vega JF. Exploring the Combined Action of Adding Pertuzumab to Branded Trastuzumab versus Trastuzumab Biosimilars for Treating HER2+ Breast Cancer. Int J Mol Sci 2024; 25:3940. [PMID: 38612751 PMCID: PMC11011846 DOI: 10.3390/ijms25073940] [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: 02/22/2024] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
The binding activity of various trastuzumab biosimilars versus the branded trastuzumab towards the glycosylated extracellular domain of the human epidermal growth factor receptor 2 (HER2) target in the presence of pertuzumab was investigated. We employed size exclusion chromatography with tetra-detection methodology to simultaneously determine absolute molecular weight, concentration, molecular size, and intrinsic viscosity. All trastuzumab molecules in solution exhibit analogous behavior in their binary action towards HER2 regardless of the order of addition of trastuzumab/pertuzumab. This analogous behavior of all trastuzumab molecules, including biosimilars, highlights the robustness and consistency of their binding activity towards HER2. Furthermore, the addition of HER2 to a mixture of trastuzumab and pertuzumab leads to increased formation of high-order HER2 complexes, up to concentrations of one order of magnitude higher than in the case of sequential addition. The observed increase suggests a potential synergistic effect between these antibodies, which could enhance their therapeutic efficacy in HER2-positive cancers. These findings underscore the importance of understanding the complex interplay between therapeutic antibodies and their target antigens, providing valuable insights for the development of more effective treatment strategies.
Collapse
Affiliation(s)
- Emma Franco-Mateos
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/Serrano 113 bis, 28006 Madrid, Spain; (E.F.-M.); (V.S.-E.); (V.L.C.); (J.R.)
| | - Virginia Souza-Egipsy
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/Serrano 113 bis, 28006 Madrid, Spain; (E.F.-M.); (V.S.-E.); (V.L.C.); (J.R.)
| | | | - José Pérez-García
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Hospital, 08017 Barcelona, Spain; (J.P.-G.); (L.G.); (P.G.); (C.O.); (J.C.)
- Medica Scientia Innovation Research (MedSIR), 08018 Barcelona, Spain
- Medica Scientia Innovation Research (MedSIR), Ridgewood, NJ 07450, USA
| | - María Gion
- Medical Oncology Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain; (M.G.); (C.S.)
| | - Laia Garrigós
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Hospital, 08017 Barcelona, Spain; (J.P.-G.); (L.G.); (P.G.); (C.O.); (J.C.)
| | | | - Cristina Saavedra
- Medical Oncology Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain; (M.G.); (C.S.)
| | - Patricia Gómez
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Hospital, 08017 Barcelona, Spain; (J.P.-G.); (L.G.); (P.G.); (C.O.); (J.C.)
| | - Carolina Ortiz
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Hospital, 08017 Barcelona, Spain; (J.P.-G.); (L.G.); (P.G.); (C.O.); (J.C.)
| | - Víctor L. Cruz
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/Serrano 113 bis, 28006 Madrid, Spain; (E.F.-M.); (V.S.-E.); (V.L.C.); (J.R.)
| | - Javier Ramos
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/Serrano 113 bis, 28006 Madrid, Spain; (E.F.-M.); (V.S.-E.); (V.L.C.); (J.R.)
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Hospital, 08017 Barcelona, Spain; (J.P.-G.); (L.G.); (P.G.); (C.O.); (J.C.)
- Medica Scientia Innovation Research (MedSIR), 08018 Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Juan F. Vega
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/Serrano 113 bis, 28006 Madrid, Spain; (E.F.-M.); (V.S.-E.); (V.L.C.); (J.R.)
| |
Collapse
|
13
|
Zhang M, Zuo Y, Chen S, Li Y, Xing Y, Yang L, Wang H, Guo R. Antibody-drug conjugates in urothelial carcinoma: scientometric analysis and clinical trials analysis. Front Oncol 2024; 14:1323366. [PMID: 38665947 PMCID: PMC11044263 DOI: 10.3389/fonc.2024.1323366] [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: 10/17/2023] [Accepted: 02/12/2024] [Indexed: 04/28/2024] Open
Abstract
In 2020, bladder cancer, which commonly presents as urothelial carcinoma, became the 10th most common malignancy. For patients with metastatic urothelial carcinoma, the standard first-line treatment remains platinum-based chemotherapy, with immunotherapy serving as an alternative in cases of programmed death ligand 1 expression. However, treatment options become limited upon resistance to platinum and programmed death 1 or programmed death ligand 1 agents. Since the FDA's approval of Enfortumab Vedotin and Sacituzumab Govitecan, the therapeutic landscape has expanded, heralding a shift towards antibody-drug conjugates as potential first-line therapies. Our review employed a robust scientometric approach to assess 475 publications on antibody-drug conjugates in urothelial carcinoma, revealing a surge in related studies since 2018, predominantly led by U.S. institutions. Moreover, 89 clinical trials were examined, with 36 in Phase II and 13 in Phase III, exploring antibody-drug conjugates as both monotherapies and in combination with other agents. Promisingly, novel targets like HER-2 and EpCAM exhibit substantial therapeutic potential. These findings affirm the increasing significance of antibody-drug conjugates in urothelial carcinoma treatment, transitioning them from posterior-line to frontline therapies. Future research is poised to focus on new therapeutic targets, combination therapy optimization, treatment personalization, exploration of double antibody-coupled drugs, and strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Jilin University, Changchun, China
| | - Yuanye Zuo
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Siyi Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yaonan Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Rui Guo
- Department of Clinical Laboratory, First Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Zimmerman BS, Esteva FJ. Next-Generation HER2-Targeted Antibody-Drug Conjugates in Breast Cancer. Cancers (Basel) 2024; 16:800. [PMID: 38398191 PMCID: PMC10887217 DOI: 10.3390/cancers16040800] [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: 01/07/2024] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) tyrosine kinase is overexpressed in 20% of breast cancers and associated with a less favorable prognosis compared to HER2-negative disease. Patients have traditionally been treated with a combination of chemotherapy and HER2-targeted monoclonal antibodies such as trastuzumab and pertuzumab. The HER2-targeted antibody-drug conjugates (ADCs) trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd) represent a novel class of therapeutics in breast cancer. These drugs augment monoclonal antibodies with a cytotoxic payload, which is attached by a linker, forming the basic structure of an ADC. Novel combinations and sequential approaches are under investigation to overcome resistance to T-DM1 and T-DXd. Furthermore, the landscape of HER2-targeted therapy is rapidly advancing with the development of ADCs designed to attack cancer cells with greater precision and reduced toxicity. This review provides an updated summary of the current state of HER2-targeted ADCs as well as a detailed review of investigational agents on the horizon. Clinical trials are crucial in determining the optimal dosing regimens, understanding resistance mechanisms, and identifying patient populations that would derive the most benefit from these treatments. These novel ADCs are at the forefront of a new era in targeted cancer therapy, holding the potential to improve outcomes for patients with HER2-positive and HER2-Low breast cancer.
Collapse
Affiliation(s)
- Brittney S. Zimmerman
- Northwell, New Hyde Park, NY 11042, USA;
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| | - Francisco J. Esteva
- Northwell, New Hyde Park, NY 11042, USA;
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| |
Collapse
|
15
|
El Alaoui M, Sivado E, Jallas AC, Mebarki L, Dyson MR, Perrez F, Valsesia-Wittmann S, El Alaoui S. Antibody and antibody fragments site-specific conjugation using new Q-tag substrate of bacterial transglutaminase. Cell Death Discov 2024; 10:79. [PMID: 38360912 PMCID: PMC10869684 DOI: 10.1038/s41420-024-01845-3] [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: 10/31/2023] [Revised: 01/16/2024] [Accepted: 02/01/2024] [Indexed: 02/17/2024] Open
Abstract
During the last few years Antibody-Drug Conjugates (ADCs) have become one of the most active and very promising therapeutic weapons. Lessons learned from the traditional chemical conjugations (via lysine or cysteine residues of the antibodies) and the clinical studies of the developed ADCs have recently paved the way to the improvement of the conjugation technologies. Use of site-specific conjugation is considered as the promising path for improving the design and development of homogeneous ADCs with controlled Drug-Antibody ratio (DAR). Moreover, some of these conjugations can be applied to antibody fragments such as Fab, scfv and VHH for which random and chemical conjugation showed significant limitations. In this study, we identified a novel small peptide substrate (Q-tag) with high affinity and specificity of bacterial transglutaminase which can be genetically fused to different formats of antibodies of interest for the development of enzymatic site-specific conjugation we named "CovIsolink" platform. We describe the synthesis of chemically defined drugs conjugation in which the site and stoichiometry of conjugation are controlled using a genetically encoded Q-tag peptide with specific amino acids which serves as a substrate of bacterial transglutaminase. This approach has enabled the generation of homogeneous conjugates with DAR 1,7 for full IgG and 0,8 drug ratio for Fab, scfv and VHH antibody fragments without the presence of significant amounts of unconjugated antibody and fragments. As a proof of concept, Q-tagged anti Her-2 (human IgG1 (Trastuzumab) and the corresponding fragments (Fab, scfv and VHH) were engineered and conjugated with different aminated-payloads. The corresponding Cov-ADCs were evaluated in series of in vitro and in vivo assays, demonstrating similar tumor cell killing potency as Trastuzumab emtansine (Kadcyla®) even with lower drug-to-antibody ratio (DAR).
Collapse
Affiliation(s)
| | - Eva Sivado
- Covalab, 1B Rue Jacques Monod, 69500, Bron, France
- Centre Léon Bérard, INSERM 1296 Radiations : défense, Santé et environnement, 28 rue Laennec, 69008, Lyon, France
| | - Anne-Catherine Jallas
- Centre Léon Bérard, INSERM 1296 Radiations : défense, Santé et environnement, 28 rue Laennec, 69008, Lyon, France
| | | | - Michael R Dyson
- IONTAS Ltd, Babraham Research Campus, Babraham, Cambridge, CB22 3AT, UK
| | - Franck Perrez
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Sandrine Valsesia-Wittmann
- Centre Léon Bérard, INSERM 1296 Radiations : défense, Santé et environnement, 28 rue Laennec, 69008, Lyon, France
| | | |
Collapse
|
16
|
Lewis GD, Li G, Guo J, Yu SF, Fields CT, Lee G, Zhang D, Dragovich PS, Pillow T, Wei B, Sadowsky J, Leipold D, Wilson T, Kamath A, Mamounas M, Lee MV, Saad O, Choeurng V, Ungewickell A, Monemi S, Crocker L, Kalinsky K, Modi S, Jung KH, Hamilton E, LoRusso P, Krop I, Schutten MM, Commerford R, Sliwkowski MX, Cho E. The HER2-directed antibody-drug conjugate DHES0815A in advanced and/or metastatic breast cancer: preclinical characterization and phase 1 trial results. Nat Commun 2024; 15:466. [PMID: 38212321 PMCID: PMC10784567 DOI: 10.1038/s41467-023-44533-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 12/14/2023] [Indexed: 01/13/2024] Open
Abstract
Approved antibody-drug conjugates (ADCs) for HER2-positive breast cancer include trastuzumab emtansine and trastuzumab deruxtecan. To develop a differentiated HER2 ADC, we chose an antibody that does not compete with trastuzumab or pertuzumab for binding, conjugated to a reduced potency PBD (pyrrolobenzodiazepine) dimer payload. PBDs are potent cytotoxic agents that alkylate and cross-link DNA. In our study, the PBD dimer is modified to alkylate, but not cross-link DNA. This HER2 ADC, DHES0815A, demonstrates in vivo efficacy in models of HER2-positive and HER2-low cancers and is well-tolerated in cynomolgus monkey safety studies. Mechanisms of action include induction of DNA damage and apoptosis, activity in non-dividing cells, and bystander activity. A dose-escalation study (ClinicalTrials.gov: NCT03451162) in patients with HER2-positive metastatic breast cancer, with the primary objective of evaluating the safety and tolerability of DHES0815A and secondary objectives of characterizing the pharmacokinetics, objective response rate, duration of response, and formation of anti-DHES0815A antibodies, is reported herein. Despite early signs of anti-tumor activity, patients at higher doses develop persistent, non-resolvable dermal, ocular, and pulmonary toxicities, which led to early termination of the phase 1 trial.
Collapse
Affiliation(s)
- Gail D Lewis
- Discovery Oncology, Genentech, South San Francisco, CA, USA.
| | - Guangmin Li
- Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Jun Guo
- Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Shang-Fan Yu
- Translational Oncology, Genentech, South San Francisco, CA, USA
| | | | - Genee Lee
- Translational Oncology, Genentech, South San Francisco, CA, USA
| | | | | | - Thomas Pillow
- Discovery Chemistry, Genentech, South San Francisco, CA, USA
| | - BinQing Wei
- Computational Chemistry, Genentech, South San Francisco, CA, USA
| | - Jack Sadowsky
- Protein Chemistry, Genentech, South San Francisco, CA, USA
- Carmot Therapeutics, Berkeley, CA, USA
| | - Douglas Leipold
- Preclinical and Translational Pharmacokinetics, Genentech, South San Francisco, CA, USA
| | - Tim Wilson
- Oncology Biomarker Development, Genentech, South San Francisco, CA, USA
| | - Amrita Kamath
- Preclinical and Translational Pharmacokinetics, Genentech, South San Francisco, CA, USA
| | - Michael Mamounas
- Project Team Leadership, Oncology, Genentech, South San Francisco, CA, USA
| | - M Violet Lee
- Bioanalytical Sciences, Genentech, South San Francisco, CA, USA
| | - Ola Saad
- Bioanalytical Sciences, Genentech, South San Francisco, CA, USA
| | | | | | - Sharareh Monemi
- Early Clinical Development, Oncology, Genentech, South San Francisco, CA, USA
| | - Lisa Crocker
- Translational Oncology, Genentech, South San Francisco, CA, USA
| | - Kevin Kalinsky
- Winship Cancer Institute at Emory University, Atlanta, GA, USA
| | - Shanu Modi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyung Hae Jung
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Erika Hamilton
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | | | - Ian Krop
- Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Melissa M Schutten
- Safety Assessment Pathology, Genentech, South San Francisco, CA, USA
- SeaGen, South San Francisco, CA, USA
| | - Renee Commerford
- Early Clinical Development, Oncology, Genentech, South San Francisco, CA, USA
- Gilead Sciences, Foster City, CA, USA
| | | | - Eunpi Cho
- Early Clinical Development, Oncology, Genentech, South San Francisco, CA, USA
| |
Collapse
|
17
|
Wei Q, Li P, Yang T, Zhu J, Sun L, Zhang Z, Wang L, Tian X, Chen J, Hu C, Xue J, Ma L, Shimura T, Fang J, Ying J, Guo P, Cheng X. The promise and challenges of combination therapies with antibody-drug conjugates in solid tumors. J Hematol Oncol 2024; 17:1. [PMID: 38178200 PMCID: PMC10768262 DOI: 10.1186/s13045-023-01509-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/06/2023] [Indexed: 01/06/2024] Open
Abstract
Antibody-drug conjugates (ADCs) represent an important class of cancer therapies that have revolutionized the treatment paradigm of solid tumors. To date, many ongoing studies of ADC combinations with a variety of anticancer drugs, encompassing chemotherapy, molecularly targeted agents, and immunotherapy, are being rigorously conducted in both preclinical studies and clinical trial settings. Nevertheless, combination therapy does not always guarantee a synergistic or additive effect and may entail overlapping toxicity risks. Therefore, understanding the current status and underlying mechanisms of ADC combination therapy is urgently required. This comprehensive review analyzes existing evidence concerning the additive or synergistic effect of ADCs with other classes of oncology medicines. Here, we discuss the biological mechanisms of different ADC combination therapy strategies, provide prominent examples, and assess their benefits and challenges. Finally, we discuss future opportunities for ADC combination therapy in clinical practice.
Collapse
Affiliation(s)
- Qing Wei
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
| | - Peijing Li
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Teng Yang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jiayu Zhu
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Sun
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ziwen Zhang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Lu Wang
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xuefei Tian
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, China
- College of Molecular Medicine, Hangzhou Institute for Advanced Study (HIAS), University of Chinese Academy of Sciences, Hangzhou, China
| | - Jiahui Chen
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Can Hu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Junli Xue
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Letao Ma
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Jieer Ying
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China.
| | - Peng Guo
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China.
| | - Xiangdong Cheng
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China.
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, China.
| |
Collapse
|
18
|
Püsküllüoğlu M, Rudzińska A, Pacholczak-Madej R. Antibody-drug conjugates in HER-2 negative breast cancers with poor prognosis. Biochim Biophys Acta Rev Cancer 2023; 1878:188991. [PMID: 37758021 DOI: 10.1016/j.bbcan.2023.188991] [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: 05/02/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
Antibody drug conjugates (ADCs) comprise a rapidly growing class of targeted drugs that selectively deliver a cytotoxic agent to cancer cells, reducing the side effects associated with conventional chemotherapy. Breast cancer (BC) is a heterogeneous entity. The need for effective therapies for HER-2 negative BCs with poor prognosis, such as triple-negative or endocrine-resistant BC, remains unmet due to the lack of potential targets for treatments. These BC subtypes are not candidates for hormonal or anti-HER-2 agents. However, ongoing clinical trials exploring the use of ADCs with a wide range of targets have shown potential for this treatment modality. In this review, we present the current state of knowledge regarding the role of ADC and speculate on novel approaches including ADC combination therapies, new molecular targets, and the role of other subclasses of ADCs (bicycle drug conjugates, bispecific ADCs, immune modulating ADCs) in this clinical scenario.
Collapse
Affiliation(s)
- Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Garncarska Street 11, 31-115 Krakow, Poland.
| | - Agnieszka Rudzińska
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Garncarska Street 11, 31-115 Krakow, Poland
| | - Renata Pacholczak-Madej
- Department of Anatomy, Jagiellonian University, Medical College, Kopernika Street 12, 31-034 Krakow, Poland; Department of Chemotherapy, The District Hospital, 22 Szpitalna Street, 34-200 Sucha Beskidzka, Poland
| |
Collapse
|
19
|
Majumder A. HER3: Toward the Prognostic Significance, Therapeutic Potential, Current Challenges, and Future Therapeutics in Different Types of Cancer. Cells 2023; 12:2517. [PMID: 37947595 PMCID: PMC10648638 DOI: 10.3390/cells12212517] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/14/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023] Open
Abstract
Human epidermal growth factor receptor 3 (HER3) is the only family member of the EGRF/HER family of receptor tyrosine kinases that lacks an active kinase domain (KD), which makes it an obligate binding partner with other receptors for its oncogenic role. When HER3 is activated in a ligand-dependent (NRG1/HRG) or independent manner, it can bind to other receptors (the most potent binding partner is HER2) to regulate many biological functions (growth, survival, nutrient sensing, metabolic regulation, etc.) through the PI3K-AKT-mTOR pathway. HER3 has been found to promote tumorigenesis, tumor growth, and drug resistance in different cancer types, especially breast and non-small cell lung cancer. Given its ubiquitous expression across different solid tumors and role in oncogenesis and drug resistance, there has been a long effort to target HER3. As HER3 cannot be targeted through its KD with small-molecule kinase inhibitors via the conventional method, pharmaceutical companies have used various other approaches, including blocking either the ligand-binding domain or extracellular domain for dimerization with other receptors. The development of treatment options with anti-HER3 monoclonal antibodies, bispecific antibodies, and different combination therapies showed limited clinical efficiency for various reasons. Recent reports showed that the extracellular domain of HER3 is not required for its binding with other receptors, which raises doubt about the efforts and applicability of the development of the HER3-antibodies for treatment. Whereas HER3-directed antibody-drug conjugates showed potentiality for treatment, these drugs are still under clinical trial. The currently understood model for dimerization-induced signaling remains incomplete due to the absence of the crystal structure of HER3 signaling complexes, and many lines of evidence suggest that HER family signaling involves more than the interaction of two members. This review article will significantly expand our knowledge of HER3 signaling and shed light on developing a new generation of drugs that have fewer side effects than the current treatment regimen for these patients.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
20
|
Xia X, Gong C, Zhang Y, Xiong H. The History and Development of HER2 Inhibitors. Pharmaceuticals (Basel) 2023; 16:1450. [PMID: 37895921 PMCID: PMC10610116 DOI: 10.3390/ph16101450] [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: 08/28/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
HER2 is highly expressed in a variety of malignant tumors and affects the prognosis of patients, making it a highly sensitive target for cancer therapy. Since the approval of the first HER2 inhibitor, trastuzumab, in 1998, HER2-targeted drugs have rapidly evolved. Currently, targeting HER2 drugs mainly include monoclonal antibodies (mAbs), tyrosine kinase inhibitors (TKIs), and antibody-drug conjugates (ADCs). This article reviews the development of HER2 inhibitors for various tumors over the past 20 years.
Collapse
Affiliation(s)
- Xiaohui Xia
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
21
|
Benhaghnazar RL, Medina-Kauwe L. Adenovirus-Derived Nano-Capsid Platforms for Targeted Delivery and Penetration of Macromolecules into Resistant and Metastatic Tumors. Cancers (Basel) 2023; 15:3240. [PMID: 37370850 PMCID: PMC10296971 DOI: 10.3390/cancers15123240] [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: 04/25/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Macromolecular therapeutics such as nucleic acids, peptides, and proteins have the potential to overcome treatment barriers for cancer. For example, nucleic acid or peptide biologics may offer an alternative strategy for attacking otherwise undruggable therapeutic targets such as transcription factors and similar oncologic drivers. Delivery of biological therapeutics into tumor cells requires a robust system of cell penetration to access therapeutic targets within the cell interior. A highly effective means of accomplishing this may be borrowed from cell-penetrating pathogens such as viruses. In particular, the cell entry function of the adenovirus penton base capsid protein has been effective at penetrating tumor cells for the intracellular deposition of macromolecular therapies and membrane-impermeable drugs. Here, we provide an overview describing the evolution of tumor-targeted penton-base-derived nano-capsids as a framework for discussing the requirements for overcoming key barriers to macromolecular delivery. The development and pre-clinical testing of these proteins for therapeutic delivery has begun to also uncover the elusive mechanism underlying the membrane-penetrating function of the penton base. An understanding of this mechanism may unlock the potential for macromolecular therapeutics to be effectively delivered into cancer cells and to provide a treatment option for tumors resisting current clinical therapies.
Collapse
Affiliation(s)
| | - Lali Medina-Kauwe
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
22
|
Khoury R, Saleh K, Khalife N, Saleh M, Chahine C, Ibrahim R, Lecesne A. Mechanisms of Resistance to Antibody-Drug Conjugates. Int J Mol Sci 2023; 24:ijms24119674. [PMID: 37298631 DOI: 10.3390/ijms24119674] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
The treatment of cancer patients has dramatically changed over the past decades with the advent of monoclonal antibodies, immune-checkpoint inhibitors, bispecific antibodies, and innovative T-cell therapy. Antibody-drug conjugates (ADCs) have also revolutionized the treatment of cancer. Several ADCs have already been approved in hematology and clinical oncology, such as trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), and sacituzumab govitecan (SG) for the treatment of metastatic breast cancer, and enfortumab vedotin (EV) for the treatment of urothelial carcinoma. The efficacy of ADCs is limited by the emergence of resistance due to different mechanisms, such as antigen-related resistance, failure of internalization, impaired lysosomal function, and other mechanisms. In this review, we summarize the clinical data that contributed to the approval of T-DM1, T-DXd, SG, and EV. We also discuss the different mechanisms of resistance to ADCs, as well as the ways to overcome this resistance, such as bispecific ADCs and the combination of ADCs with immune-checkpoint inhibitors or tyrosine-kinase inhibitors.
Collapse
Affiliation(s)
- Rita Khoury
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Khalil Saleh
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Nadine Khalife
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Mohamad Saleh
- Department of Hematology and Oncology, Lebanese American University Medical Center-Rizk Hopsital, Beirut 1100, Lebanon
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Rebecca Ibrahim
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Axel Lecesne
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| |
Collapse
|
23
|
Pegram M, Jackisch C, Johnston SRD. Estrogen/HER2 receptor crosstalk in breast cancer: combination therapies to improve outcomes for patients with hormone receptor-positive/HER2-positive breast cancer. NPJ Breast Cancer 2023; 9:45. [PMID: 37258523 DOI: 10.1038/s41523-023-00533-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/04/2023] [Indexed: 06/02/2023] Open
Abstract
The human epidermal growth factor receptor 2 (HER2) is overexpressed in 13-22% of breast cancers (BC). Approximately 60-70% of HER2+ BC co-express hormone receptors (HRs). HR/HER2 co-expression modulates response to both anti-HER2-directed and endocrine therapy due to "crosstalk" between the estrogen receptor (ER) and HER2 pathways. Combined HER2/ER blockade may be an effective treatment strategy for patients with HR+/HER2+ BC in the appropriate clinical setting(s). In this review, we provide an overview of crosstalk between the ER and HER2 pathways, summarize data from recently published and ongoing clinical trials, and discuss clinical implications for targeted treatment of HR+/HER2+ BC.
Collapse
Affiliation(s)
- Mark Pegram
- Stanford Cancer Institute, Stanford, CA, USA.
| | - Christian Jackisch
- Obstetrics and Gynaecology and Breast Cancer Center, Klinikum Offenbach GmbH, Offenbach, Germany
| | | |
Collapse
|
24
|
Zhu Y, Zhou M, Kong W, Li C. Antibody-drug conjugates: the clinical development in gastric cancer. Front Oncol 2023; 13:1211947. [PMID: 37305567 PMCID: PMC10250015 DOI: 10.3389/fonc.2023.1211947] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 06/13/2023] Open
Abstract
Gastric cancer (GC) is a prevalent malignant tumor of the digestive system worldwide, ranking among the top five in terms of incidence and mortality. However, the clinical efficacy of conventional treatments for gastric cancer remains limited, with a median overall survival of approximately eight months for advanced cases. In recent years, researchers have increasingly focused on antibody-drug conjugates (ADCs) as a promising approach. ADCs are potent chemical drugs that selectively target cancer cells by binding to specific cell surface receptors with antibodies. Notably, ADCs have demonstrated promising results in clinical studies and have made significant strides in the treatment of gastric cancer. Currently, several ADCs are under investigation in clinical trials for gastric cancer patients, targeting various receptors such as EGFR, HER-2, HER-3, CLDN18.2, Mucin 1, among others. This review offers a comprehensive exploration of ADC drug characteristics and provides an overview of the research progress in ADC-based therapies for gastric cancer.
Collapse
Affiliation(s)
- Yingze Zhu
- School of Clinical Medicine, Affiliated Hospital, North China University of Science and Technology, Tangshan, China
| | - Miao Zhou
- Tangshan Central Hospital, Tangshan, China
| | - Wenyue Kong
- School of Clinical Medicine, Affiliated Hospital, North China University of Science and Technology, Tangshan, China
| | - Congling Li
- School of Clinical Medicine, Affiliated Hospital, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
25
|
Shastry M, Gupta A, Chandarlapaty S, Young M, Powles T, Hamilton E. Rise of Antibody-Drug Conjugates: The Present and Future. Am Soc Clin Oncol Educ Book 2023; 43:e390094. [PMID: 37229614 DOI: 10.1200/edbk_390094] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Antibody-drug conjugates (ADCs) embody a simple, but elegant, vision for cancer therapy-the delivery of a potent cytotoxic agent to tumor cells with minimal damage to normal cells-so-called smart chemo. Although there were significant challenges in achieving this milestone culminating in the first Food and Drug Administration approval in 2000, subsequent advancements in technology have led to rapid drug development with regulatory approvals for ADCs targeting a variety of tumor types. The most successful application for solid tumors has been in breast cancer, with ADCs becoming the standard of care across traditional human epidermal growth factor receptor 2 (HER2)+, hormone receptor+ (HR+) and triple-negative disease subtypes. Moreover, the improved features and gains in potency with the development of ADCs have expanded the treatment-eligible population to those with low/heterogeneous expression of the target antigen on the tumor with trastuzumab deruxtecan or in the case of sacituzumab govitecan, agnostic to target expression. Despite their antibody-directed homing, these novel agents come with their share of toxicities obligating appropriate patient selection and vigilant monitoring while on treatment. As more ADCs are included in the treatment armamentarium, mechanisms of resistance need to be studied and understood for optimal sequencing. Modifying the payload to use immune-stimulating agents or combination therapies with immunotherapy and other effective targeted therapies may further extend the utility of these agents in the treatment of solid tumors.
Collapse
Affiliation(s)
| | - Avantika Gupta
- Department of Medicine, Human Oncology and Pathogenesis Program, MSKCC, New York, NY
| | - Sarat Chandarlapaty
- Department of Medicine, Human Oncology and Pathogenesis Program, MSKCC, New York, NY
| | - Matthew Young
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Thomas Powles
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Erika Hamilton
- Sarah Cannon Research Institute, Nashville, TN
- Tennessee Oncology, Nashville, TN
| |
Collapse
|
26
|
Grinda T, Rassy E, Pistilli B. Antibody-Drug Conjugate Revolution in Breast Cancer: The Road Ahead. Curr Treat Options Oncol 2023; 24:442-465. [PMID: 36966267 PMCID: PMC10122624 DOI: 10.1007/s11864-023-01072-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2023] [Indexed: 03/27/2023]
Abstract
OPINION STATEMENT Antibody drug-conjugates (ADCs) have revolutionized the treatment of many types of cancer, including breast cancer. Recently, two new ADCs have been approved, trastuzumab deruxtecan and sacituzumab govitecan; both have demonstrated impressive improvements in overall survival, trastuzumab deruxtecan in all three subtypes of metastatic breast cancer and sacituzumab govitecan in luminal and triple negative metastatic breast cancer. These drugs are the results of significant progress and innovation in the construction of the three components of an ADC, the monoclonal antibody, the payload, and the linker, and of the discovery of new target antigens. ADC engineering has profoundly changed the paradigm of cancer treatment, on one side being effective on tumors considered inherently resistant to the payload class of drugs and on the other side demonstrating activity in tumors with very low target expression. Yet, it is likely that we are just at the beginning of a new era as the identification of new targets and the introduction of new ADC constructs and combinations will expand the field of ADC rapidly over the coming years.
Collapse
Affiliation(s)
- Thomas Grinda
- Department of Cancer Medicine, Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| | - Barbara Pistilli
- Department of Cancer Medicine, Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France.
| |
Collapse
|
27
|
Fuentes-Antrás J, Genta S, Vijenthira A, Siu LL. Antibody-drug conjugates: in search of partners of choice. Trends Cancer 2023; 9:339-354. [PMID: 36746689 DOI: 10.1016/j.trecan.2023.01.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023]
Abstract
Antibody-drug conjugates (ADCs) have become a credentialled class of anticancer drugs for both solid and hematological malignancies, with regulatory approvals mainly as single agents. Despite extensive preclinical and clinical efforts to develop rational ADC-based combinations, to date only a limited number have demonstrated survival improvements over standard of care. The most appealing partners for ADCs are those that offer additive or synergistic effects on tumor cells or their microenvironment without unacceptable overlapping toxicities. Coadministration with antiangiogenic compounds, HER2-targeting drugs, DNA-damage response agents and immune checkpoint inhibitors (ICIs) represent active forerunners. Through the identification of targets with tumor-specific expression, improved conjugation technologies, and novel linkers and payloads offering superior therapeutic indices, the next generation of ADCs brings optimism to combinatorial approaches.
Collapse
Affiliation(s)
- Jesús Fuentes-Antrás
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Sofia Genta
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Abi Vijenthira
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
28
|
Chen Y, Xu Y, Shao Z, Yu K. Resistance to antibody-drug conjugates in breast cancer: mechanisms and solutions. Cancer Commun (Lond) 2023; 43:297-337. [PMID: 36357174 PMCID: PMC10009672 DOI: 10.1002/cac2.12387] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/13/2022] [Accepted: 10/26/2022] [Indexed: 11/12/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a rapidly developing therapeutic approach in cancer treatment that has shown remarkable activity in breast cancer. Currently, there are two ADCs approved for the treatment of human epidermal growth factor receptor 2-positive breast cancer, one for triple-negative breast cancer, and multiple investigational ADCs in clinical trials. However, drug resistance has been noticed in clinical use, especially in trastuzumab emtansine. Here, the mechanisms of ADC resistance are summarized into four categories: antibody-mediated resistance, impaired drug trafficking, disrupted lysosomal function, and payload-related resistance. To overcome or prevent resistance to ADCs, innovative development strategies and combination therapy options are being investigated. Analyzing predictive biomarkers for optimal therapy selection may also help to prevent drug resistance.
Collapse
Affiliation(s)
- Yu‐Fei Chen
- Department of Breast SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Ying‐ying Xu
- Department of Breast SurgeryFirst Affiliated Hospital of China Medical UniversityShenyangLiaoning110001P. R. China
| | - Zhi‐Ming Shao
- Department of Breast SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Ke‐Da Yu
- Department of Breast SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| |
Collapse
|
29
|
Abelman RO, Wu B, Spring LM, Ellisen LW, Bardia A. Mechanisms of Resistance to Antibody-Drug Conjugates. Cancers (Basel) 2023; 15:cancers15041278. [PMID: 36831621 PMCID: PMC9954407 DOI: 10.3390/cancers15041278] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Antibody-drug conjugates (ADCs), with antibodies targeted against specific antigens linked to cytotoxic payloads, offer the opportunity for a more specific delivery of chemotherapy and other bioactive payloads to minimize side effects. First approved in the setting of HER2+ breast cancer, more recent ADCs have been developed for triple-negative breast cancer (TNBC) and, most recently, hormone receptor-positive (HR+) breast cancer. While antibody-drug conjugates have compared favorably against traditional chemotherapy in some settings, patients eventually progress on these therapies and require a change in treatment. Mechanisms to explain the resistance to ADCs are highly sought after, in hopes of developing next-line treatment options and expanding the therapeutic windows of existing therapies. These resistance mechanisms are categorized as follows: change in antigen expression, change in ADC processing and resistance, and efflux of the ADC payload. This paper reviews the recently published literature on these mechanisms as well as potential options to overcome these barriers.
Collapse
|
30
|
Rassy E, Rached L, Pistilli B. Antibody drug conjugates targeting HER2: Clinical development in metastatic breast cancer. Breast 2022; 66:217-226. [PMID: 36334569 PMCID: PMC9636477 DOI: 10.1016/j.breast.2022.10.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022] Open
Abstract
The identification of the HER2 alteration as an actionable oncogenic driver in breast cancer has propelled the development of HER-targeting monoclonal antibodies (mAb) such as trastuzumab and pertuzumab, which led to dramatic improvements in survival outcomes. Lately, the great strides made toward developing antibody-conjugation methods have led to the development of a new class of compelling compounds, the antibody-drug conjugates (ADCs) targeting HER2 which have profoundly transformed the treatment landscape of breast cancer. HER2-targeting ADCs, trastuzumab-emtansine and trastuzumab-deruxtecan, have improved the overall survival in the second and third-line settings with manageable adverse events. Other HER2-targeting ADCs using novel technological advances in the antibody, linker and/or payload conception have shown promising activity in preclinical and clinical studies and some of them are now being evaluated in larger clinical trials. Multiple challenges still impede the success of ADCs in breast cancer namely the lack of a comprehensive understanding of resistance mechanisms as well as the mechanisms of action of ADCs in special subgroups of patients such as those with low or ultra-low HER2 expression and patients with brain or leptomeningeal metastases (BM). In this framework, we review the approved indications and ongoing trials for HER2-targeting ADCs, across patient subgroups, including those with BM and discuss the associated potential mechanisms of action and resistance. Last, we provide an overview of the future perspectives involving HER2-targeting ADCs in breast cancer.
Collapse
Affiliation(s)
- Elie Rassy
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Layal Rached
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Barbara Pistilli
- Department of Medical Oncology, Gustave Roussy, Villejuif, France.
| |
Collapse
|
31
|
Gandullo-Sánchez L, Ocaña A, Pandiella A. HER3 in cancer: from the bench to the bedside. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:310. [PMID: 36271429 PMCID: PMC9585794 DOI: 10.1186/s13046-022-02515-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022]
Abstract
The HER3 protein, that belongs to the ErbB/HER receptor tyrosine kinase (RTK) family, is expressed in several types of tumors. That fact, together with the role of HER3 in promoting cell proliferation, implicate that targeting HER3 may have therapeutic relevance. Furthermore, expression and activation of HER3 has been linked to resistance to drugs that target other HER receptors such as agents that act on EGFR or HER2. In addition, HER3 has been associated to resistance to some chemotherapeutic drugs. Because of those circumstances, efforts to develop and test agents targeting HER3 have been carried out. Two types of agents targeting HER3 have been developed. The most abundant are antibodies or engineered antibody derivatives that specifically recognize the extracellular region of HER3. In addition, the use of aptamers specifically interacting with HER3, vaccines or HER3-targeting siRNAs have also been developed. Here we discuss the state of the art of the preclinical and clinical development of drugs aimed at targeting HER3 with therapeutic purposes.
Collapse
Affiliation(s)
- Lucía Gandullo-Sánchez
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Alberto Ocaña
- grid.411068.a0000 0001 0671 5785Hospital Clínico San Carlos and CIBERONC, 28040 Madrid, Spain
| | - Atanasio Pandiella
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| |
Collapse
|
32
|
LeNoue-Newton ML, Chen SC, Stricker T, Hyman DM, Blauvelt N, Bedard PL, Meric-Bernstam F, Punglia RS, Schrag D, Lepisto EM, Andre F, Smyth L, Dogan S, Yu C, Wathoo C, Levy M, Eli LD, Xu F, Mann G, Lalani AS, Ye F, Micheel CM, Arnedos M. Natural History and Characteristics of ERBB2-mutated Hormone Receptor-positive Metastatic Breast Cancer: A Multi-institutional Retrospective Case-control Study from AACR Project GENIE. Clin Cancer Res 2022; 28:2118-2130. [PMID: 35190802 DOI: 10.1158/1078-0432.ccr-21-0885] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/21/2021] [Accepted: 02/16/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE We wanted to determine the prognosis and the phenotypic characteristics of hormone receptor-positive advanced breast cancer tumors harboring an ERBB2 mutation in the absence of a HER2 amplification. EXPERIMENTAL DESIGN We retrospectively collected information from the American Association of Cancer Research-Genomics Evidence Neoplasia Information Exchange registry database from patients with hormone receptor-positive, HER2-negative, ERBB2-mutated advanced breast cancer. Phenotypic and co-mutational features, as well as response to treatment and outcome were compared with matched control cases ERBB2 wild type. RESULTS A total of 45 ERBB2-mutant cases were identified for 90 matched controls. The presence of an ERBB2 mutation was not associated with worse outcome determined by overall survival (OS) from first metastatic relapse. No significant differences were observed in phenotypic characteristics apart from higher lobular infiltrating subtype in the ERBB2-mutated group. ERBB2 mutation did not seem to have an impact in response to treatment or time-to-progression (TTP) to endocrine therapy compared with ERBB2 wild type. In the co-mutational analyses, CDH1 mutation was more frequent in the ERBB2-mutated group (FDR < 1). Although not significant, fewer co-occurring ESR1 mutations and more KRAS mutations were identified in the ERBB2-mutated group. CONCLUSIONS ERBB2-activating mutation was not associated with a worse OS from time of first metastatic relapse, or differences in TTP on treatment as compared with a series of matched controls. Although not significant, differences in coexisting mutations (CDH1, ESR1, and KRAS) were noted between the ERBB2-mutated and the control group.
Collapse
Affiliation(s)
| | - Sheau-Chiann Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Thomas Stricker
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Philippe L Bedard
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, Texas
| | - Rinaa S Punglia
- Department of Radiation Oncology, DFCI, Harvard Medical School, Boston, Massachusetts
| | - Deborah Schrag
- Division of Population Sciences and the Department of Medical Oncology, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Eva M Lepisto
- Division of Population Sciences and the Department of Medical Oncology, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Fabrice Andre
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM Unit, U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Lillian Smyth
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Semih Dogan
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM Unit, U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Celeste Yu
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Chetna Wathoo
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, Texas
| | - Mia Levy
- Departments of Biomedical Informatics and Medicine, Division of Hematology/Oncology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lisa D Eli
- PUMA Biotechnology, Los Angeles, California
| | - Feng Xu
- PUMA Biotechnology, Los Angeles, California
| | - Grace Mann
- PUMA Biotechnology, Los Angeles, California
| | | | - Fei Ye
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christine M Micheel
- Department of Medicine, Division of Hematology/Oncology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Monica Arnedos
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM Unit, U981, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
33
|
NRG1/ERBB3/ERBB2 Axis Triggers Anchorage-Independent Growth of Basal-like/Triple-Negative Breast Cancer Cells. Cancers (Basel) 2022; 14:cancers14071603. [PMID: 35406375 PMCID: PMC8997077 DOI: 10.3390/cancers14071603] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/09/2022] [Accepted: 03/16/2022] [Indexed: 01/12/2023] Open
Abstract
Simple Summary Breast cancer is a heterogeneous disease, categorized into distinct subgroups with different clinical prognoses and treatment strategies. This study aimed to evaluate the role of ERBB3 in different molecular subtypes of breast cancers. Despite ERBB3/HER3 and its partner ERBB2/HER2 showing low expression levels in basal-like/triple-negative breast cancers, stratification of basal-like patients according to ERBB3 mRNA expression levels highlighted a correlation between higher ERBB3 levels and shorter relapse-free patients’ survival. In vitro analyses unveiled that the activation of the NRG1/ERBB3/ERBB2 axis robustly induces anchorage-independent growth of basal-like/triple-negative breast cancer cellular models, without significant effects on cell proliferation, differentiation, and migration in adhesion. Overall, our data suggest that ERBB2/ERBB3 plays an oncogenic role in basal-like/triple-negative breast cancer patients, suggesting its neutralization as a therapeutic strategy for these breast cancer subtypes, which today have very limited treatment opportunities. Abstract ERBB3, also known as HER3, is a tyrosine kinase transmembrane receptor of the ERBB family. Upon binding to neuregulin 1 (NRG1), ERBB3 preferentially dimerizes with HER2 (ERBB2), in turn inducing aggressive features in several cancer types. The analysis of a dataset of breast cancer patients unveiled that higher ERBB3 mRNA expression correlates with shorter relapse-free survival in basal-like breast cancers, despite low ERBB3 expression in this breast cancer subtype. Administration of neuregulin 1 beta (NRG1β) significantly affected neither cellular proliferation nor the basal migratory ability of basal-like/triple-negative quasi-normal MCF10A breast cells, cultured in mono-layer conditions. Furthermore, no significant regulation in cell morphology or in the expression of basal/myoepithelial and luminal markers was observed upon stimulation with NRG1β. In non-adherent conditions, NRG1β administration to MCF10A cells did not significantly influence cell survival; however, it robustly induced cell growth as spheroids (3D growth). Intriguingly, a remarkable upregulation of ERBB3 and ERBB2 protein abundance was observed in 3D compared to 2D cell cultures, and NRG1β-induced 3D cell growth was efficiently prevented by the anti-HER2 monoclonal antibody pertuzumab. Similar results were obtained by the analysis of basal-like/triple-negative breast cancer cellular models, MDA-MB-468 and MDA-MB-231 cells, in which NRG1β induced anchorage-independent cell growth that in turn was prevented or reduced by the simultaneous administration of anti-HER2 neutralizing antibodies. Finally, the ability of pertuzumab in suppressing NRG1β-induced 3D growth was also evaluated and confirmed in MCF10A engineered with HER2-overexpression. We suggest that the NRG1/ERBB3/ERBB2 pathway promotes the anchorage-independent growth of basal-like breast cancer cells. Importantly, we provide evidence that ERBB2 neutralization, in particular by pertuzumab, robustly inhibits this process. Our results pave the way towards the development of novel anticancer strategies for basal-like breast cancer patients based on the interception of the NRG1/ERBB3/ERBB2 signaling axis.
Collapse
|
34
|
Boghaert ER, Cox MC, Vaidya KS. Pathophysiological and pharmacological considerations to improve the design and application of antibody-drug conjugates. Cancer Res 2022; 82:1858-1869. [PMID: 35298624 DOI: 10.1158/0008-5472.can-21-3236] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/01/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADC) have emerged as one of the pillars of clinical disease management in oncology. The biggest hurdle to widespread development and application of ADCs has been a narrow therapeutic index. Advances in antibody technologies and formats as well as novel linker and payload chemistries have begun to facilitate structural improvements to ADCs. However, the interplay of structural characteristics with physiologic and pharmacologic factors determining therapeutic success has garnered less attention. This review elaborates on the pharmacology of ADCs, the pathophysiology of cancerous tissues, and the reciprocal consequences on ADC properties and functions. While most currently approved ADCs utilize either microtubule inhibition or DNA damage as primary mechanisms of action, we present arguments to expand this repertoire and highlight the need for payload mechanisms that exploit disease-specific vulnerabilities. We promote the idea that the choice of antibody format, targeting antigen, linker properties, and payload of an ADC should be deliberately fit for purpose by taking the pathophysiology of disease and the specific pharmacology of the drug entity into account, thus allowing a higher probability of clinical success.
Collapse
Affiliation(s)
| | - Megan C Cox
- Abbvie, Inc., North Chicago, IL, United States
| | - Kedar S Vaidya
- Jazz Pharmaceuticals (United States), Palo Alto, CA, United States
| |
Collapse
|
35
|
Bête Noire of Chemotherapy and Targeted Therapy: CAF-Mediated Resistance. Cancers (Basel) 2022; 14:cancers14061519. [PMID: 35326670 PMCID: PMC8946545 DOI: 10.3390/cancers14061519] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Tumor cells struggle to survive following treatment. The struggle ends in either of two ways. The drug combination used for the treatment blocks the proliferation of tumor cells and initiates apoptosis of cells, which is a win for the patient, or tumor cells resist the effect of the drug combination used for the treatment and continue to evade the effect of anti-tumor drugs, which is a bête noire of therapy. Cancer-associated fibroblasts are the most abundant non-transformed element of the microenvironment in solid tumors. Tumor cells play a direct role in establishing the cancer-associated fibroblasts’ population in its microenvironment. Since cancer-associated fibroblasts are activated by tumor cells, cancer-associated fibroblasts show unconditional servitude to tumor cells in their effort to resist treatment. Thus, cancer-associated fibroblasts, as the critical or indispensable component of resistance to the treatment, are one of the most logical targets within tumors that eventually progress despite therapy. We evaluate the participatory role of cancer-associated fibroblasts in the development of drug resistance in solid tumors. In the future, we will establish the specific mode of action of cancer-associated fibroblasts in solid tumors, paving the way for cancer-associated-fibroblast-inclusive personalized therapy. Abstract In tumor cells’ struggle for survival following therapy, they resist treatment. Resistance to therapy is the outcome of well-planned, highly efficient adaptive strategies initiated and utilized by these transformed tumor cells. Cancer cells undergo several reprogramming events towards adapting this opportunistic behavior, leading them to gain specific survival advantages. The strategy involves changes within the transformed tumors cells as well as in their neighboring non-transformed extra-tumoral support system, the tumor microenvironment (TME). Cancer-Associated Fibroblasts (CAFs) are one of the components of the TME that is used by tumor cells to achieve resistance to therapy. CAFs are diverse in origin and are the most abundant non-transformed element of the microenvironment in solid tumors. Cells of an established tumor initially play a direct role in the establishment of the CAF population for its own microenvironment. Like their origin, CAFs are also diverse in their functions in catering to the pro-tumor microenvironment. Once instituted, CAFs interact in unison with both tumor cells and all other components of the TME towards the progression of the disease and the worst outcome. One of the many functions of CAFs in influencing the outcome of the disease is their participation in the development of resistance to treatment. CAFs resist therapy in solid tumors. A tumor–CAF relationship is initiated by tumor cells to exploit host stroma in favor of tumor progression. CAFs in concert with tumor cells and other components of the TME are abettors of resistance to treatment. Thus, this liaison between CAFs and tumor cells is a bête noire of therapy. Here, we portray a comprehensive picture of the modes and functions of CAFs in conjunction with their role in orchestrating the development of resistance to different chemotherapies and targeted therapies in solid tumors. We investigate the various functions of CAFs in various solid tumors in light of their dialogue with tumor cells and the two components of the TME, the immune component, and the vascular component. Acknowledgment of the irrefutable role of CAFs in the development of treatment resistance will impact our future strategies and ability to design improved therapies inclusive of CAFs. Finally, we discuss the future implications of this understanding from a therapeutic standpoint and in light of currently ongoing and completed CAF-based NIH clinical trials.
Collapse
|
36
|
Teicher BA, Morris J. Antibody-Drug Conjugate Targets, Drugs and Linkers. Curr Cancer Drug Targets 2022; 22:463-529. [PMID: 35209819 DOI: 10.2174/1568009622666220224110538] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/22/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
Abstract
Antibody-drug conjugates offer the possibility of directing powerful cytotoxic agents to a malignant tumor while sparing normal tissue. The challenge is to select an antibody target expressed exclusively or at highly elevated levels on the surface of tumor cells and either not all or at low levels on normal cells. The current review explores 78 targets that have been explored as antibody-drug conjugate targets. Some of these targets have been abandoned, 9 or more are the targets of FDA-approved drugs, and most remain active clinical interest. Antibody-drug conjugates require potent cytotoxic drug payloads, several of these small molecules are discussed, as are the linkers between the protein component and small molecule components of the conjugates. Finally, conclusions regarding the elements for the successful antibody-drug conjugate are discussed.
Collapse
Affiliation(s)
- Beverly A Teicher
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| | - Joel Morris
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| |
Collapse
|
37
|
Krop IE, Im SA, Barrios C, Bonnefoi H, Gralow J, Toi M, Ellis PA, Gianni L, Swain SM, Im YH, De Laurentiis M, Nowecki Z, Huang CS, Fehrenbacher L, Ito Y, Shah J, Boulet T, Liu H, Macharia H, Trask P, Song C, Winer EP, Harbeck N. Trastuzumab Emtansine Plus Pertuzumab Versus Taxane Plus Trastuzumab Plus Pertuzumab After Anthracycline for High-Risk Human Epidermal Growth Factor Receptor 2-Positive Early Breast Cancer: The Phase III KAITLIN Study. J Clin Oncol 2022; 40:438-448. [PMID: 34890214 PMCID: PMC8824393 DOI: 10.1200/jco.21.00896] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE We aimed to improve efficacy and reduce toxicity of high-risk human epidermal growth factor receptor 2 (HER2)-positive early breast cancer (EBC) treatment by replacing taxanes and trastuzumab with trastuzumab emtansine (T-DM1). METHODS The phase III KAITLIN study (NCT01966471) included adults with excised HER2-positive EBC (node-positive or node-negative, hormone receptor-negative, and tumor > 2.0 cm). Postsurgery, patients were randomly assigned 1:1 to anthracycline-based chemotherapy (three-four cycles) and then 18 cycles of T-DM1 plus pertuzumab (AC-KP) or taxane (three-four cycles) plus trastuzumab plus pertuzumab (AC-THP). Adjuvant radiotherapy/endocrine therapy was permitted. Coprimary end points were invasive disease-free survival (IDFS) in the intention-to-treat node-positive and overall populations with hierarchical testing. RESULTS The median follow-up was 57.1 months (interquartile range, 52.1-60.1 months) for AC-THP (n = 918) and 57.0 months (interquartile range, 52.1-59.8 months) for AC-KP (n = 928). There was no significant IDFS difference between arms in the node-positive (n = 1,658; stratified hazard ratio [HR], 0.97; 95% CI, 0.71 to 1.32) or overall population (n = 1846; stratified HR, 0.98; 95% CI, 0.72 to 1.32). In the overall population, the three-year IDFS was 94.2% (95% CI, 92.7 to 95.8) for AC-THP and 93.1% (95% CI, 91.4 to 94.7) for AC-KP. Treatment completion rates (ie, 18 cycles) were 88.4% for AC-THP and 65.0% for AC-KP (difference driven by T-DM1 discontinuation because of laboratory abnormalities [12.5%]). Similar rates of grade ≥ 3 (55.4% v 51.8%) and serious adverse events (23.3% v 21.4%) occurred with AC-THP and AC-KP, respectively. KP decreased clinically meaningful deterioration in global health status versus THP (stratified HR, 0.71; 95% CI, 0.62 to 0.80). CONCLUSION The primary end point was not met. Both arms achieved favorable IDFS. Trastuzumab plus pertuzumab plus chemotherapy remains the standard of care for high-risk HER2-positive EBC.
Collapse
Affiliation(s)
- Ian E. Krop
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA,Ian E. Krop, MD, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215; e-mail:
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Carlos Barrios
- Oncology Research Unit, Oncoclínicas, HSL, PUCRS, Porto Alegre, Brazil
| | - Hervé Bonnefoi
- Institut Bergonié Unicancer and Bordeaux University, Bordeaux, France
| | | | - Masakazu Toi
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Paul A. Ellis
- Guy's Hospital and Sarah Cannon Research Institute, London, UK
| | | | - Sandra M. Swain
- Georgetown University Medical Center and MedStar Health, Washington, DC
| | - Young-Hyuck Im
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | - Zbigniew Nowecki
- Maria Sklodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Chiun-Sheng Huang
- National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | | - Yoshinori Ito
- Breast Oncology Center, The Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | | | | | | | | | | | - Eric P. Winer
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Nadia Harbeck
- Breast Center, Dept. OB&GYN, LMU University Hospital, Munich, Germany
| |
Collapse
|
38
|
Díaz-Rodríguez E, Gandullo-Sánchez L, Ocaña A, Pandiella A. Novel ADCs and Strategies to Overcome Resistance to Anti-HER2 ADCs. Cancers (Basel) 2021; 14:154. [PMID: 35008318 PMCID: PMC8750930 DOI: 10.3390/cancers14010154] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/27/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022] Open
Abstract
During recent years, a number of new compounds against HER2 have reached clinics, improving the prognosis and quality of life of HER2-positive breast cancer patients. Nonetheless, resistance to standard-of-care drugs has motivated the development of novel agents, such as new antibody-drug conjugates (ADCs). The latter are a group of drugs that benefit from the potency of cytotoxic agents whose action is specifically guided to the tumor by the target-specific antibody. Two anti-HER2 ADCs have reached the clinic: trastuzumab-emtansine and, more recently, trastuzumab-deruxtecan. In addition, several other HER2-targeted ADCs are in preclinical or clinical development, some of them with promising signs of activity. In the present review, the structure, mechanism of action, and potential resistance to all these ADCs will be described. Specific attention will be given to discussing novel strategies to circumvent resistance to ADCs.
Collapse
Affiliation(s)
- Elena Díaz-Rodríguez
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
- Departamento de Bioquímica y Biología Molecular, University of Salamanca, 37007 Salamanca, Spain
| | - Lucía Gandullo-Sánchez
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
| | - Alberto Ocaña
- Hospital Clínico San Carlos, Centro de Investigación Biomédica en Red de Oncología (CIBERONC), 28040 Madrid, Spain;
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
| |
Collapse
|
39
|
Advances with antibody-drug conjugates in breast cancer treatment. Eur J Pharm Biopharm 2021; 169:241-255. [PMID: 34748933 DOI: 10.1016/j.ejpb.2021.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/21/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022]
Abstract
Antibody-drug conjugate-based therapy for treatment of cancer has attracted much attention because of its enhanced efficacy against numerous cancer types. Commonly, an ADC includes a mAb linked to a therapeutic payload. Antibody, linker and payload are the three main components of ADCs. The high specificity of antibodies is integrated with the strong potency of payloads in ADCs. ADCs with potential cytotoxic small molecules as payloads, generate antibody-mediated cancer therapy. Recently, ADCs with DNA-damaging agents have shown favor over microtubule-targeting agents as payloads. Although ADC resistance can be a barrier to effectiveness, several ADC therapies have been either approved or are in clinical trials for cancer treatment. The ADC-based treatments of breast cancers, particularly TNBC, MDR and metastatic breast cancers, have shown promise in recent years. This review discusses ADC drug designs, and developed for different types of breast cancer including TNBC, MDR and metastatic breast cancer.
Collapse
|
40
|
Benedetti F, Stadlbauer K, Stadlmayr G, Rüker F, Wozniak-Knopp G. A Tetravalent Biparatopic Antibody Causes Strong HER2 Internalization and Inhibits Cellular Proliferation. Life (Basel) 2021; 11:life11111157. [PMID: 34833033 PMCID: PMC8624325 DOI: 10.3390/life11111157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
The overexpression of tyrosine kinase HER2 in numerous cancers, connected with fierce signaling and uncontrolled proliferation, makes it a suitable target for immunotherapy. The acquisition of resistance to currently used compounds and the multiplicity of signaling pathways involved prompted research into the discovery of novel binders as well as treatment options with multiple targeting and multispecific agents. Here we constructed an anti-HER2 tetravalent and biparatopic symmetrical IgG-like molecule by combining the Fab of pertuzumab with a HER2-specific Fcab (Fc fragment with antigen binding), which recognizes an epitope overlapping with trastuzumab. In the strongly HER2-positive cell line SK-BR-3, the molecule induced a rapid and efficient reduction in surface HER2 levels. A potent anti-proliferative effect, specific for the HER2-positive cell line, was observed in vitro, following the induction of apoptosis, and this could not be achieved with treatment with the mixture of pertuzumab and the parental Fcab. The inhibitory cytotoxic effect of our antibody as a single agent makes it a promising contribution to the armory of anti-cancer molecules directed against HER2-addicted cells.
Collapse
|
41
|
Guardia C, Bianchini G, Arpí-LLucià O, Menendez S, Casadevall D, Galbardi B, Dugo M, Servitja S, Montero JC, Soria-Jiménez L, Sabbaghi M, Peña R, Madoz-Gúrpide J, Lloveras B, Lluch A, Eroles P, Arribas J, Pandiella A, Gianni L, Rojo F, Rovira A, Albanell J. Preclinical and Clinical Characterization of Fibroblast-derived Neuregulin-1 on Trastuzumab and Pertuzumab Activity in HER2-positive Breast Cancer. Clin Cancer Res 2021; 27:5096-5108. [PMID: 34385295 DOI: 10.1158/1078-0432.ccr-20-2915] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 05/29/2021] [Accepted: 07/12/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE To characterize expression of neuregulin-1 (NRG1), an HER3 ligand, in HER2-positive breast cancer and its relation with the efficacy of trastuzumab with or without pertuzumab. EXPERIMENTAL DESIGN Characterization of NRG1 expression in tumor cell lines, in tumor specimens, and in cancer-associated fibroblasts (CAFs). Patient-derived CAFs were used to investigate NRG1 impact on the activity of trastuzumab with or without pertuzumab in HER2-positive breast cancer cells. The relationship between NRG1 expression and pathologic response to anti-HER2-based neoadjuvant therapy was assessed in a retrospective patient cohort and in the NeoSphere trial. RESULTS NRG1 was expressed in HER2-positive breast cancer-derived fibroblasts at significantly higher levels than in cancer cells. NRG1 and the conditioned media (CM) from CAFs phosphorylated HER3 and AKT in cancer cells and mediated trastuzumab resistance. Stable genetic depletion of NRG1 from CAFs overcame trastuzumab resistance. Pertuzumab effectively suppressed trastuzumab resistance mediated by either NRG1 or CAF's CM. NRG1 engaged an epithelial-to-mesenchymal transition that was prevented by trastuzumab and pertuzumab. In clinical samples, stromal and/or tumor cell expression of NRG1 determined by immunohistochemistry was uncommon (13.2%) yet significantly linked with residual disease following trastuzumab-based neoadjuvant therapy. In the NeoSphere trial, the magnitude of the difference of pathologic complete response rates favoring the pertuzumab arm was higher in the NRG1-high group. CONCLUSIONS CAF-derived NRG1 mediates trastuzumab resistance through HER3/AKT, which might be reverted by pertuzumab. In patients with HER2-positive breast cancer, high expression of NRG1 was associated to poor response to trastuzumab, but not in combination with pertuzumab.
Collapse
Affiliation(s)
- Cristina Guardia
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | | | - Oriol Arpí-LLucià
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Silvia Menendez
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - David Casadevall
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Barbara Galbardi
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Matteo Dugo
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Sonia Servitja
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | | | - Luis Soria-Jiménez
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - MohammadA Sabbaghi
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Raul Peña
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Juan Madoz-Gúrpide
- Department of Pathology, IIS-Fundación Jiménez Díaz-CIBERONC, Madrid, Spain
| | - Belen Lloveras
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Ana Lluch
- INCLIVA Biomedical Research Institute, Hospital Clínico de Valencia-CIBERONC, Universitat de València, Barcelona, Spain
| | - Pilar Eroles
- INCLIVA Biomedical Research Institute, Hospital Clínico de Valencia-CIBERONC, Universitat de València, Barcelona, Spain.,INCLIVA, Biomedical Research Institute, Valencia, Spain
| | - Joaquin Arribas
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer, IBSAL-CSIC-CIBERONC, Salamanca, Spain
| | | | - Federico Rojo
- Department of Pathology, IIS-Fundación Jiménez Díaz-CIBERONC, Madrid, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. .,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
42
|
Lucas AT, Moody A, Schorzman AN, Zamboni WC. Importance and Considerations of Antibody Engineering in Antibody-Drug Conjugates Development from a Clinical Pharmacologist's Perspective. Antibodies (Basel) 2021; 10:30. [PMID: 34449544 PMCID: PMC8395454 DOI: 10.3390/antib10030030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/04/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Antibody-drug conjugates (ADCs) appear to be in a developmental boom, with five FDA approvals in the last two years and a projected market value of over $4 billion by 2024. Major advancements in the engineering of these novel cytotoxic drug carriers have provided a few early success stories. Although the use of these immunoconjugate agents are still in their infancy, valuable lessons in the engineering of these agents have been learned from both preclinical and clinical failures. It is essential to appreciate how the various mechanisms used to engineer changes in ADCs can alter the complex pharmacology of these agents and allow the ADCs to navigate the modern-day therapeutic challenges within oncology. This review provides a global overview of ADC characteristics which can be engineered to alter the interaction with the immune system, pharmacokinetic and pharmacodynamic profiles, and therapeutic index of ADCs. In addition, this review will highlight some of the engineering approaches being explored in the creation of the next generation of ADCs.
Collapse
Affiliation(s)
- Andrew T. Lucas
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.T.L.); (A.N.S.)
- Carolina Center of Cancer Nanotechnology Excellence, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Amber Moody
- Carolina Center of Cancer Nanotechnology Excellence, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Allison N. Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.T.L.); (A.N.S.)
| | - William C. Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.T.L.); (A.N.S.)
- Carolina Center of Cancer Nanotechnology Excellence, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Glolytics, LLC, Chapel Hill, NC 27517, USA
| |
Collapse
|
43
|
Hatschek T, Foukakis T, Bjöhle J, Lekberg T, Fredholm H, Elinder E, Bosch A, Pekar G, Lindman H, Schiza A, Einbeigi Z, Adra J, Andersson A, Carlsson L, Dreifaldt AC, Isaksson-Friman E, Agartz S, Azavedo E, Grybäck P, Hellström M, Johansson H, Maes C, Zerdes I, Hartman J, Brandberg Y, Bergh J. Neoadjuvant Trastuzumab, Pertuzumab, and Docetaxel vs Trastuzumab Emtansine in Patients With ERBB2-Positive Breast Cancer: A Phase 2 Randomized Clinical Trial. JAMA Oncol 2021; 7:1360-1367. [PMID: 34165503 DOI: 10.1001/jamaoncol.2021.1932] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Importance Trastuzumab emtansine (T-DM1) is presently approved for treatment of advanced breast cancer and after incomplete response to neoadjuvant therapy, but the potential of T-DM1 as monotherapy is so far unknown. Objective To assess pathologic complete response (pCR) to standard neoadjuvant therapy of combination docetaxel, trastuzumab, and pertuzumab (DTP) vs T-DM1 monotherapy in patients with ERBB2 (formerly HER2)-positive breast cancer. Design, Setting, and Participants This randomized phase 2 trial, conducted at 9 sites in Sweden, enrolled 202 patients between December 1, 2014, and October 31, 2018. Participants were 18 years or older, with ERBB2-positive tumors larger than 20 mm and/or verified lymph node metastases. Analysis was performed on an intention-to-treat basis. Interventions Patients were randomized to receive 6 cycles of DTP (standard group) or T-DM1 (investigational group). Crossover was recommended at lack of response or occurrence of intolerable toxic effects. Assessment with fluorine 18-labeled fluorodeoxyglucose (18F-FDG) positron emission tomography combined with computed tomography (PET-CT) was performed at baseline and after 2 and 6 treatment cycles. Main Outcome and Measures Pathologic complete response, defined as ypT0 or Tis ypN0. Secondary end points were clinical and radiologic objective response; event-free survival, invasive disease-free survival, distant disease-free survival, and overall survival; safety; health-related quality of life (HRQoL); functional and biological tumor characteristics; and frequency of breast-conserving surgery. Results Overall, 202 patients were randomized; 197 (99 women in the standard group [median age, 51 years (range, 26-73 years)] and 98 women in the investigational group [median age, 53 years (range, 28-74 years)]) were evaluable for the primary end point. Pathologic complete response was achieved in 45 patients in the standard group (45.5%; 95% CI 35.4%-55.8%) and 43 patients in the investigational group (43.9%; 95% CI 33.9%-54.3%). The difference was not statistically significant (P = .82). In a subgroup analysis, the pCR rate was higher in hormone receptor-negative tumors than in hormone receptor-positive tumors in both treatment groups (45 of 72 [62.5%] vs 45 of 125 [36.0%]). Three patients in the T-DM1 group experienced progression during therapy. In an exploratory analysis, tumor-infiltrating lymphocytes at 10% or more (median) estimated pCR significantly (odds ratio, 2.76; 95% CI, 1.42-5.36; P = .003). Response evaluation with 18F-FDG PET-CT revealed a relative decrease of maximum standardized uptake value by more than 31.3% (median) was associated with pCR (odds ratio, 6.67, 95% CI, 2.38-20.00; P < .001). Conclusions and Relevance In this study, treatment with standard neoadjuvant combination DTP was equal to T-DM1. Trial Registrations ClinicalTrials.gov Identifier: NCT02568839; EudraCT number: 2014-000808-10.
Collapse
Affiliation(s)
- Thomas Hatschek
- Breast Cancer Center, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Theodoros Foukakis
- Breast Cancer Center, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Judith Bjöhle
- Breast Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Tobias Lekberg
- Breast Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Hanna Fredholm
- Breast Cancer Center, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Ana Bosch
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Gyula Pekar
- Department of Pathology, Skåne University Hospital, Lund, Sweden
| | - Henrik Lindman
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Aglaia Schiza
- Department of Immunology, Genetics and Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Zakaria Einbeigi
- Department of Oncology, Southern Älvsborg Hospital, Borås, Sweden
| | - Jamila Adra
- Department of Oncology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Anne Andersson
- Department of Radiation Sciences, Oncology Unit, Umeå University Hospital, Umeå, Sweden
| | - Lena Carlsson
- Department of Oncology, Sundsvall Hospital, Sundsvall, Sweden
| | | | | | - Susanne Agartz
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Edward Azavedo
- Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Per Grybäck
- Department of Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mats Hellström
- Central Trial Office, Clinical Trial Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Hemming Johansson
- Central Trial Office, Clinical Trial Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Claudia Maes
- Central Trial Office, Clinical Trial Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Zerdes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yvonne Brandberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Bergh
- Breast Cancer Center, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Bon G, Pizzuti L, Laquintana V, Loria R, Porru M, Marchiò C, Krasniqi E, Barba M, Maugeri-Saccà M, Gamucci T, Berardi R, Livi L, Ficorella C, Natoli C, Cortesi E, Generali D, La Verde N, Cassano A, Bria E, Moscetti L, Michelotti A, Adamo V, Zamagni C, Tonini G, Barchiesi G, Mazzotta M, Marinelli D, Tomao S, Marchetti P, Valerio MR, Mirabelli R, Russo A, Fabbri MA, D'Ostilio N, Veltri E, Corsi D, Garrone O, Paris I, Sarobba G, Giotta F, Garufi C, Cazzaniga M, Del Medico P, Roselli M, Sanguineti G, Sperduti I, Sapino A, De Maria R, Leonetti C, Di Leo A, Ciliberto G, Falcioni R, Vici P. Loss of HER2 and decreased T-DM1 efficacy in HER2 positive advanced breast cancer treated with dual HER2 blockade: the SePHER Study. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:279. [PMID: 33302999 PMCID: PMC7731769 DOI: 10.1186/s13046-020-01797-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/02/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND HER2-targeting agents have dramatically changed the therapeutic landscape of HER2+ advanced breast cancer (ABC). Within a short time frame, the rapid introduction of new therapeutics has led to the approval of pertuzumab combined with trastuzumab and a taxane in first-line, and trastuzumab emtansine (T-DM1) in second-line. Thereby, evidence of T-DM1 efficacy following trastuzumab/pertuzumab combination is limited, with data from some retrospective reports suggesting lower activity. The purpose of the present study is to investigate T-DM1 efficacy in pertuzumab-pretreated and pertuzumab naïve HER2 positive ABC patients. We also aimed to provide evidence on the exposure to different drugs sequences including pertuzumab and T-DM1 in HER2 positive cell lines. METHODS The biology of HER2 was investigated in vitro through sequential exposure of resistant HER2 + breast cancer cell lines to trastuzumab, pertuzumab, and their combination. In vitro experiments were paralleled by the analysis of data from 555 HER2 + ABC patients treated with T-DM1 and evaluation of T-DM1 efficacy in the 371 patients who received it in second line. Survival estimates were graphically displayed in Kaplan Meier curves, compared by log rank test and, when possibile, confirmed in multivariate models. RESULTS We herein show evidence of lower activity of T-DM1 in two HER2+ breast cancer cell lines resistant to trastuzumab+pertuzumab, as compared to trastuzumab-resistant cells. Lower T-DM1 efficacy was associated with a marked reduction of HER2 expression on the cell membrane and its nuclear translocation. HER2 downregulation at the membrane level was confirmed in biopsies of four trastuzumab/pertuzumab-pretreated patients. Among the 371 patients treated with second-line T-DM1, median overall survival (mOS) from diagnosis of advanced disease and median progression-free survival to second-line treatment (mPFS2) were 52 and 6 months in 177 patients who received trastuzumab/pertuzumab in first-line, and 74 and 10 months in 194 pertuzumab-naïve patients (p = 0.0006 and 0.03 for OS and PFS2, respectively). CONCLUSIONS Our data support the hypothesis that the addition of pertuzumab to trastuzumab reduces the amount of available plasma membrane HER2 receptor, limiting the binding of T-DM1 in cancer cells. This may help interpret the less favorable outcomes of second-line T-DM1 in trastuzumab/pertuzumab pre-treated patients compared to their pertuzumab-naïve counterpart.
Collapse
Affiliation(s)
- Giulia Bon
- Cellular Network and Molecular Therapeutic Target Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Laura Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | | | - Rossella Loria
- Cellular Network and Molecular Therapeutic Target Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Manuela Porru
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Caterina Marchiò
- Department of Medical Sciences, University of Turin, Turin, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Eriseld Krasniqi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Maddalena Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | | | - Rossana Berardi
- Oncology Clinic, "Ospedali Riuniti di Ancona" Hospital, Ancona, Italy
| | - Lorenzo Livi
- Radiotherapy Unit, Department of Oncology, Careggi University Hospital, Florence, Italy
| | | | - Clara Natoli
- Department of Medical, Oral and Biotechnological Sciences, University Gabriele D'Annunzio, Chieti, Italy
| | - Enrico Cortesi
- Department of Medical Oncology, University La Sapienza, Rome, Italy
| | | | - Nicla La Verde
- Oncology Unit, ASST Fatebenefratelli Sacco-PO Fatebenefratelli, Milan, Italy
| | - Alessandra Cassano
- Oncology Unit, IRCCS Foundation Polyclinic University A. Gemelli, University Cattolica Del Sacro Cuore, Rome, Italy
| | - Emilio Bria
- Oncology Unit, IRCCS Foundation Polyclinic University A. Gemelli, University Cattolica Del Sacro Cuore, Rome, Italy.,University of Verona, Verona, Italy
| | - Luca Moscetti
- Department of Oncology and Hematology, University Hospital, Modena, Italy
| | | | - Vincenzo Adamo
- Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Messina, Italy
| | - Claudio Zamagni
- Medical Oncology Unit, Addarii Institute of Oncology, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Giuseppe Tonini
- Department of Oncology, University Campus Biomedico, Rome, Italy
| | - Giacomo Barchiesi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Marco Mazzotta
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Daniele Marinelli
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.,Medical Oncology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Silverio Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, University La Sapienza, Umberto I University Hospital, Rome, Italy
| | - Paolo Marchetti
- Department of Medical Oncology, University La Sapienza, Rome, Italy.,Medical Oncology Unit, Sant'Andrea University Hospital, Rome, Italy
| | | | - Rosanna Mirabelli
- Department of Ematology & Oncology, Pugliese-Ciaccio Hospital, Catanzaro, Italy
| | - Antonio Russo
- Medical Oncology, Paolo Giaccone University Hospital, Palermo, Italy
| | | | | | - Enzo Veltri
- Medical Oncology Unit, Santa Maria Goretti Hospital, Latina, Italy
| | - Domenico Corsi
- Medical Oncology Unit, Fatebenefratelli Hospital, Rome, Italy
| | - Ornella Garrone
- Medical Oncology AO S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Ida Paris
- Gynaecology - Oncology Unit, University Cattolica del Sacro Cuore, Rome, Italy
| | | | - Francesco Giotta
- Department of Medical Oncology, IRCCS Giovanni Paolo II, Bari, Italy
| | - Carlo Garufi
- Division of Medical Oncology, Pescara Hospital, Pescara, Italy
| | - Marina Cazzaniga
- Research Unit Phase I Trials and Oncology Unit, ASST, Monza, Italy
| | - Pietro Del Medico
- Division of Medical Oncology, Reggio Calabria General Hospital, Reggio Calabria, Italy
| | - Mario Roselli
- Department of Systems Medicine, Medical Oncology, University Tor Vergata, Rome, Italy
| | - Giuseppe Sanguineti
- Radiotherapy Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Sperduti
- Biostatistics Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Ruggero De Maria
- Institute of General Pathology, University Cattolica del Sacro Cuore, Rome, Italy.,Department of Medical Oncology, IRCCS Foundation University A. Gemelli, Rome, Italy
| | - Carlo Leonetti
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Angelo Di Leo
- Sandro Pitigliani Medical Oncology Department, Hospital of Prato, Prato, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Rita Falcioni
- Cellular Network and Molecular Therapeutic Target Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Patrizia Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| |
Collapse
|
45
|
Hafeez U, Parakh S, Gan HK, Scott AM. Antibody-Drug Conjugates for Cancer Therapy. Molecules 2020; 25:E4764. [PMID: 33081383 PMCID: PMC7587605 DOI: 10.3390/molecules25204764] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 01/03/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are novel drugs that exploit the specificity of a monoclonal antibody (mAb) to reach target antigens expressed on cancer cells for the delivery of a potent cytotoxic payload. ADCs provide a unique opportunity to deliver drugs to tumor cells while minimizing toxicity to normal tissue, achieving wider therapeutic windows and enhanced pharmacokinetic/pharmacodynamic properties. To date, nine ADCs have been approved by the FDA and more than 80 ADCs are under clinical development worldwide. In this paper, we provide an overview of the biology and chemistry of each component of ADC design. We briefly discuss the clinical experience with approved ADCs and the various pathways involved in ADC resistance. We conclude with perspectives about the future development of the next generations of ADCs, including the role of molecular imaging in drug development.
Collapse
Affiliation(s)
- Umbreen Hafeez
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC 3084, Australia, (U.H.)
- Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Melbourne, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Sagun Parakh
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC 3084, Australia, (U.H.)
- Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Melbourne, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Hui K Gan
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC 3084, Australia, (U.H.)
- Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Melbourne, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC 3084, Australia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC 3084, Australia, (U.H.)
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC 3084, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC 3084, Australia
| |
Collapse
|
46
|
Skeie M, Nikolaysen F, Chitano Y, Stang E. Hsp90 inhibition and co-incubation with pertuzumab induce internalization and degradation of trastuzumab: Implications for use of T-DM1. J Cell Mol Med 2020; 24:10258-10262. [PMID: 32672902 PMCID: PMC7520337 DOI: 10.1111/jcmm.15643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/11/2020] [Accepted: 06/28/2020] [Indexed: 12/15/2022] Open
Abstract
The receptor tyrosine kinase HER2 is associated with a number of human malignancies and is an important therapeutic target. The antibody-drug conjugate trastuzumab emtansine (T-DM1; Kadcyla® ) is recommended as a first-line treatment for patients with HER2-positive metastatic breast cancer. T-DM1 combines the antibody-induced effects of the anti-HER2 antibody trastuzumab (Herceptin® ) with the cytotoxic effect of the tubulin inhibitor mertansine (DM1). For DM1 to have effect, the T-DM1-HER2 complex has to be internalized and the trastuzumab part of T-DM1 has to be degraded. HER2 is, however, considered endocytosis-resistant. As a result of this, trastuzumab is only internalized to a highly limited extent, and if internalized, it is rapidly recycled. The exact reasons for the endocytosis resistance of HER2 are not clear, but it is stabilized by heat-shock protein 90 (Hsp90) and Hsp90 inhibitors induce internalization and degradation of HER2. HER2 can also be internalized upon activation of protein kinase C, and contrary to trastuzumab alone, the combination of two or more anti-HER2 antibodies can induce efficient internalization and degradation of HER2. With intention to find ways to improve the action of T-DM1, we investigated how different ways of inducing HER2 internalization leads to degradation of trastuzumab. The results show that although both Hsp90 inhibition and activation of protein kinase C induce internalization of trastuzumab, only Hsp90 inhibition induces degradation. Furthermore, we find that antibody internalization and degradation are increased when trastuzumab is combined with the clinically approved anti-HER2 antibody pertuzumab (Perjeta® ).
Collapse
Affiliation(s)
- Marianne Skeie
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Filip Nikolaysen
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Ylenia Chitano
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Stang
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
47
|
Fukui F, Hayashi SI, Yamaguchi Y. Heregulin controls ERα and HER2 signaling in mammospheres of ERα-positive breast cancer cells and interferes with the efficacy of molecular targeted therapy. J Steroid Biochem Mol Biol 2020; 201:105698. [PMID: 32404282 DOI: 10.1016/j.jsbmb.2020.105698] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/27/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022]
Abstract
Estrogen receptor (ER)α and the human epidermal growth factor receptor (HER) family are inversely expressed in ERα-positive cancer in association with resistance to hormonal therapy, but the mechanism underlying their relationship remains unknown. We analyzed the effect of HER family ligands on the expression of ER and the HER family in ERα-positive MCF-7 and T47D breast cancer cell lines in 3D spheroid culture. Here, we demonstrated for the first time that heregulin-1β (HRG), a HER3 and HER4 ligand, most effectively regulated ER/HER family expression by decreasing ERα mRNA expression and increasing HER family mRNA expression. HRG treatment attenuated fulvestrant-mediated growth inhibition, and promoted the migration of MCF-7 cells. Moreover, HRG increased the CD44+/CD24- cell fraction and side population cells, both of which are recognized as prospective breast cancer stem cell markers. HRG activated both phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) and mitogen-activated protein kinase (MAPK) pathways. Inhibitors of these pathways reduced the growth of MCF-7 cells, but the addition of HRG has different effects on these pathways. HRG blocked the inhibitory effect of mTOR inhibitors, such as rapamycin and everolimus, on cell growth but not that of a PI3K inhibitor. Furthermore, HRG slightly decreased the inhibitory effect of an AKT inhibitor on cell growth. In contrast, HRG enhanced the MEK inhibitor-induced inhibition of cell growth. These findings suggest that HRG-stimulated signaling pathways allow ERα-positive breast cancer cells to escape from growth inhibition caused by everolimus, via MAPK signaling and/or other signaling pathways. Everolimus improves progression-free survival in combination with exemestane as second-line therapy for metastatic hormone receptor-positive breast cancer. Our study suggests that HRG is a novel target for ERα-positive breast cancer therapy.
Collapse
Affiliation(s)
- Fumiyo Fukui
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan; Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Sendai, Japan.
| | - Shin-Ichi Hayashi
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Sendai, Japan.
| | - Yuri Yamaguchi
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan.
| |
Collapse
|
48
|
D’Amico S, Krasnowska EK, Manni I, Toietta G, Baldari S, Piaggio G, Ranalli M, Gambacurta A, Vernieri C, Di Giacinto F, Bernassola F, de Braud F, Lucibello M. DHA Affects Microtubule Dynamics Through Reduction of Phospho-TCTP Levels and Enhances the Antiproliferative Effect of T-DM1 in Trastuzumab-Resistant HER2-Positive Breast Cancer Cell Lines. Cells 2020; 9:E1260. [PMID: 32438775 PMCID: PMC7290969 DOI: 10.3390/cells9051260] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/13/2020] [Accepted: 05/16/2020] [Indexed: 12/11/2022] Open
Abstract
Trastuzumab emtansine (T-DM1) is an anti-human epidermal growth factor receptor 2 (HER2) antibody-drug conjugated to the microtubule-targeting agent emtansine (DM1). T-DM1 is an effective agent in the treatment of patients with HER2-positive breast cancer whose disease has progressed on the first-line trastuzumab containing chemotherapy. However, both primary and acquired tumour resistance limit its efficacy. Increased levels of the phosphorylated form of Translationally Controlled Tumour Protein (phospho-TCTP) have been shown to be associated with a poor clinical response to trastuzumab therapy in HER2-positive breast cancer. Here we show that phospho-TCTP is essential for correct mitosis in human mammary epithelial cells. Reduction of phospho-TCTP levels by dihydroartemisinin (DHA) causes mitotic aberration and increases microtubule density in the trastuzumab-resistant breast cancer cells HCC1954 and HCC1569. Combinatorial studies show that T-DM1 when combined with DHA is more effective in killing breast cells compared to the effect induced by any single agent. In an orthotopic breast cancer xenograft model (HCC1954), the growth of the tumour cells resumes after having achieved a complete response to T-DM1 treatment. Conversely, DHA and T-DM1 treatment induces a severe and irreversible cytotoxic effect, even after treatment interruption, thus, improving the long-term efficacy of T-DM1. These results suggest that DHA increases the effect of T-DM1 as poison for microtubules and supports the clinical development of the combination of DHA and T-DM1 for the treatment of aggressive HER2-overexpressing breast cancer.
Collapse
Affiliation(s)
- Silvia D’Amico
- National Research Council of Italy, Institute of Translational Pharmacology (IFT-CNR), 00133 Rome, Italy; (S.D.); (E.K.K.)
| | - Ewa Krystyna Krasnowska
- National Research Council of Italy, Institute of Translational Pharmacology (IFT-CNR), 00133 Rome, Italy; (S.D.); (E.K.K.)
| | - Isabella Manni
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (I.M.); (G.P.)
| | - Gabriele Toietta
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (G.T.); (S.B.)
| | - Silvia Baldari
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (G.T.); (S.B.)
| | - Giulia Piaggio
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (I.M.); (G.P.)
| | - Marco Ranalli
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy; (M.R.); (A.G.); (F.B.)
| | - Alessandra Gambacurta
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy; (M.R.); (A.G.); (F.B.)
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (C.V.); (F.d.B.)
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Flavio Di Giacinto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Francesca Bernassola
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy; (M.R.); (A.G.); (F.B.)
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (C.V.); (F.d.B.)
- Oncology and Hemato-Oncology Department, University of Milan, 20122 Milan, Italy
| | - Maria Lucibello
- National Research Council of Italy, Institute of Translational Pharmacology (IFT-CNR), 00133 Rome, Italy; (S.D.); (E.K.K.)
| |
Collapse
|
49
|
Gandullo-Sánchez L, Capone E, Ocaña A, Iacobelli S, Sala G, Pandiella A. HER3 targeting with an antibody-drug conjugate bypasses resistance to anti-HER2 therapies. EMBO Mol Med 2020; 12:e11498. [PMID: 32329582 PMCID: PMC7207167 DOI: 10.15252/emmm.201911498] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/24/2020] [Accepted: 03/29/2020] [Indexed: 02/06/2023] Open
Abstract
Despite impressive clinical benefit obtained with anti‐HER2‐targeted therapies, in advances stages, especially in the metastatic setting, HER2‐positive tumors remain incurable. Therefore, it is important to develop novel strategies to fight these tumors, especially when they become resistant to available therapies. We show here that the anti‐HER3 antibody–drug conjugate EV20/MMAF exerted potent anti‐tumoral properties against several models of primary resistance and secondary resistance to common anti‐HER2 available therapies, including trastuzumab, lapatinib, neratinib, and trastuzumab‐emtansine. HER3 was expressed in these HER2+ breast cancer cells and knockdown experiments demonstrated that HER3 expression was required for the action of EV20/MMAF. In mice injected with trastuzumab‐resistant HER2+ cells, a single dose of EV20/MMAF caused complete and long‐lasting tumor regression. Mechanistically, EV20/MMAF bound to cell surface HER3 and became internalized to the lysosomes. Treatment with EV20/MMAF caused cell cycle arrest in mitosis and promoted cell death through mitotic catastrophe. These findings encourage the clinical testing of EV20/MMAF for several indications in the HER2+ cancer clinic, including situations in which HER2+ tumors become refractory to approved anti‐HER2 therapies.
Collapse
Affiliation(s)
- Lucía Gandullo-Sánchez
- Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Salamanca, Spain
| | - Emily Capone
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studiesand Technology (CAST), University of Chieti-Pescara, Chieti, Italy
| | | | | | - Gianluca Sala
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studiesand Technology (CAST), University of Chieti-Pescara, Chieti, Italy.,MediaPharma s.r.l, Chieti, Italy
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Salamanca, Spain
| |
Collapse
|
50
|
Nadal-Serrano M, Morancho B, Escrivá-de-Romaní S, Bernadó Morales C, Luque A, Escorihuela M, Espinosa Bravo M, Peg V, Dijcks FA, Dokter WH, Cortés J, Saura C, Arribas J. The Second Generation Antibody-Drug Conjugate SYD985 Overcomes Resistances to T-DM1. Cancers (Basel) 2020; 12:cancers12030670. [PMID: 32183023 PMCID: PMC7139846 DOI: 10.3390/cancers12030670] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/29/2022] Open
Abstract
Trastuzumab-emtansine (T-DM1) is an antibody-drug conjugate (ADC) approved for the treatment of HER2 (human epidermal growth factor receptor 2)-positive breast cancer. T-DM1 consists of trastuzumab covalently linked to the cytotoxic maytansinoid DM1 via a non-cleavable linker. Despite its efficacy, primary or acquired resistance frequently develops, particularly in advanced stages of the disease. Second generation ADCs targeting HER2 are meant to supersede T-DM1 by using a cleavable linker and a more potent payload with a different mechanism of action. To determine the effect of one of these novel ADCs, SYD985, on tumors resistant to T-DM1, we developed several patient-derived models of resistance to T-DM1. Characterization of these models showed that previously described mechanisms-HER2 downmodulation, impairment of lysosomal function and upregulation of drug efflux pumps-account for the resistances observed, arguing that mechanisms of resistance to T-DM1 are limited, and most of them have already been described. Importantly, SYD985 was effective in these models, showing that the resistance to first generation ADCs can be overcome with an improved design.
Collapse
Affiliation(s)
- Mercedes Nadal-Serrano
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (M.N.-S.); (B.M.); (C.B.M.); (A.L.); (M.E.); (J.C.)
| | - Beatriz Morancho
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (M.N.-S.); (B.M.); (C.B.M.); (A.L.); (M.E.); (J.C.)
| | - Santiago Escrivá-de-Romaní
- Breast Cancer and Melanoma Group, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (S.E.-d.-R.); (M.E.B.); (V.P.); (C.S.)
- Medical Oncology Department, Vall d´Hebron University Hospital (HUVH), 08035 Barcelona, Spain
| | - Cristina Bernadó Morales
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (M.N.-S.); (B.M.); (C.B.M.); (A.L.); (M.E.); (J.C.)
| | - Antonio Luque
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (M.N.-S.); (B.M.); (C.B.M.); (A.L.); (M.E.); (J.C.)
| | - Marta Escorihuela
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (M.N.-S.); (B.M.); (C.B.M.); (A.L.); (M.E.); (J.C.)
| | - Martín Espinosa Bravo
- Breast Cancer and Melanoma Group, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (S.E.-d.-R.); (M.E.B.); (V.P.); (C.S.)
- Medical Oncology Department, Vall d´Hebron University Hospital (HUVH), 08035 Barcelona, Spain
| | - Vicente Peg
- Breast Cancer and Melanoma Group, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (S.E.-d.-R.); (M.E.B.); (V.P.); (C.S.)
- Medical Oncology Department, Vall d´Hebron University Hospital (HUVH), 08035 Barcelona, Spain
| | - Fred A. Dijcks
- Preclinical R&D, Synthon Biopharmaceuticals BV, 6545 CM Nijmegen, The Netherlands; (F.A.D.)
| | - Wim H.A. Dokter
- Preclinical R&D, Synthon Biopharmaceuticals BV, 6545 CM Nijmegen, The Netherlands; (F.A.D.)
| | - Javier Cortés
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (M.N.-S.); (B.M.); (C.B.M.); (A.L.); (M.E.); (J.C.)
| | - Cristina Saura
- Breast Cancer and Melanoma Group, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (S.E.-d.-R.); (M.E.B.); (V.P.); (C.S.)
- Medical Oncology Department, Vall d´Hebron University Hospital (HUVH), 08035 Barcelona, Spain
| | - Joaquín Arribas
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain; (M.N.-S.); (B.M.); (C.B.M.); (A.L.); (M.E.); (J.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08035 Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autónoma de Barcelona, Campus de la UAB, 08193 Bellaterra, Spain
- Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
- Correspondence:
| |
Collapse
|